Back to Journals » OncoTargets and Therapy » Volume 18

The Landscape of PARP Inhibitors in Solid Cancers

Authors Muzzana M, Broggini M , Damia G

Received 15 October 2024

Accepted for publication 5 February 2025

Published 2 March 2025 Volume 2025:18 Pages 297—317

DOI https://doi.org/10.2147/OTT.S499226

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr John Riches



Marta Muzzana,1 Massimo Broggini,2 Giovanna Damia2

1Oncology Department, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy; 2Experimental Oncology Department, Istituto Di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy

Correspondence: Massimo Broggini, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, via Mario Negri, Milan, 2 20156, Italy, Email [email protected]

Abstract: PARP inhibitors are a class of agents that have shown significant preclinical activity in models defective in homologous recombination (HR). The identification of synthetic lethality between HR defects and PARP inhibition led to several clinical trials in tumors with known HR defects (initially mutations in BRCA1/2 genes and subsequently in other genes involved in HR). These studies demonstrated significant responses in breast and ovarian cancers, which are known to have a significant proportion of patients with HR defects. Since the approval of the first PARP inhibitor (PARPi), olaparib, several other inhibitors have been developed, expanding the armamentarium available to clinicians in this setting. The positive results obtained in breast and ovarian cancer have expanded the use of PARPi in other solid tumors with HR defects, including prostate and pancreatic cancer in which these defects have been identified. The clinical trials have demonstrated responses to PARPi which are now also available for the subset of patients with prostate and pancreatic cancer with HR defects. This review summarizes the results obtained in solid tumors with PARPi and their potential use when combined with other agents, including immune checkpoint inhibitors that are likely to further increase the survival of these patients which still needs a dramatic improvement.

Keywords: DNA damage, homologous recombination, PARP inhibitors, solid tumors, BRCA

Introduction

Poly-ADP-ribose polymerase inhibitors (PARPi) represent one of the first example of synthetic lethality approach in oncology.1 Synthetic lethality refers to a genetic interaction in which inactivation of either gene individually has no effect on cell viability, while their concomitant loss of function causes cell death. Importantly, the inactivation can be a gene deletion, an inactivating mutation or a pharmacological treatment. PARPi were shown to be in synthetic lethality with BRCA1/2 (breast-related cancer antigen protein 1 and 2) mutations, which favored their clinical use in cancers harboring these mutations. However, their use was later extended to a broader clinical context, in tumors with deficiency in homologous recombination (HR) repair. Deficiency in HR (condition known as BRCAness or HRD) is due to functional inactivation of mutation/deletion and/or lack of expression (due to hypermethylation) of genes involved in the HR repair pathway.2 HR is a pathway that repairs the DNA double-strand breaks (DNA-DSBs) in an error-free manner because it uses the sister chromatid as a template with no genetic loss/alteration. The accuracy and fidelity of DNA-DSBs is of paramount importance as these are the most deleterious DNA lesions and can result in chromosomal aberrations, insertions and deletions and many other mutagenic outcomes that promote tumorigenesis.3

Treatment of HRD cells with inhibitors of PARPi results in cell death, with an almost 100-fold difference in sensitivity between HR-proficient and HR-deficient cells.4,5 The basis of the synthetic interaction is not fully understood and may be based on the inability of PARPi-treated cells to process DNA single strands, generating DNA-DSBs, that are highly toxic in HR-deficient cells; on the ability of PARPi to trap PARP1 on DNA with the subsequent accumulation of DNA damage,6 replication fork stalling and cell death,7 and on the accumulation of replication gaps.8 In addition, the single-strand DNA breaks in HRD cells treated with PARPi cause replication fork collapse and the resulting DNA-DSBs, which cannot be repaired by HR, are processed by error-prone non-homologous end joining (NHEJ), with the accumulation of genetic damage ultimately leading to cell death.

