Skip to main content
Therapeutic Advances in Medical Oncology logoLink to Therapeutic Advances in Medical Oncology
. 2024 Jan 9;16:17588359231221337. doi: 10.1177/17588359231221337

Development of PARP inhibitors in advanced prostate cancer

Maria Teresa Bourlon 1, Paola Valdez 2, Elena Castro 3,
PMCID: PMC10777773  PMID: 38205078

Abstract

The relatively high prevalence of alterations in the homologous recombination repair (HRR) pathway described in advanced prostate cancer provides a unique opportunity to develop therapeutic strategies that take advantage of the decreased tumor ability to repair DNA damage. Poly ADP-ribose polymerase (PARP) inhibitors have been demonstrated to improve the outcomes of metastatic castration-resistant prostate cancer (mCRPC) patients with HRR defects, particularly in those with BRCA1/2 alterations. To expand the benefit of PARPi to patients without detectable HRR alterations, multiple studies are addressing potential synergies between PARP inhibition (PARPi) and androgen receptor pathway inhibitors (ARSi), radiation, radioligand therapy, chemotherapy, or immunotherapy, and these strategies are also being evaluated in the hormone-sensitive setting. In this review, we summarize the development of PARPi in prostate cancer, the potential synergies, and combinations being investigated as well as the future directions of PARPi for the management of the disease.

Keywords: BRCA, HRR, niraprib, olaparib, PARP inhibition, prostate cancer, rucaparib, talazoparib

Plain language summary

Development of PARP inhibitors in advanced prostate cancer

Alterations in the mechanisms responsible for repairing damaged DNA are frequently altered in advanced prostate cancer. This provides a unique opportunity to develop therapies that exploit the decreased ability of these prostate tumours to repair DNA. Poly ADP-ribose polymerase (PARP) inhibitors have been successfully used to treat other tumor types with similar deficiencies and recently, multiple studies have demonstrated its efficacy also in prostate cancer, particularly in tumors with BRCA1/2 alterations. To expand the benefit of PARPi to patients without detectable DNA repair alterations, multiple studies are addressing potential synergies between PARP inhibition (PARPi) and androgen receptor pathway inhibitors (ARSi), radiation, radiopharmaceuticals, chemotherapy and immunotherapy in different disease stages. In this review, we summarize the development of PARPi in prostate cancer, the potential synergies and combinations being evaluated as well as the future directions of PARPi for the management of the disease.

Introduction

Prostate cancer is the second most frequent cancer and the fifth leading cause of cancer-related deaths in men globally. 1 Androgen deprivation therapy (ADT) remains the cornerstone of management for advanced prostate cancer; nonetheless, the addition of a variety of therapies to ADT including taxane-based chemotherapy (docetaxel and cabazitaxel), androgen receptor signaling inhibitors (ARSi, abiraterone, enzalutamide, darolutamide, and apalutamide), and radiopharmaceuticals (radium223 and lutetium177) have demonstrated to prolong the overall survival (OS) of these men. 2 However, response to these agents is heterogeneous and even those patients who achieve significant responses eventually become resistant. In consequence, the disease remains incurable, and new therapeutic strategies are needed.

In the last decade, multiple studies have described the molecular landscape of advanced prostate cancer and the mechanisms behind treatment resistance.35 A better understanding of the molecular heterogeneity of the disease allows the search for new treatment options. Alterations in genes involved in the homologous recombination repair (HRR) pathway have been reported in a quarter of patients with advanced prostate cancer3,6 and provide a unique opportunity to develop therapeutic strategies that take advantage of the decreased tumor ability to repair DNA damage. Poly ADP-ribose polymerase (PARP) inhibitors have been investigated in patients with metastatic castration-resistant prostate cancer (mCRPC) with HRR defects demonstrating great efficacy for the treatment of these tumors, particularly for those with BRCA1/2 alterations. Nonetheless, the role of PARP inhibitors (PARPi) is evolving as preclinical models suggest potential synergies with other agents, and these combinations are being evaluated in multiple clinical trials in mCRPC but also in the hormone-sensitive setting.

Prevalence and clinical implications of HRR alterations in prostate cancer

Alterations in DNA Damage Repair DDR genes have been observed in 10% of localized prostate tumors and almost a third of metastatic cases.3,4 Among these genes, the HRR pathway is the most commonly impaired DNA repair pathway in prostate cancer. In 28% of the samples analyzed in the PROFound study, at least one HRR alteration was found. The most frequently altered gene was BRCA2 (8.7%), followed by CDK12 (6.3%), ATM (5.9%), CHEK2 (1.2%), and BRCA1 (1%). Co-occurring aberrations in two or more HRR genes were identified in 2.2% of cases. 6 Importantly, the analysis of paired samples from primary tumors and metastases at the time of castration resistance in various series has revealed no difference in the prevalence of DDR (and HRR) aberrations, suggesting that these events occur early in the progression of lethal prostate cancer.5,7,8 Importantly, a significant proportion of HRR alterations found in tumors originates from the germline, including 50% of BRCA2 mutations.9,10

Germline BRCA2 mutations are a well-established poor prognosis factor for localized1113 and advanced14,15 prostate cancer. The role of concurrent genomic events is unclear but it seems that certain events (i.e. RB1-BRCA2 co-deletion, MYC amplification) could modulate the effect of inherited BRCA2 mutations on patient outcomes. 16 A recent study suggests that somatic BRCA2 alterations would have a negative prognostic impact on mCRPC patients conventionally treated with ARSi and taxanes. 17 The clinical implications of other HRR genes are not well established except for biallelic CDK12 inactivation,18,19 consistently reported to also have a detrimental effect. Inactivating CDK12 mutations leads to gene fusion-induced neoantigens and a potential sensitivity to immunotherapy that is being addressed in several studies. 19

Mechanism of action of Poly (ADP-Ribose) polymerase inhibitors

Poly (ADP-ribose) polymerases (PARP) is a family of ADP-ribosyl transferase enzymes, of which PARP1 is the most abundant, that transfer negatively charged poly ADP-ribose (PAR) groups from donor NAD+ molecules onto their target proteins. Through these PAR chains, PARPs control a wide array of cellular processes, such as DNA repair, transcriptional regulation, and RNA interference among others. 20 PARP1 is a key component of the base excision repair mechanism that acts as a DNA damage sensor and a signal transducer. It detects single-strand breaks, binds to the DNA adjacent to the damage, and then synthesizes PAR chains on target proteins (PARylation) leading to the recruitment of additional factors that complete the DNA repair process. PARP1 autoPARylation leads to its own release for the site of DNA damage.21,22

Based on this critical role in initiating response to DNA damage, PARP has been an attractive pharmacological target for cancer therapeutics. PARP inhibitors (PARPi) mimic the structure of nicotinamide and have two general effects: catalytic inhibition of PARP1 (preventing PARylation) and ‘trapping’ PARP1 on damaged DNA. The precise mechanism explaining trapping is unclear but two possibilities have been proposed: autoPARylation inhibition prevents the release of PARP1 from DNA or PARPi binding to the catalytic site modifies PARP1 structure enhancing DNA avidity.22,23 Then, trapped PARP1 stalls the progress of replication forks, that in normal cells would be repaired by HRR proteins but in HRR-deficient cells (i.e. due to BRCA2 mutations) alternative error-prone DNA repair mechanisms, such as nonhomologous end-joining repair (NHEJ), are used, leading to catastrophic genomic instability and cell death. 24 Initial reports described that BRCA1-/- or BRCA2-/- cell lines displayed a 60- to 1000-fold greater sensitivity to olaparib and talazoparib precursors than BRCA+/+ cell lines. These observations have been replicated numerous times with other PARPi. 25

Several PARPi have been developed that differ in their ability to inhibit its catalytic activity and trap PARP in damaged DNA complexes23,25 and subsequently show different antitumor activity. 24 At the same time, increased PARP trapping has been associated with high myelosupression. 26 Most PARPi clinically developed have limited selectivity for PARP1 over PARP2, although preclinical data suggest that synthetic lethality with BRCA½ mutations was caused solely by PARP1 inhibition. 23 PARP2 seems to play a major role in the survival of hematopoietic/stem progenitor cells and its inhibition could result in the hematological toxicity observed in the clinic. 27 Other common class-specific side effects include fatigue and gastrointestinal toxicity. The type, grade, or frequency of side effects do not seem to be associated with the presence of BRCA½ or other HRR alterations.

After a variable period, resistance to PARPi eventually occurs. Resistance mechanisms to these drugs are complex and include events that restore HRR function such as secondary mutations restoring the open reading frame of HRR genes; replication folk protection; point mutations in the DNA-binding domain of PARP1; and alterations in drug efflux pumps affecting drug pharmacokinetics, among others.24,28

Development of PARP inhibitors in prostate cancer

PARPi were initially developed for cancer treatment as radio and chemo-sensitizing drugs but early preclinical observations supported their development in monotherapy for BRCA-deficient tumors. 24 Germline BRCA1/2 mutation carriers with different tumor types were the initial target population to test the PARPi-BRCA synthetic lethal hypothesis in the clinic. A first-in-human clinical trial with KU-0059436 (a precursor of Olaparib) was conducted to determine safety, the adverse-event profile, establish a recommended dose, and the pharmacokinetic and pharmacodynamic profiles. Once these were established, the expansion cohorts included only cancer patients with germline BRCA mutations to test the hypothesis that patients with BRCA-related tumors would show an objective antitumor response to single-agent olaparib. Twelve of the 19 (63%) carriers with evaluable BRCA-related tumors achieved objective response or disease stabilization for ⩾4 months. One germline BRCA2 mutation carrier with prostate cancer was included who benefited from the drug for over 2 years. 29

The first-in-man clinical trial of niraparib (MK4827) included an exploratory expansion cohort of 18 mCRPC patients, in addition to 3 germline BRCA mutation carriers with CRPC recruited during the dose-escalation phase. Nine of those 21 mCPRC patients (43%) had stable disease for a median duration of 8.5 months; no radiological responses per RECIST were documented although one patient experienced a >50% decrease in prostate-specific antigen (PSA) on treatment, remaining on treatment for 10 months.

TOPARP-A 30 was a phase II trial to identify possible biomarkers that could help identify mCRPC patients who would benefit from treatment with olaparib. The study enrolled 50 molecularly unselected mCRPC heavily pretreated. The main objective was the composite response rate, defined either as objective response rate (ORR) by response evaluation criteria in solid tumors 1.1 (RECIST 1.1), a decline in PSA level of at least 50% (PSA50), and/or conversion in circulating tumor cell (CTC) count from ⩾5 cells/7.5 mL to <5 cells/7.5 mL of blood. Overall, 16/49 (32%) evaluable patients fulfilled the prespecified definition of response. These included 11/49 (22%) patients achieving at least 50% PSA fall and 6/32 (19%) patients with measurable disease in CT scan achieving a RECIST response. All patients donated fresh and/or archival tumor samples for molecular analysis, and next-generation targeted sequencing was pursued to investigate putative biomarkers of response. A patient was classified as ‘biomarker-positive’ if a homozygous deletion or deleterious mutation was identified in any of the DNA repair genes analyzed. Almost all patients who responded to therapy (14/16) were biomarker positive, including seven patients harboring somatic or germline BRCA2 alterations that responded to olaparib. Other responding patients had alterations in ATM, PALB2, CHEK2, and other genes. Conversely, these genomic aberrations were uncommon in non-responding patients. Disease control lasted at least 6 months in many of these heavily pretreated cases, with four patients receiving the drug for over a year. These data led to a second stage of the trial (TOPARP-B 31 ) in which patients were pre-selected based on the detection of tumor genomic aberrations associated with sensitivity to PARPi.

Further phase II trials have investigated the efficacy and safety of other PARPi in patients with HRR-altered mCRPC patients and disease progression to several treatment lines, with the primary endpoints being ORR and/or PSA50. TOPARP-B, 31 TRITON2,3234 TALAPRO-1, 35 and GALAHAD 36 have, respectively, assessed the efficacy of olaparib, rucaparib, talazoparib, and niraparib in patients with mCRPC who had progressed on prior ARSIs and taxanes. In all of them, a marked benefit was seen in the BRCA 1/2 population, with ORR ranging from 34% to 52% and PSA response from 43% to 76%. The antitumor activity associated with alterations in non-BRCA HRR genes is heterogeneous; while the clinical benefit of PARPi in patients with ATM, CDK12, or CHEK2 alterations seems to be limited at best,31,32,35,36 relevant response rates have been reported in patients with PALB2 alterations31,33,35 (Table 1).

Table 1.

Phase II studies have investigated the efficacy of PARP inhibitors in monotherapy for the treatment of mCRPC.

Study characteristics TOPARP-B TRITON2 TALAPRO-1 GALAHAD
Drug Olaparib Rucaparib Talazoparib Niraparib
Study design and population Phase II, single arm
mCRPC after taxanes
(ARSi allowed)
N = 98
Phase II, single arm
mCRPC after ARSi and taxane
N = 193
Phase II, single arm
mCRPC ARSi and taxane
N = 127
Phase II, single arm
mCRPC ARSi and taxane
N = 223
Primary objective Composite endpoint:
ORR, PSA50, CTC conversion
ORR in pts with DDR alterations ORR in pts with DDR alterations ORR in biallelic BRCA1/2
Results Composite response (1ry endpoint)
-By gene:
 85% BRCA1/2
 37% ATM
 25% CDK12
 57% PALB2
 20% other
-By dose cohort:
 54% with 400 mg bid
 39% with 300 mg bid
ORR (1ry endpoint)
 43.5% BRCA1/2
 10.5% ATM
 11.1% CHEK2
 0% CDK12
 28.5% other DDR
PSA50 (2ry endpoint)
 54.8% BRCA1/2
 4.1% ATM
 16.7% CHEK2
 6.7% CDK12
 36% other DDR
ORR (1ry endpoint)
 46% BRCA 1/2
 25% PALB2
 12% ATM
 0% other DDR
PSA50 (2ry endpoint)
 66% BRCA 1/2
 75% PALB2
 7% ATM
 6% other DDR
ORR (1ry and 2ry endpoint):
 26% BRCA1/2
 5% in non-BRCA DDR
PSA50 (exploratory endpoint)
 43% BRCA1/2
 5% non-BRCA DDR
Specimen tested Tumor tissue
Central
Plasma or tumor tissue
Central/local
Tumor tissue
Central
Plasma or tumor tissue
Central analysis
Test used Targeted customized NGS panel FoundationOne CDx® FoundationOne CDx® Resolution-HRD®
FoundationOne CDx®
Genes screened 113 DDR genes ATM, BARD1, BRCA1, BRCA2, BRIP1, CDK12, CHEK2, FANCA, NBN, PALB2, RAD51, RAD51B, RAD51C, RAD51D, RAD54L ATM, ATR, BRCA1, BRCA2, CHEK2, FANCA, MLH1, MRE11A, NBN, PALB2, RAD51C ATM, BRCA1, BRCA2, BRIP1, CHEK2, FANCA, HDAC2, PALB2
Genomic alt. required Mono- and biallelic DDR alterations Biallelic or germline DDR alt.

mCRPC, metastatic castration-resistant prostate cancer; NGS, Next generation sequencing; PSA, prostate-specific antigen.