BRCA1 and BRCA2 are tumor suppressor genes that are mutated in breast, ovarian, pancreatic and prostate cancers. Germline mutations in the BRCA1/2 genes significantly increase the lifetime risk of developing breast (up to 85%) and ovarian cancer (15–56%),9 pancreatic cancer (2–7%)10 and prostate cancer,11 usually by inactivating the second allele, resulting in almost complete loss of protein function. BRCA1/2 proteins play a key role in HR and their functional inactivation results in HRD with increased levels of unrepaired DNA-DSBs that promote tumorigenesis.12 Since HR is a multi-step process involving many other proteins, HR deficiency has been reported to be caused by mutations in other genes (eg, PALB2, RAD51C, BRIP1, etc.) found in several cancers.13

Four PARPi (olaparib, rucaparib, niraparib and talazoparib) are now approved in the clinic with different indications (Figure 1) mainly in BRCA1/2 mutated/HRD tumors, including ovarian carcinoma, breast, and pancreatic with very interesting results. Even PARPi activity has also been reported in HD proficient (HRP) tumors,14 suggesting that other defects not detectable by HRD tests may contribute to their anticancer activity, the highest activity has been reported in HRD tumors and HRD is indeed considered the most important predictive biomarker of response to PARPi. In the last decades, a number of HRD tests have been developed that have been the companion tests for the development of PARPi in the clinical setting (recently reviewed in).15

Figure 1 Structure of the four FDA and EMA-approved PARPi.

We will summarize the most important clinical data of PARPi in ovarian, breast, pancreatic and prostate cancer alone or in combination therapy. Specifically, we will describe and discuss the trials in these different tumor types that lead to the approval of the different PARPi in specific settings. We will also discuss the possible mechanism of PARPi resistance and the new emerging PARPi, designed to be more specific, more potent and potentially less toxic that are now being tested in clinical trials.

Clinical Results in Ovarian Cancer

PARPi were first tested in ovarian cancer (OC), based on the evidence that 50% of the high-grade serous ovarian carcinomas are HRD2 and at least 17 Phase III clinical trials have been published (recently reviewed in).16 Table 1 shows the different PARPi and their current FDA and EMA approved indications in OC.

Table 1 PARP I Approved by FDA and EMA in Ovarian and Breast Cancer

Table 2 reports the results of the most important trials of PARPi in OC. Olaparib was approved as maintenance therapy for newly diagnosed OC based on the results of the SOLO1 trial, which showed that 2-years olaparib treatment extended the progression-free survival (PFS) in patients with BRCA1/2 mutations and that the PFS benefit was maintained 3 years after discontinuation of olaparib treatment.17 Similar results were obtained with niraparib in the PRIMA trial,18 which further extended the activity of niraparib not only in patients with BRCA1/2 mutations but also in patients with HRD, and with rucaparib in the ATHENA-MONO trial.19

Table 2 Summary of the Phase III Trials Assessing the Use of PARPi in Patients with Ovarian Cancer

The PAOLA-1 trial evaluated the addition of olaparib maintenance to bevacizumab after first-line platinum-taxane therapy and demonstrated a statistically significant increase in PFS in the olaparib+bevacizumab (22.1 months) versus placebo+bevacizumab (16.6 months) and this effect appeared to be greater in HRD patients;22 however, no survival benefit was later observed.

In the setting of recurrent platinum-sensitive OC, olaparib as maintenance therapy in patients with gBRCAmut (BRCA germline mutation) was associated with a statistically significant increase in PFS in the SOLO231 and in the Study 19 trials.32 The ARIEL3 trial evaluated the effect of rucaparib treatment versus placebo as maintenance therapy in platinum-sensitive relapsed OC with a significant improvement in PFS benefit independent of BRCA mutations or HRD status.29 However, no overall survival (OS) benefit was demonstrated.33 In the same setting, niraparib was tested versus placebo in both patients with gBRCAmut and non-gBRCAmut and again a significant PFS improvement was observed regardless of BRCA mutation or HRD tumor status.34 All of these studies support the indication of these drugs as maintenance monotherapy in relapsed-sensitive OC, establishing a new standard of care; interestingly, for some of the drugs, the effect was independent of the BRCA mutation and HRD status.