In light of these encouraging findings, the phase III trials PROfound and TRITON3 were developed (Table 2). PROfound6,37 has been the first randomized phase III biomarker-driven trial in prostate cancer aimed to assess the potential benefits of olaparib (300 mg bid) compared to a second ARSi (enzalutamide or abiraterone) in patients with mCRPC who had disease progression after receiving a prior ARSi, either in the context of castration-naïve or castration-resistant disease. The primary endpoint was radiologic progression-free survival (rPFS) in cohort A and the overall population. A total of 4858 tissue samples from 4047 patients were tested for genomic alterations in ATM, BRCA1, BRCA2, BARD1, BRIP1, CDK12, CHEK1, CHEK2, FANCl, PALB2, PPPR2A, RAD51B, RAD51C, and RAD54L. Next-generation sequencing was successful in 58% of samples (69% of patients). Out of these, 28% (778 patients) had a qualifying deleterious alteration in at least one of the 15 pre-specified genes. Finally, 387 patients met all the entry criteria and were enrolled in the trial. According to the identified mutation, patients were assigned to cohort A (BRCA1, BRCA2, or ATM alterations, n = 245) or cohort B (other genes assessed, n = 142). The frequency of mutations in cohort A was 13 (5%) in BRCA1, 127 (33%) in BRCA2, and 84 (21%) in ATM. Olaparib showed rPFS benefit, with an absolute increased survival of 3.8 months (HR 0.34, p < 0.001) in cohort A and 2.3 months (HR 0.49; 95%CI 0.38–0.63, p < 0.001) in the overall population. Olaparib also improved OS in cohort A (19.1 versus 14.7 months, HR 0.69, 95% CI 0.50–0.97) but not in cohort B (HR 0.96 95%CI 0.63–1.49). A benefit in OS was noted in the overall population (HR 0.79, 95% CI 0.61–1.03), mostly driven by patients in cohort A. Importantly, 66% of patients in the control arm crossed over to olaparib after progression and a sensitivity analysis adjusted for the crossover showed a 58% decrease in the risk of death for patients in cohort A. A gene-by-gene exploratory analysis suggested the greatest benefit from olaparib for BRCA2-altered patients and a less clear benefit for those with ATM alterations. The median duration of treatment was 7.6 months (0.03–29) in the olaparib group and 3.9 (0.6–29.1) in the control group. The most common adverse events related to olaparib were anemia (39%), nausea (36%), and fatigue/asthenia (32%). Olaparib was discontinued because of anemia in 7% of patients and because of neutropenia, thrombocytopenia, nausea, vomiting, or fatigue/asthenia in 1% of patients. One case of fatal acute myeloid leukemia was reported in a patient with a germline BRCA2 mutation diagnosed 54 days after discontinuation of olaparib. In view of these results, the FDA-approved olaparib for patients with alterations in the HRR genes tested in the PROFound study while EMA restricted the approval only to patients with BRCA1 and BRCA2 alterations.

Table 2.

Summary of phase III trials TRITON 3 and PROFOUND cohort A.

Study characteristics TRITON 3 PROFOUND – COHORT A
Rucaparib (n = 270) Physician’s choice second ARPi (n = 60) Olaparib (n = 162) Physician’s choice
second ARPi (n = 83)
BRCA1 29 (11%) 9 (12%) 8 (5%) 5 (6%)
BRCA2 172 (64%) 51 (68%) 80 (49%) 47 (57%)
ATM 69 (26%) 15 (20%) 60 (37%) 24 (29%)
Median age, years (range) 70 (45–90) 72 (54–92) 68 (47–86) 67 (49–86)
ECOG PS 0, n (%) 132 (49%) 33 (55%) 84 (52%) 34 (41%)
Baseline PSA, ng/mL 27 (0.1–1247) 29 (0–1039) 62.2 (21.9–280.4) 112.9 (34.3–317.1)
Gleason score ⩾8, n (%) 173 (64%) 37 (62%) 105/157 (67%) 54/80 (67%)
Measurable disease at baseline 106 (39%) 21 (35%) 95 (59%) 46 (55%)
Visceral metastasis at baseline 74 (27%) 46 (34%) 49 (30%) 23 (28%)
Prior ARSi
 1 100% 100% 130 (80%) 69 (83%)
 ⩾2 0 0 32 (20%) 14 (17%)
Prior Taxanes
 1 63 (23%)* 16 (27%)* 76 (47%) 32 (39%)
 ⩾2 0 0 29 (18%) 20 (24%)
Imaging-based PFS 10.2 versus 4.5 months
HR 0.47 (95%CI 0.34–0.66)
p < 0.0001
7.39 versus 3.55 months
HR 0.34 (95%CI 0.25–0.47)
p < 0.0001
Overall survival 23.6 versus 20.9 months$
HR 0.94 (95% CI, 0.72–1.23)
19.1 versus 14.7 months
HR 0.69 (95% CI 0.5–0.97)
p = 0.017

*Docetaxel for metastatic hormone-sensitive prostate cancer.

$

Interim overall survival data reported with 59% data maturity.

PFS, progression-free survival; PSA, prostate-specific antigen.

The TRITON-3 study 38 has assessed rucaparib monotherapy (600 mg bid) for the treatment of mCRPC patients with BRCA1, BRCA2, or ATM after progression to an ARSi. Of the 405 patients enrolled, 44 had alterations in BRCA1, 258 in BRCA2 and 103 in ATM. Patients were randomized to receive rucaparib or a physician’s choice (docetaxel or a second ARSi). At 62 months, the median rPFS in the intention-to-treat population was significantly longer in the rucaparib than in the control arm (10.2 months versus 6.4 months; HR 0.61, 95% CI 0.47–0.8). The difference in rPFS was driven by BRCA patients (11.2 months versus 6.4 months; HR 0.5, 95% CI 0.36–0.69) as no benefit was noted in the ATM-mutated population (8.1 versus 6.8 months; HR 0.95, 95% CI 0.59–1.52). TRITON-3 has provided the first direct comparison between a PARPi and docetaxel; in patients with BRCA1/2 alterations, treatment with rucaparib resulted in more prolonged rPFS (11.2 versus 8.3 months; HR 0.53 95% CI 0.37–0.77), while no difference was noted in ATM patients (8.1 versus 8.1 months, HR 1.1, 95% CI 0.57–2.11). In line with the PROfound study, compared to a second ARSi, rucaparib resulted in improved rPFS in patients with BRCA alterations (11.2 versus 4.5 months, HR 0.38, 95% CI 0.25–0.58) with less clear benefit in ATM patients (8.1 versus 5.7 months, HR 0.82, 95% CI 0.47–1.45). The most common adverse events related to rucaparib were fatigue (61%), nausea (50%), anemia (47%), and decreased appetite (36%). The most frequent grade ⩾3 toxicity were anemia (24%), neutropenia (7%), fatigue (7%), and thrombocytopenia (6%). No cases of myelodysplastic syndrome or acute myeloid leukemia have been reported. No mature OS data are still available. FDA has approved rucaparib for the treatment of BRCA-mutated mCRPC that has progressed to ARSi.

PARPi in combinations for the treatment of advanced prostate cancer

The role of PARPi in monotherapy to treat prostate tumors with alterations in BRCA1, BRCA2, and some other HRR genes has consistently been demonstrated in the studies mentioned above. Potential therapeutic synergies that could deepen response in these patients but may also expand the benefit of PARPi to a broader unselected population are now being addressed in multiple trials. Some studies are combining PARPi with other compounds that target alternative DDR nodes (i.e. Wee1, HSP90, or ATR inhibitors) or induce DNA damage3941 (i.e. 177Lutetium, 223 Radium) aiming to increase DNA damage during G1 and S phases of the cell cycle and to minimize repair during G2, leading to cell death. Combination with DNA-damaging chemotherapy 42 (i.e. carboplatin) would maximize the effect of DNA damage but is also challenging due to overlapping toxicities. A second strategy consists of combining PARPi with drugs targeting other biological pathways that are modulated and/or to modulate HRR function, such as the PI3K/AKT pathway, 43 Vascular Endotelial Growth Factors (VEGFR), 44 and Androgen Receptor (AR) signaling4547 pathways. Finally, the rationale for developing combinations of PARPi and immunotherapy is that the genomic instability associated with HRR and other DDR defects may lead to neoantigen production and T-cell activation. Furthermore, the accumulation of cytosolic DNA may activate the innate immune system through the cGAS-STING pathway, inducing interferon-mediated response. This would subsequently lead to the activation of natural killer cells and macrophages and the infiltration, proliferation, and antitumor response of CD4 and CD8 T cells into the tumor. Paradoxically, the STING pathway and PARP inhibition also activate the expression of PD-L1.4850

While most studies are still ongoing, the trials reported to date have shown limited antitumor activity from the combination of PARPi with cediranib, 44 pembrolizumab, 51 or nivolumab 52 in mCRPC patients without detectable HRR alterations and for those with HRR defects it is unclear whether the combinations add any benefit to PARPi in monotherapy. The most promising results have been reported from trials investigating the combination of PARPi with ARSi, with several lines of work showing a close interplay between the AR pathway and the DDR machinery, supporting the synergistic effect of ARSi and PARPi. It has been shown that AR transcriptionally stimulates the expression of a wide range of DNA repair genes, some of them through direct interaction of AR with AR binding sites in gene enhancer regions.53,54 Prominent among the AR-regulated DDR genes seem to be those encoding components of the nonhomologous end-joining repair (NHEJ) but also genes involved in homologous recombination, mismatch repair, base excision repair, and the Fanconi anemia pathway. In line with these observations, Li et al. 55 reported that in both, AR-dependent and AR-independent cell lines, enzalutamide reduced the expression of a specific set of HR genes, including BRCA1, RAD51C, RAD54L, and RMI2, thus creating an HR deficiency and a BRCA-loss-like state (referred to as ‘BRCAness’). Although AR inhibitor-induced ‘BRCAness’ may be transient and shallow, a treatment strategy in which enzalutamide was followed by olaparib resulted in improved antitumoral activity in murine prostate cancer xenografts compared to monotherapy with either agent. 55 On the other hand, beyond its role in DNA damage repair, PARP1 has been suggested to be involved in the transcriptional control of AR 56 as its enzymatic activity is enhanced and required for AR function, tumor growth, and progression to castration resistance. 57 Based on this, early exposure to PARP inhibition might modulate AR signaling with downstream effects that maintain hormone-responsive disease for a longer period. Multiple interactions between the AR and HRR pathways have been reported although it remains unclear which of these mechanisms play a role in the synergy between ARSi and PARPi and whether there is inter- or intra- patient heterogeneity.

The potential synergy between ARSi and PARPi was tested by STUDY 8, 58 a phase II study that compared abiraterone with abiraterone plus olaparib in 142 mCRPC patients unselected for HRR alterations. The trial met its primary endpoint, with a median PFS of 13.8 months (95% CI 10.8–30.4) from the combination, compared with 8.2 months (95% CI 5.5–9.7, p = 0.034) in the control arm. No significant difference was observed in the secondary endpoint of OS, although the study may have been underpowered. Due to collection and testing issues, it was not possible to establish the presence of HRR alterations in a significant proportion of patients and there may be a disbalance of these genomic events between arms. A 25% increase in the incidence of grade 3–4 adverse events was noted in the combination arm, largely due to anemia, as well as increased pneumonia and myocardial infarction events.

Despite its limitations, STUDY 8 paved the way for the development of phase III trials addressing the combination of PARPi with different abilities to inhibit their target and ARSi with different mechanisms of action (abiraterone is an androgen biosynthesis inhibitor while enzalutamide is an AR antagonist). It is unclear whether these different mechanisms of action may affect a potential synergy between PARPi and ARSi. The three randomized phase III trials reported to date (MAGNITUD, PROPEL, and TALAPRO-2) have evaluated PARPi and ARSi combinations as first-line treatment for mCRPC and shared rPFS as the primary outcome, but otherwise had remarkably different designs including the prospective or retrospective HRR testing, the stratifying factors, the percentage of patients with HRR and BRCA alterations or the prior use of ARSi (Table 3).

Table 3.

Summary of phase III trials MGANITUDE, PROPEL, and TALAPRO-2.