The SOLO3 trial compared olaparib (single agent) versus non-platinum-based chemotherapy (PDL, gemcitabine or topotecan) in platinum-sensitive gBRCAmut who had already received ≥2 prior platinum-based chemotherapies. While in the primary analysis an increase in OS and PFS in the olaparib arm compared to single-agent chemotherapy was reported, in the final analysis, the OS was similar, and only the time to second objective disease progression (PFS2) was longer in the olaparib arm.35 The efficacy of rucaparib was evaluated in a phase III trial in OC patients with germline and somatic BRCA mutations, who had already received ≥2 prior lines of chemotherapy, with varying degrees of platinum sensitivity compared to standard chemotherapy (ARIEL4).36 Despite the PFS advantage of rucaparib over chemotherapy (7.4 months −95% Confidence Interval – CI-: 7.3–9.1 versus 5.7 months 95% CI: 5.5–7.3), the median OS was 19.4 months and 25.4 months, respectively. Although these data could be inferred from the high crossover rate (69%), Clovis Oncology voluntarily withdrew rucaparib in this setting.16

PARPi have also been studied in recurrent and platinum-resistance settings, although very few trials considered the possible common mechanism of resistance between platinum and PARPi.37 In the CLIO trial, olaparib monotherapy was comparable to chemotherapy in platinum-resistant OC, including patients with gBRCA wild-type.38 An overall response rate of 28% (95% CI: 15.6–42.6; p=0.00053) was observed in the QUADRA trial evaluating niraparib monotherapy (as ≥4 lines of therapy) and was evaluated in 463 patients with disease recurrence within 6 months of last platinum-based therapy.39 The effect was more pronounced in HRD, platinum-sensitive disease. Olaparib monotherapy was found to have activity in patients with gBRCAmut advanced OC who had received more than three prior lines of chemotherapy (overall response rate – ORR – 34%; CI: 26–42).40

The introduction of PARPi in the treatment of OC has clearly changed the therapeutic approach in this tumor type. However, the updated long-term OS data in the recurrent setting (both as maintenance therapy after completion of chemotherapy and as monotherapy) did not correlate with the longer PFS survival reported in various studies. Final OS data from ARIEL4 were 19.4 months in the rucaparib arm versus 25.4 months with standard of care.36 Similar data were seen in the SOLO3 trial, with the post-hoc subgroup analysis showing a potential detrimental effect of olaparib compared to standard of care. Concerns have been raised in the relapse maintenance setting. Long-term follow-up data from the phase III NOVA trial, which evaluated the role of niraparib versus placebo in 553 patients with platinum-sensitive recurrent ovarian cancer (gBRCAmut and non-gBRCAmut cohorts) after standard platinum therapy, reported no difference in OS, although a trend over increased OS was observed in the niraparib-treated gBRAmut cohort versus placebo-treated patients (43.8 versus 34.1 months; Hazard Ratio HR, 0.66, CI95%: 0.44–0.99).41 Similar OS data were reported in the primary analysis of ARIEL3 trial in the intention to treat population (ITT), the mOS in patients treated with rucaparib was 36 months versus 43.2 months in the placebo group (HR 0.995, 95% CI: 0.809–1.223) despite the significant improvement in PFS.33 There could be several reasons for the lack of a clear OS benefit, including the fact that OS was a secondary end-point in many of the trials and these trials were underpowered for this endpoint, the high crossover rate between arms and the recent advances in the treatment of OC that have increased the mOS in these patients.

Nevertheless, the mature OS data from the first-line maintenance setting are positive. A clear OS benefit was reported after 7 years of follow-up in the SOLO1 trial, which had a median follow-up time of 88 months, with an OS HR of 0.55 in the olaparib maintenance group versus placebo and a substantial proportion of the olaparib patients not receiving subsequent lines of chemotherapy compared with placebo patients.20 Again, the PAOLA 1 5-year OS data clearly showed a benefit in favor of the combination of olaparib plus bevacizumab (OS at 5 years 65.5% versus 48.4%, HR: 0.62, 95% CI; 0.45–0.85) in the HRD patient cohort.22

There are few trials of PARPi in combination with immune-checkpoint inhibitors (ICIs), including anti-PD-1 and antiPD-L1 antibodies, in advanced OC. The clinical efficacy of ICIs in OC is limited and is considered a post-treatment option. However, emerging preclinical evidence suggests that PARPi have immunomodulatory effects, being able to activate the cGAS/STING pathway, to upregulate PD-L1 expression through the ATM/ATR/chk1 pathway, and to induce INF-1 release and expression of the chemokines CCL5 and CXCL10. Finally, PARPi treatment increases genomic instability, potentially leading to a higher tumor mutation burden and immune responsiveness.42 Given this background, the combination of PARPi and ICI may be a viable approach. Olaparib and durvalumab (an anti-PD-L1) combination in platinum-sensitive recurrent gBRCAmut OC patients showed a good safety profile and promising efficacy. The results of this Phase II trial have been recently published43 with an ORR was 92.2% and more than 40% patients presenting a complete response (CR). Based on these results, the DUO-O phase II trial was activated with the aim to compare in newly diagnosed OC patients the following schedules: Arm a) chemotherapy + bevacizumab + placebo followed by bevacizumab + placebo maintenance treatment; Arm b) chemotherapy + bevacizumab + durvalumab followed by bevacizumab + durvalumab + placebo maintenance treatment; or arm c) chemotherapy + bevacizumab + durvalumab followed by bevacizumab + durvalumab + olaparib maintenance treatment. Enrollment began in January 2019, and the updated final PFS, interim OS and updated safety were recent.44 A clinically meaningful PFS was observed for Arm 3 versus Arm 1 in the non-tBRCAm HRD population: HR 0.46 (95% CI: 0.33–0.65), with a mPFS of 45.1 versus 23.3 months, and a PFS rate at 24 months of 72.9% versus 46.5%, respectively. In the non-tBRCAm ITT population for Arm 3 versus Arm 1 an HR 0.61 (95% CI: 0.51–0.73), with mPFS of 25.1 versus 19.3 months, and PFS rate at 24 months of 53.0% versus 33.2%, respectively, have been found. The mPFS of 45.1 months observed in patients with HRD-positive disease treated with the triplet (Arm 3) is the longest ever reported in this setting. Although the interim OS analysis was not statistically significant in the ITT population, a favorable trend in overall survival was also observed in the BRCA wild-type, HRD-positive population. In the HRD-negative population, Arm 3 demonstrated an improvement in mPFS with a HR of 0.68 compared with Arm 1, even if the interim OS analysis did not show a significant difference between the treatment arms.44

The TOPACIO/KEYNOTE-162, a phase III study, evaluated the combination of niraparib and pembrolizumab in relapsed platinum-resistant OC. Among the 62 patients treated, the ORR was 18% (90% CI, 11–29%), with a disease control rate of 65% (90% CI, 54–75%). Interestingly, higher than expected responses were observed in patients without tumor BRCA mutations or non-HRD cancers.45 Many other studies are ongoing (recently reviewed in42), and data on efficacy are awaited.

The unraveling of the molecular characteristic of high-grade serous OC (HRD and its synthetic interaction with inhibition of PARPi greatly foster their clinical development with very positive results and a change in the therapeutic management of OC. The positive data from SOLO-1, PRIMA, and ATHENA trial have led to the approval of olaparib as for first-line maintenance treatment of advanced OC with BRCA mutated OC and niraparib regardless of HRD status. Recently, the PAOLA-1 data granted the approval of olaparib in combination with bevacizumab as first-line maintenance therapy in HRD OC; in addition, the long-term follow-up results are awaited from the DUO-O study to possibly include ICI in combination with PARPi and bevacizumab.