AR signaling inhibitor MAGNITUDE PROPEL TALAPRO-2
Abiraterone (AA) Abiraterone (AA) Enzalutamide
PARP inhibitor Niraparib Olaparib Talazoparib
Study design Inclusion criteria 1L mCRPC
BPI-SF ⩽3
⩽4 months AA for mCRPC
HRR alt only
1L mCRPC
ECOG PFS 0-1
No prior AA in mCRPC
ARSi allowed if stopped 12 months prior
All comers
1L mCRPC
ECOG PFS 0-1
Docetaxel and AA for mHSCP allowed
All comers
Molecular testing Prospective
Plasma: Resolution Biosciences
Tissue: FoundationOne®CDx
Retrospective
Tissue: FoundationOne®CDx
Plasma:FoundationOne®Liquid CDx
Prospective
Tissue: FoundationOne®CDx
Plasma: FoundationOne®Liquid CDx
Genes analyzed ATM, BRCA1, BRCA2, BRP1, CDK12, CHEK2, FANCA, HDAC2, PALB2 ATM, BRCA1, BRCA2, BARD1, BRIP1, CDK12, CHEK1, CHEK2, FANCL, PALB2, RAD51B, RAD51C, RAD51D, RAD54L ATM, ATR, BRCA1, BRCA2, CHEK2, FANCA, MLH1, MRE11A, NBN, PALB2, RAD51C
Stratification factors Prior docetaxel for mHSPC
Prior ARSi for nmCRPC or mHSPC
Prior AA for 1L mCRPC
BRCA1/2 versus non-BRCA HRR
Site of metastases
Prior taxane at mHSCP
HRR status
Prior AA or docetaxel for mHSCP
Primary endpoint rPFS by central review in HRRm rPFS by the investigator in all comers rPFS by central reviewer in all comers
Population Experimental arm Control arm Experimental arm Control arm Experimental arm Control arm
Patients 212 211 399 397 402 403
HRRm 100% 100% 28% 29% 21% 21%
Age, median (range), y 69 (45–199) 69 (43–88) 69 (43–91) 70 (46–88) 71 (41–90) 71 (36–91)
PSA at study entry (ng/mL) 21.4 (0–4826.5) 17.4 (0.1–4400) 17.9 (6.09–67) 16.81 (6.26–53.3) 18.2 (0.1–2796) 16.2 (0.1–2285)
ECOG
 0 130 (61%) 146 (69%) 286 (72%) 272 (68%) 259 (64%) 271 (67%)
 1 82 (39%) 65 (31%) 112 (28%) 124 (31%) 143 (36%) 132 (33%)
Site of metastases
 Bone 183 (86.3%) 170 (80.6%) 349 (88%) 339 (85%) 349 (87%) 342 (85%)
 Visceral 51 (24.1%) 39 (18.5%) 55 (14%) 60 (15%) 57 (14%) 77 (19%)
Prior docetaxel mHSPC 41 (19.3%) 44 (20.9%) 90 (23%) 89 (22%) 86 (21% 93 (23%)
Prior ARPi for nmCRPC/mHSPC 8 (3.8%) 5 (2.4%) 1 (0.3%) 0 21 (5%) 25 (6%)
Prior ARPi for L1 mCRPC 50 (23.6%) 48 (22.7%) 0 0 0 0
Efficacy rPFS all comers _________ 24.8 versus 16.6 months
HR 0.66 (95% CI 0.54-0.81)
NR versus 21.9 months
HR 0.63 (95% CI 0.5-0.78)
rPFS BRCA subgroup 19.5 versus 10.9 months
HR 0.55 (95% CI 0.36-0.79)
NR versus 8.4 months
HR 0.23 (95% CI 0.12-0.43)
NR versus 11 months
HR 0.20 (95%CI 0.11-0.36)
rPFS HRRm subgroup 16.5 versus 13.7 months
HR 0.73 (95% CI 0.56–0.96)
NR versus 13.9 months
HR 0.50 (95% CI 0.34–0.73)
27.9 vs 16.4 months
HR 0.46 (95% CI 0.3–0.7)
rPFS non-HRR/unknown _________ 24.1 versus 19 months
HR 0.76 (95% CI 0.60–0.97)
NR vs 22.5 months
HR 0.70 (95% CI 0.54–0.89)
OS all comers _________ 42.1 versus 34.7 months
HR 0.81 (95% CI 0.67–1.00)
36.4 versus NR months*
HR 0.89 (95% CI0.69–1.14)
OS BRCA subgroup HR 0.54 (95%CI 0.33–0.90)* NR versus 23 months
HR 0.29 (95% CI 0.14–0.56)
HR 0.61 (95% CI 0.31–1.23)*
OS HRR subgroup HR 0.70 (95%CI 0.49–0.99)* NR-28.5 months
HR 0.66 (95% CI 0.45–0.95)
NR versus 33.7 months*
HR 0.69 (95% CI 0.46–1.03)
OS non-HRR/unknown _________ 42.1 versus 38.9 months
HR 0.89 (95% CI 0.7–1.14)
Not reported yet
Time to PSA progression 18.5 versus 9.3 months
HR 0.57 (95% CI 0.43-0.76)
p < 0.001
NR versus 12 months
HR 0.55 95% CI 0.45-0.68)
26.7 versus 17.5 months
HR 0.72 (95% CI 0.58-0.89)
p = 0.002
Objective response rate (ORR) 60% versus 28%
p < 0.001
58% versus 48%
p = 0.041
62% versus 44%
p = 0.005
Safety Frequent Adverse Events (Any grade – ⩾G3) Experimental arm Control arm Experimental arm Control arm Experimental arm Control arm
 Anemia 46–30% 20–8% 46–15% 16–3% 66–43% 16–4%
 Thrombocytopenia 21–7% 9–2% <10% <10% 25–5% 4–0.7%
 Neutropenia 14–6% 6–1% <10% <10% 36–17% 7–1%
 Nausea 24–0.5% 14–0 28–0.3% 13–0.3% 21–0.5% 13–0.7%
 Fatigue/asthenia 26–3% 17–4% 37–2.3% 28–2% 34–4% 29–2%
 Embolic/thrombotic events <10% <10% 9–8% 6–4% 3–2% 1–1%
*

Immature overall survival data.

mCRPC, metastatic castration-resistant prostate cancer; NR, not reached; OS, overall survival; PSA, prostate-specific antigen; rPFS, radiologic progression-free survival.

The PROpel trial 46 randomized 796 men to receive olaparib (300 mg bid) and abiraterone acetate (1000 mg/day) plus prednisone (10 mg/day) vs placebo and abiraterone as first-line treatment for mCPRC. Patients could have received docetaxel in the metastatic hormone-sensitive (mHSPC) stage but no prior ARSi was allowed. Participants were stratified by site of distant metastases and prior docetaxel at mHSPC. Tumor tissue and plasma samples were collected at study entry but HRR status was retrospectively analyzed. Aggregate data from tumor tissue and circulating DNA were used to classify patients as HRR mutant (HRRm, 28.4%), non-HRR mutant (non-HRRm, 69.3%), or unknown (2.3%), without significant differences noted between treatment arms. The study included 85 patients with BRCA1/2 alterations. In the overall population, a benefit in rPFS was observed in patients receiving laparib and abiraterone (24.8 versus 16.6 months; HR 0.66, 95% CI 0.54–0.81), although the magnitude of the benefit varied by HRR status with BRCA altered patients benefiting the most (HR 0.23, 95% CI 0.12–0.43) followed by the broader HRRm subgroup (HR 0.50, 95% CI 0.34–0.73), and the Non-HRRm subgroup (HR 0.76; 95% CI 0.60–0.97). This benefit in rPFS only translated in more prolonged OS in patients with HRRm (HR 0.66, 95% CI 0.66–0.95) and particularly in those with BRCA alterations (HR 0.29; 95% CI 0.14–0.56). In the overall population, the 7.4 months improvement in OS (HR 0.81, 95% CI 0.67–1) was not statistically significant. The most frequent adverse events attributed to the combination included anemia (46%), fatigue/asthenia (37.2%), and nausea (28%). The most frequent grade ⩾3 side effects in the experimental arm were anemia (15%), venous embolisms (6.8%), hypertension (3.5%), and fatigue (2.3%). Olaparib plus abiraterone is currently approved by EMA as first-line therapy for mCRPC patients regardless of HRR status while the FDA restricted the approval to BRCA1 and BRCA2 mCRPC patients.

The TALAPRO-2 trial 47 explored the use of enzalutamide (160 mg/day) in combination with talazoparib (0.5 mg/day) as first-line therapy for mCRPC. Prior treatment with docetaxel or abiraterone for mHSPC and HRR status were stratification factors. In cohort 1, the study enrolled 805 patients who were prospectively tested for HRR status on tumor tissue samples, detecting HRR alterations in 20.9% of participants. Median rPFS was significantly more prolonged in the combination arm (not reached versus 21.9 months, HR 0.63, 95% CI 0.51–0.78) in the overall population with a greater benefit in the HRR-deficient population (HR 0.46, 95% CI 0.3–0.7). Importantly, an exploratory analysis limited to patients without detectable HRR in tumor tissue suggests a benefit in rPFS from combining enzalutamide with talazoparib (HR 0.66, 95% CI 0.49–0.91), supporting preclinical reports of a synergy from the inhibition of the AR and HR pathways. The TALAPRO-2 study also included a second cohort of patients (cohort 2, n = 230) to expand the HRR deficient population to a total of 399, including 155 patients with BRCA1/2 alterations, 59 and confirmed the rPFS benefit from the combination in this population (HR 0.44, 95% CI 0.32–0.60). Consistently with the PROPEL study, in TALAPRO-2 the greatest benefit was also observed in BRCA patients (HR 0.20, 95% CI 0.11–0.36). An exploratory analysis of cohort 2 suggests that as in monotherapy, the antitumor efficacy of PARPi combined with ARSi may depend on the HRR gene altered. The data presented at ASCO 2023 59 suggest limited benefit from the combination in patients with ATM alterations, but a potential advantage for patients with CDK12 alterations that do not seem to respond to PARPi alone. Anemia was the most frequent side effect reported in the experimental arm (66%), followed by neutropenia (36%), fatigue (34%), thrombocytopenia (25%), decreased appetite (22%), and nausea (21%). Grade ⩾3 hematological toxicities were frequent in the combination arm (anemia 46%, neutropenia 18%, and thrombocytopenia 7%). 47 Talazoparib plus enzalutamide is currently approved by the FDA as first-line therapy for HRR-deficient mCRPC patients.