Clinical Results in Breast Cancer

The unraveling of the molecular characteristics of breast cancer (BC) has made it possible to define that nearly 10% of this type of tumor carries mutations in DNA repair genes (mainly BRCA1 and 2 genes) with the acquisition of an HDR phenotype. These tumors tend to be triple-negative and of high-grade and are generally more aggressive than the sporadic BCs.46 As HRD, they could benefit from a PARPi therapy. Indeed, two PARPi have been approved in BC: olaparib and talazoparib (Table 1). The specific indications were granted based on the results obtained in the OlympAD and EMBRACA trials47,48 (Table 3).

Table 3 Summary of the OlympiA, OlympiAD, EMBRACA, and BROCADE3 Phase III Trials Assessing the Use of PARPi in Patients with Breast Cancer

The phase III OlympAD trial, compared olaparib monotherapy with investigator’s choice (capecitabine, eribulin, or vinorelbine- standard) therapy in metastatic BC patients with a gBRCA mutation and human epidermal growth factor receptor type 2 (HER2)-negative, who had received no more than two prior chemotherapy regimens.50 The mPFS was 2.8 months longer in patients treated with olaparib than in patients treated with standard therapy; in addition, the risk of disease progression or death was 42% lower with olaparib monotherapy than with standard therapy. The final survival data did not show a statistically significant improvement in OS with olaparib compared to standard therapy; however, a significant OS benefit was observed in patients who had not received chemotherapy for metastatic disease.47 In an extended further analysis, the 3-year survival was 27.9% for olaparib versus 21.2% for standard of care, 8.8% of patients received olaparib treatment for ≥3 years versus none with chemotherapy treated patients.53 Following these results, a phase III trial patients with (HER2)-negative early breast cancer with BRCA1 or BRCA2 germline pathogenic or likely pathogenic variants and high-risk clinicopathologic factors who had received local treatment and neoadjuvant or adjuvant chemotherapy were randomized to receive olaparib or placebo for 1 year.48 At 3-year follow-up, invasive disease-free survival was 85.9% in the olaparib group versus 77.1% in the placebo group (HR 0.58; 99.5% CI; 0.41 to 0.82; p<0.001) and the distant disease-free survival was 87.5% versus 80.4%, respectively (HR 0.57; 99.5% CI; 0.39 to 0.83; p<0.001).

Similar results were observed in the EMBRACA trial in which patients with gBRCA1/2-mutated HER2-negative advanced BC patients were randomized to receive talazoparib or physician’s choice of chemotherapy51 The EMBRACA trial randomized 431 patients to receive talazoparib versus single-agent chemotherapy of physician’s choice and is the largest PARP monotherapy trial to date in this setting. Significant increase in PFS was observed in talazoparib treated patients versus chemotherapy treated patients: HR: 0.542 (95% CI: 0.413–0.711; p < 0.0001) with mPFS of 8.6 months versus 5.6 months. The results from the patient-reported outcomes (PRO) favored talazoparib, with significant overall improvement and significant delay in time to clinically meaningful deterioration of the global health status/quality of life (GHS/QoL) and breast symptom scales.52,54 Safety profile was similar to other PARPi treatment and could be managed by supportive care and dose modification. However, the final results of the OS,51 evaluated as a key secondary endpoint in this trial, did not show any statistically significant difference between arms, the HR was 0.848 (95% CI 0.670–1.073; p = 0.17); it was suggested that subsequent treatments the patients have undergone may have impacted results. Overall, these data supported the incorporation of talazoparib in clinical practice as a treatment option for patients with advanced BC with a gBRCA1/2 mutation. Clinical trials evaluating the role of rucaparib and niraparib in triple negative BC are ongoing, and results are awaited.55

All these data have suggested that patients with high‐risk, HER2‐negative early BC harboring gBRCA1/2 mutations can receive olaparib in the adjuvant setting after neoadjuvant or adjuvant chemotherapy. In addition, patients with HER-negative, gBRCA1/2 mutation locally advances or metastatic BC can receive talazoparib or olaparib in the metastatic setting after prior exposure to chemotherapy.