MAGNITUDE45,60 investigated the benefit of niraparib plus abiraterone as first-line treatment for mCRPC patients. An earlier phase Ib study recommended a dose reduction of niraparib to 200 mg/day (from 300 mg/day) when combined with abiraterone. 61 Eligible patients were prospectively screened for alterations in ATM, BRCA1, BRCA2, BRP1, CDK12, CHEK2, FANCA, HDAC2, and PALB2 and included in cohort 1 if an alteration was present or in cohort 2 if no alteration was found. Patients could start abiraterone prior to randomization for up to 4 months while testing was being performed. Run-in treatment with abiraterone and prior mHSPC treatment were the stratification factors. cohort 2 was closed after a pre-planned futility analysis suggested a lack of benefit from the combination in patients without HRR alterations. Cohort 1 finally included 423 patients, of which 190 (45%) had BRCA1/2 alterations. The addition of niraparib to abiraterone resulted in more prolonged rPFS (16.5 versus 13.7 months; HR 0.76, 95% CI 0.6–0.97) with the greatest benefit also noted in the BRCA1/2 subgroup (16.6 versus 10.9 months; HR 0.53, 95% CI 0.36–0.79). Anemia (46%), hypertension (31%), constipation (31%), fatigue (26%), nausea (24%), thrombocytopenia (21.2%), and dyspnea (16%) were more frequent in the experimental arm. In this group, frequent grade ⩾3 toxicities included anemia (28.3%), hypertension (14.6%), and neutropenia (5.2%). Niraparib in combination with abiraterone has been approved by both, the FDA and EMA, as first-line therapy for mCRPC patients with BRCA1/2 alterations.

An important conclusion from these three studies, PROPEL, TALAPRO-2, and MAGNITUDE is that a benefit hierarchy aligned with known biology can be established: BRCA2-deficient tumors >> all HRR-deficient tumors >> unselected patients >> HRR-proficient tumors. 62

Future directions

PARPi are the first targeted therapies approved for the treatment of advanced prostate cancer but their use is being limited by the low testing rates. 63 Increased awareness and education are needed to encourage testing but also broader access to somatic genomic profiling to ensure that all patients with advanced disease have their tumors tested.

A major limitation to implementing PROPEL, TALAPRO-2, and MAGNITUDE in clinical practice is the differences between the population enrolled in those studies, mostly unexposed to prior ARSi, and the current and future mCRPC patients, the majority of whom have already received an ARSi for the treatment of mHSPC or non-metastatic castration-resistant prostate cancer (nmCRPC). The available evidence demonstrates that treatment with a second ARSi after disease progression to a prior one results in limited responses6466 and it has not been demonstrated that resistance to ARSi could be overcome by the addition of a PARPi. Furthermore, an exploratory analysis of the MAGNITUDE trial, 67 that allowed starting abiraterone before randomization reported decreased antitumor efficacy on patients who were on abiraterone ⩾2–4 months before starting niraparib, suggesting that the offset impact negatively the potential synergistic effect.

It will be important to understand the synergistic effect of ARSi and PARPi in early disease stages. AMPLITUDE 68 and TALAPRO-3 69 are, respectively, evaluating the combination of niraparib plus abiraterone and talazoparib plus enzalutamide in HRR-deficient mHSPC patients, previously unexposed to ARSi. This strategy is expected to extend the duration of hormone sensitivity and modify disease trajectory.

As mentioned earlier, patients will eventually become resistant to PARPi and there is currently no evidence for the management of patients with known BRCA1/2 alterations and/or HRR defects after disease progression to PARPi. Preclinical studies have shown that PARPi and platinum-resistant cancer models may be re-sensitized to PARPi when combined with other drugs that target molecular vulnerabilities, including cell cycle checkpoints 70 and replication stress. 71 Early-phase trials have shown a signal of activity with PARP inhibition re-challenge in combination with ATR or Wee1 inhibition in various tumor types. Other potent inhibitors that target different key nodes along the DNA damage repair cascade are also in clinical development, including CHK1/2, DNA-PK, ATM, and POLθ inhibitors, 72 which may represent other potential combination partners for PARPi.

Finally, a new generation of selective PARP1 inhibitors without activity upon PARP2, expected to retain anticancer efficacy with decreased toxicity, is being developed. 73

Acknowledgments

None.

Footnotes

Contributor Information

Maria Teresa Bourlon, Hemato-Oncology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.

Paola Valdez, Hemato-Oncology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.

Elena Castro, Department of Medical Oncology, Hospital Universitario 12 de Octubre, Av. Cordoba s/n, 28041, Madrid, Spain.

Declarations

Ethics approval and consent to participate: Not applicable.

Consent for publication: Not applicable.

Author contributions: Maria Teresa Bourlon: Conceptualization; Methodology; Supervision; Writing – original draft; Writing – review & editing.

Paola Valdez: Conceptualization; Investigation; Writing – original draft; Writing – review & editing.

Elena Castro: Conceptualization; Data curation; Investigation; Methodology; Project administration; Supervision; Validation; Writing – original draft; Writing – review & editing.

Funding: The authors disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: EC is supported by Asociación Española Contra el Cancer (grant CLSEN223433).

MTB reports Speaker’s bureau: Janssen, Pfizer, Foundation One and travel support from Janssen and Pfizer. PV has no conflicts to declare. EC reports consulting/advisory role for Astellas Pharma, AstraZeneca, Bayer, Daiichi-Sankyo, Janssen, Lilly, Medscape, Merck, MSD Oncology, and Pfizer; and speakers’ bureau from AstraZeneca, Janssen, Novartis, and Pfizer; travel support from AstraZeneca, Bayer, Janssen, and Pfizeer; honoraria from Astellas Pharma, AstraZeneca, Bayer, Clovis Oncology, Janssen-Cilag, Medscape, Pfizer, Roche, Telix; and research funding from AstraZeneca, Bayer, Janssen.

Availability of data and materials: Not applicable.