PARPi Inhibitors in Tumors Different from Breast and Ovarian Cancer

PARPi have been shown to have activity in other solid tumors besides breast and ovarian cancer, particularly in prostate cancer and pancreatic cancer where their use is approved as reported in Table 4.

Table 4 PARP Inhibitors Approved by FDA and EMA in Prostate and Pancreatic Cancer

Prostate Cancer

Prostate cancer (PC) is the second most commonly diagnosed cancer and the fifth leading cause of cancer death in men in Western countries.56 While early-stage PC is generally responsive to antiandrogen therapy, advanced-stage and metastatic PC (metastatic Castration-Resistant Prostate Cancer, mCRPC) is a very heterogeneous disease with poor response to therapy57 and with a very dismal prognosis (median OS of about 2 years).58 The molecular characterization of PC revealed that approximately 10% of localized tumors and up to 30% of mCRCP show defects in DNA damage response (DDR) for mutations in BRCA1, BRCA2, CDK12, ATM, and CHK2 genes leading to their loss of function and impairment of HR.59,60 For this reason, all patients with PC should undergo germline testing, and somatic testing is recommended in patients with recurrent and metastatic disease.58 The presence of HR defects has opened up the way for the use of PARPi, particularly in mCRPC, changing the standard of care for these patients.61

PARPi were first evaluated in phase II trials as monotherapy in mCRPC.57 In all these studies, patients with mCRPC progressing after taxane or after taxane and androgen receptor signaling inhibitors (ARSi) with DDR alterations, directly or indirectly related to HR, were treated with olaparib (TORAP-B),62 rucaparib (TRITON2),63 talazoparib (TALAPRO-1)64 and niraparib (GALAHAD).65 The objective response rate (ORR) in patients with DDR alterations was the primary endpoint in all the phases II except for the GALAHAD trials, where the primary endpoint was the ORR in patients with biallelic inactivation of BRCA1/2. The ORRs were, respectively, 39.1 and 54.3% (for the 300 and 400mg olaparib twice-daily doses), 43%, 29.8% and 34.2%. Interestingly, all the studies reported higher response rates in the BRCA1/2 cohorts of patients than in other cohorts of patients with DDR genomic alterations.

These exciting results prompted the implementation of phase III clinical trials: the PROfound60 and TRITON366 trials, the positive results of which led to the approval of olaparib and rucaparib in mCRPC patients with alteration in DDR gene alterations (Table 5).

The TRIOTON 3 is a phase III trial that enrolled mCRPC with BRCA1/2 or ATM alterations that progressed after treatment with a second-generation androgen receptor pathway inhibitor (APRi). A total of 4855 patients were screened and 270 were assigned to the rucaparib arm (600mg twice daily) and 135 to the physician’s choice of docetaxel or a second-generation ARPi (abiraterone acetate or enzalutamide) arm. Patients with BRCA1 alterations comprised 75% of both groups. In the control arm, 56% of the patients received docetaxel. The primary endpoint was the median duration of imaging-based progression-free survival, and at 62 months follow-up, it was significantly longer in the rucaparib group than in the control group both in the BRCA subgroup (median, 11.2 months vs 6.4 months, HR: 0.50, 95% CI: 0.36–0.69, p < 0.001) and in the intention to-treat group (median, 10.2 months and 6.4 months, HR: 0.61, 95% CI: 0.47–0.80, p < 0.001). No difference was observed in the ATM mutated patients with a median duration of imaging-based progression-free survival of 8.1 months (95% CI, 5.5 to 8.3) in the rucaparib group and of 6.8 months (95% CI, 4.0 to 10.4) in the control group (HR 0.95; 95% CI, 0.59–1.52). OS data are awaited.