References

  • 1. Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021; 71: 209–249. [DOI] [PubMed] [Google Scholar]
  • 2. Gillessen S, Bossi A, Davis ID, et al. Management of patients with advanced prostate cancer-metastatic and/or castration-resistant prostate cancer: report of the advanced prostate cancer consensus conference (APCCC) 2022. Eur J Cancer 2023; 185: 178–215. [DOI] [PubMed] [Google Scholar]
  • 3. Robinson D, Van Allen EM, Wu YM, et al. Integrative clinical genomics of advanced prostate cancer. Cell 2015; 161: 1215–1228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Armenia J, Wankowicz SAM, Liu D, et al. The long tail of oncogenic drivers in prostate cancer. Nat Genet 2018; 50: 645–651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Mateo J, Seed G, Bertan C, et al. Genomics of lethal prostate cancer at diagnosis and castration resistance. J Clin Invest 2020; 130: 1743–1751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. de Bono J, Mateo J, Fizazi K, et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med 2020; 382: 2091–2102. [DOI] [PubMed] [Google Scholar]
  • 7. Warner E, Herberts C, Fu S, et al. BRCA2, ATM, and CDK12 defects differentially shape prostate tumor driver genomics and clinical aggression. Clin Cancer Res 2021; 27: 1650–1662. [DOI] [PubMed] [Google Scholar]
  • 8. Schweizer MT, Sivakumar S, Tukachinsky H, et al. Concordance of DNA repair gene mutations in paired primary prostate cancer samples and metastatic tissue or cell-free DNA. JAMA Oncol 2021; 7: 1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Truong H, Breen K, Nandakumar S, et al. Gene-based confirmatory germline testing following tumor-only sequencing of prostate cancer. Eur Urol 2023; 83: 29–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Kuzbari Z, Bandlamudi C, Loveday C, et al. Germline-focused analysis of tumour-detected variants in 49,264 cancer patients: ESMO Precision Medicine Working Group recommendations. Ann Oncol 2023; 34: 215–227. [DOI] [PubMed] [Google Scholar]
  • 11. Carter HB, Helfand B, Mamawala M, et al. Germline mutations in ATM and BRCA1/2 are associated with grade reclassification in men on active surveillance for prostate cancer. Eur Urol 2019; 75: 743–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Castro E, Goh C, Leongamornlert D, et al. Effect of BRCA mutations on metastatic relapse and cause-specific survival after radical treatment for localised prostate cancer. Eur Urol 2015; 68: 186–193. [DOI] [PubMed] [Google Scholar]
  • 13. Castro E, Goh C, Olmos D, et al. Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer. J Clin Oncol 2013; 31: 1748–1757. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Castro E, Romero-Laorden N, Del Pozo A, et al. 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; 37: 490–503. [DOI] [PubMed] [Google Scholar]
  • 15. Annala M, Struss WJ, Warner EW, et al. Treatment outcomes and tumor loss of heterozygosity in germline dna repair-deficient prostate cancer. Eur Urol 2017; 72: 34–42. [DOI] [PubMed] [Google Scholar]
  • 16. Lozano R, Castro E, Lopez-Campos F, et al. Impact of concurrent tumour events on the prostate cancer outcomes of germline BRCA2 mutation carriers. Eur J Cancer 2023; 185: 105–118. [DOI] [PubMed] [Google Scholar]
  • 17. Olmos D, Lorente D, Alameda D, et al. Presence of somatic/germline homologous recombination repair (HRR) mutations and outcomes in metastatic castration-resistant prostate cancer (mCRPC) patients (pts) receiving first-line (1L) treatment stratified by BRCA status. J Clin Oncol 2023; 41: 5003–5003. [Google Scholar]
  • 18. Rescigno P, Gurel B, Pereira R, et al. Characterizing CDK12-mutated prostate cancers. Clin Cancer Res 2021; 27: 566–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Antonarakis ES, Isaacsson Velho P, Fu W, et al. CDK12-altered prostate cancer: clinical features and therapeutic outcomes to standard systemic therapies, poly (ADP-Ribose) polymerase inhibitors, and PD-1 inhibitors. JCO Precis Oncol 2020; 4: 370–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Hakme A, Wong HK, Dantzer F, et al.The expanding field of poly(ADP-ribosyl)ation reactions. ‘protein modifications: beyond the usual suspects’ review series. EMBO Rep 2008; 9: 1094–1100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Langelier MF, Eisemann T, Riccio AA, et al. PARP family enzymes: regulation and catalysis of the poly(ADP-ribose) posttranslational modification. Curr Opin Struct Biol 2018; 53: 187–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Murai J, Huang SY, Renaud A, et al. Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib. Mol Cancer Ther 2014; 13: 433–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Murai J, Huang SY, Das BB, et al. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res 2012; 72: 5588–5599. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Mateo J, Lord CJ, Serra V, et al. A decade of clinical development of PARP inhibitors in perspective. Ann Oncol 2019; 30: 1437–1447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Rudolph J, Jung K, Luger K. Inhibitors of PARP: Number crunching and structure gazing. Proc Natl Acad Sci USA 2022; 119: e2121979119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Hopkins TA, Ainsworth WB, Ellis PA, et al. PARP1 trapping by PARP inhibitors drives cytotoxicity in both cancer cells and healthy bone marrow. Mol Cancer Res 2019; 17: 409–419. [DOI] [PubMed] [Google Scholar]
  • 27. Farres J, Martin-Caballero J, Martinez C, et al. Parp-2 is required to maintain hematopoiesis following sublethal gamma-irradiation in mice. Blood 2013; 122: 44–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Gonzalez-Ochoa E, Oza AM. An attempt to stretch the benefit: Re-challenge with PARP inhibitors in ovarian cancer. Clin Cancer Res 2023; 29: 2563–2566. [DOI] [PubMed] [Google Scholar]
  • 29. Fong PC, Boss DS, Yap TA, et al. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carriers. N Engl J Med 2009; 361: 123–134. [DOI] [PubMed] [Google Scholar]
  • 30. Mateo J, Carreira S, Sandhu S, et al. DNA-repair defects and olaparib in metastatic prostate cancer. N Engl J Med 373: 1697–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Mateo J, Porta N, McGovern UB, et al. TOPARP-B: a phase II randomized trial of the poly(ADP)-ribose polymerase (PARP) inhibitor olaparib for metastatic castration resistant prostate cancers (mCRPC) with DNA damage repair (DDR) alterations. J Clin Oncol 2019; 37: 5005–5005. [Google Scholar]
  • 32. Abida W, Campbell D, Patnaik A, et al. Non-BRCA DNA damage repair gene alterations and response to the PARP inhibitor rucaparib in metastatic castration-resistant prostate cancer: analysis from the phase 2 TRITON2 study. Clin Cancer Res 2020; 26: 2487–2496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Abida W, Campbell D, Patnaik A, et al. Rucaparib for the treatment of metastatic castration-resistant prostate cancer associated with a DNA damage repair gene alteration: final results from the phase 2 TRITON2 study. Eur Urol 2023; 84: 321–330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Abida W, Patnaik A, Campbell D, et al. Rucaparib in men with metastatic castration-resistant prostate cancer harboring a BRCA1 or BRCA2 gene alteration. J Clin Oncol 2020; 38: 3763–3772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. de Bono JS, Mehra N, Scagliotti GV, et al. Talazoparib monotherapy in metastatic castration-resistant prostate cancer with DNA repair alterations (TALAPRO-1): an open-label, phase 2 trial. Lancet Oncol 2021; 22: 1250–1264. [DOI] [PubMed] [Google Scholar]
  • 36. Smith MR, Scher HI, Sandhu S, et al. 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; 23: 362–373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Hussain M, Mateo J, Fizazi K, et al. Survival with olaparib in metastatic castration-resistant prostate cancer. N Engl J Med, 2020; 383: 2345–2357. [DOI] [PubMed] [Google Scholar]
  • 38. Fizazi K, Piulats JM, Reaume MN, et al. Rucaparib or physician’s choice in metastatic prostate cancer. N Engl J Med 2023; 388: 719–732. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Pan E, Xie W, Ajmera A, et al. A phase I study of combination olaparib and radium-223 in men with metastatic castration-resistant prostate cancer (mCRPC) with bone metastases (COMRADE). Mol Cancer Ther 2023; 22: 511–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Sandhu S, Joshua AM, Emmett L, et al. LuPARP: Phase 1 trial of 177Lu-PSMA-617 and olaparib in patients with metastatic castration resistant prostate cancer (mCRPC). J Clin Oncol 2023; 41: 5005–5005.37639648 [Google Scholar]
  • 41. Konstantinopoulos PA, Cheng SC, Supko JG, et al. Combined PARP and HSP90 inhibition: preclinical and Phase 1 evaluation in patients with advanced solid tumours. Br J Cancer 2022; 126: 1027–1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Pilié PG, Tidwell RS, Viscuse PV, et al. Randomized phase II study of olaparib (Ola) maintenance following cabazitaxel-carboplatin induction chemotherapy (CabCarb) in men with aggressive variant prostate cancer (AVPC). J Clin Oncol 2023; 41: 196–196. [Google Scholar]
  • 43. Yap TA, Kristeleit R, Michalarea V, et al. Phase I trial of the PARP inhibitor olaparib and AKT inhibitor capivasertib in patients with BRCA1/2- and non-BRCA1/2-mutant cancers. Cancer Discov 2020; 10: 1528–1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Kim JW, McKay RR, Radke MR, et al. Randomized trial of olaparib with or without cediranib for metastatic castration-resistant prostate cancer: the results from National Cancer Institute 9984. J Clin Oncol 2023; 41: 871–880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Chi KN, Sandhu S, Smith MR, 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. Ann Oncol 2023; 34: 772–782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Clarke NW, Armstrong AJ, Thiery-Vuillemin A, et al. Final overall survival (OS) in PROpel: Abiraterone (abi) and olaparib (ola) versus abiraterone and placebo (pbo) as first-line (1L) therapy for metastatic castration-resistant prostate cancer (mCRPC). J Clin Oncol 2023; 41: LBA16-LBA16. [Google Scholar]
  • 47. Agarwal N, Azad AA, 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 2023; 402: 291–303. [DOI] [PubMed] [Google Scholar]
  • 48. Jiao S, Xia W, Yamaguchi H, et al. PARP inhibitor upregulates PD-L1 expression and enhances cancer-associated immunosuppression. Clin Cancer Res 2017; 23: 3711–3720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Touat M, Sourisseau T, Dorvault N, et al. DNA repair deficiency sensitizes lung cancer cells to NAD+ biosynthesis blockade. J Clin Invest 2018; 128: 1671–1687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Revythis A, Limbu A, Mikropoulos C, et al. Recent insights into PARP and immuno-checkpoint inhibitors in epithelial ovarian cancer. Int J Environ Res Public Health 2022; 19: 8577. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Antonarakis ES, Park SH, Goh JC, et al. Pembrolizumab plus olaparib for patients with previously treated and biomarker-unselected metastatic castration-resistant prostate cancer: the randomized, open-label, PHASE III KEYLYNK-010 trial. J Clin Oncol 2023; 41: 3839–3850. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Fizazi K, Retz M, Petrylak DP, et al. Nivolumab plus rucaparib for metastatic castration-resistant prostate cancer: results from the phase 2 CheckMate 9KD trial. J Immunother Cancer 2022; 10: e004761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Polkinghorn WR, Parker JS, Lee MX, et al. Androgen receptor signaling regulates DNA repair in prostate cancers. Cancer Discov 2013; 3: 1245–1253. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Goodwin JF, Schiewer MJ, Dean JL, et al. A hormone-DNA repair circuit governs the response to genotoxic insult. Cancer Discov 2013; 3: 1254–1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Li L, Karanika S, Yang G, et al. Androgen receptor inhibitor-induced ‘BRCAness’ and PARP inhibition are synthetically lethal for castration-resistant prostate cancer. Sci Signal 2017; 10: eaam7479 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Schiewer MJ, Goodwin JF, Han S, et al. Dual roles of PARP-1 promote cancer growth and progression. Cancer Discov 2012; 2: 1134–1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Schiewer MJ, Knudsen KE: Transcriptional roles of PARP1 in cancer. Mol Cancer Res 2014; 12: 1069–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58. Clarke N, Wiechno P, Alekseev B, et al. Olaparib combined with abiraterone in patients with metastatic castration-resistant prostate cancer: a randomised, double-blind, placebo-controlled, phase 2 trial. Lancet Oncol 2018; 19: 975–986. [DOI] [PubMed] [Google Scholar]
  • 59. Fizazi K, Azad A, Matsubara N, et al. TALAPRO-2: phase 3 study of talazoparib (TALA) + enzalutamide (ENZA) versus placebo (PBO) + ENZA as first-line (1L) treatment for patients (pts) with metastatic castration-resistant prostate cancer (mCRPC) harboring homologous recombination repair (HRR) gene alterations. J Clin Oncol 2023; 41: 5004–5004. [Google Scholar]
  • 60. Chi KN, Rathkopf D, Smith MR, et al. Niraparib and Abiraterone Acetate for Metastatic Castration-Resistant Prostate Cancer. J Clin Oncol 2023; 41: 3339–3351. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Saad F, Chi KN, Shore ND, et al. Niraparib with androgen receptor-axis-targeted therapy in patients with metastatic castration-resistant prostate cancer: safety and pharmacokinetic results from a phase 1b study (BEDIVERE). Cancer Chemother Pharmacol 88:25-37, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Beije N, Abida W, Antonarakis ES, et al. PARP inhibitors for prostate cancer: tangled up in PROfound and PROpel (and TALAPRO-2) Blues. Eur Urol 2023; 84: 253–256. [DOI] [PubMed] [Google Scholar]
  • 63. Leith A, Ribbands A, Kim J, et al. Real-world homologous recombination repair mutation testing in metastatic castration-resistant prostate cancer in the USA, Europe and Japan. Future Oncol 2022; 18: 937–951. [DOI] [PubMed] [Google Scholar]
  • 64. Khalaf DJ, Annala M, Taavitsainen S, et al. Optimal sequencing of enzalutamide and abiraterone acetate plus prednisone in metastatic castration-resistant prostate cancer: a multicentre, randomised, open-label, phase 2, crossover trial. Lancet Oncol 2019; 20: 1730–1739. [DOI] [PubMed] [Google Scholar]
  • 65. de Wit R, de Bono J, Sternberg CN, et al. Cabazitaxel versus abiraterone or enzalutamide in metastatic prostate cancer. N Engl J Med 2019; 381: 2506–2518. [DOI] [PubMed] [Google Scholar]
  • 66. Attard G, Borre M, Gurney H, et al. Abiraterone alone or in combination with enzalutamide in metastatic castration-resistant prostate cancer with rising prostate-specific antigen during enzalutamide treatment. J Clin Oncol 2018; 36: 2639–2646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Castro E, Chi KN, Sandhu S, et al. Impact of run-in treatment with abiraterone acetate and prednisone (AAP) in the MAGNITUDE study of niraparib (NIRA) and AAP in patients (pts) with metastatic castration-resistant prostate cancer (mCRPC) and homologous recombination repair (HRR) gene alterations. J Clin Oncol 2023; 41: 172–172. [Google Scholar]
  • 68. Rathkopf DE, Chi KN, Olmos D, et al. AMPLITUDE: a study of niraparib in combination with abiraterone acetate plus prednisone (AAP) versus AAP for the treatment of patients with deleterious germline or somatic homologous recombination repair (HRR) gene-altered metastatic castration-sensitive prostate cancer (mCSPC). J Clin Oncol 2021; 39: TPS176-TPS176. [Google Scholar]
  • 69. Agarwal N, Saad F, Azad A, et al. TALAPRO-3: a phase 3, double-blind, randomized study of enzalutamide (ENZA) plus talazoparib (TALA) vs placebo plus ENZA in patients with DDR gene-mutated, metastatic castration-sensitive prostate cancer (mCSPC). J Clin Oncol 41: TPS279–TPS279. [Google Scholar]
  • 70. Kim H, Xu H, George E, et al. Combining PARP with ATR inhibition overcomes PARP inhibitor and platinum resistance in ovarian cancer models. Nat Commun 2020; 11: 3726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Parsels LA, Karnak D, Parsels JD, et al. PARP1 trapping and DNA replication stress enhance radiosensitization with combined WEE1 and PARP inhibitors. Mol Cancer Res 2018; 16: 222–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72. Yap TA, Plummer R, Azad NS, et al. The DNA damaging revolution: PARP inhibitors and beyond. ASCO Educ Book 2019; 39: 185–195. [DOI] [PubMed] [Google Scholar]
  • 73. Illuzzi G, Staniszewska AD, Gill SJ, et al. Preclinical characterization of AZD5305, a next-generation, highly selective PARP1 inhibitor and trapper. Clin Cancer Res 2022; 28: 4724–4736. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Therapeutic Advances in Medical Oncology are provided here courtesy of SAGE Publications

RESOURCES