The PROfound study is a randomized, open-label, biomarker-driven phase III trial in mCRPC that has progressed while receiving enzalutamide or abiraterone to evaluate the efficacy of olaparib. Only patients with alterations in prespecified genes directly or indirectly associated with HR were enrolled in two cohorts: cohort A (245 patients) with alterations in BRCA1, BRCA 2 and ATM and cohort B (142 patients) with alterations on other prespecified DDR genes. Primary end point was radiological PFS (rPFS) of olaparib compared to androgen receptor signaling inhibitors (ARSi) in the BRCA cohort and in the overall population. Olaparib treatment significantly improved rPFS in cohort A (median,7.4 months vs 3.6 months; HR 0.39 95% CI 0.34; 95% CI, 0.25 to 0.47; p<0.001) and in the overall population (median, 5.8 versus 3.5 months; HR, 0.49; 95% CI, 0.38 to 0.63; p<0.001). In addition, in cohort A the OS interim analysis shows a benefit in the olaparib-treated arm that did not reach a statistical significance (19.1 versus 14.7 months, HR: 0.69, 95% CI: 0.50–0.97, p=0.02); these latter data could be inferred from the 80% of crossover to olaparib arm. The main criticism of these data was the fact that the treatment in the control arm was ARSi after prior ARSi therapy, which has been reported to have limited efficacy in mCRPC due to cross-resistance mechanisms,67,68 and a taxane-based chemotherapy would probably have been a better control arm.

Preclinical evidence strongly suggests a synergistic effect between PARPi and ARSi. Indeed, the androgen receptor (AR) has been shown to enhance the expression of DNA damage repair (DDR) genes69 and PARP1 supports the AR-X transcriptional activity,70,71 suggesting that androgen receptor blockade could induce a BRCAness phenotype. With this in mind, the efficacy and tolerability of olaparib and abiraterone were evaluated in 142 mCRPC patients randomized to receive olaparib/abiraterone and placebo/abiraterone.72 The rPFS was 13.8 in the olaparib group versus 8.2 in the placebo group (HR 065, 95% CI0.11–0.97, p=0.034). Based on these results, the combination of PARPi and ARSi was evaluated as a first-line therapy in mCRPC in three randomized, double-blind, placebo-controlled prospective Phase 3 trials: PROpel,73 MAGNITUDE,74 and TALAPRO-275 (Table 5).

Table 5 Summary of the Phase III Trials Assessing the Role of PAPRi in Monotherapy or in Combination in mCRPC

The PROpel study73 enrolled 796 mCRPC patients, regardless of HRR gene mutation status, to receive abiraterone/olaparib versus abiraterone/placebo in the first-line setting. The primary endpoint (rPFS) was met with a median rPFS of 24.8 months observed in the combination group versus 16.6 months in the placebo-combination arm (HR: 0.66, 95% CI: 0.54–0.81, p < 0.001). In patients with the HRR gene mutations, rPSF was not yet reached in the olaparib group compared to 13.9 months in the control combo group. However, at 36.5 months of follow-up, no difference was observed between the two treatment arms in the prespecified analysis of mOS (42.1 vs 34.7 months, HR: 0.81, 95% CI: 0.67–1.00, p = 0.054). However, in a post-hoc exploratory subgroup analysis, the combination of abiraterone/olaparib reduced the risk of death by 71% of in BRCA1/2 patients (mOS: not reached vs 23.0 months, HR: 0.29, 95% CI: 0.14–0.56) and by 34% in patients with HRR gene mutations (mOS: not reached vs 28.5 months, HR: 0.66, 95% CI: 0.45–0.95).

In the MAGNITUDE74 trial evaluated the combination of niraparib/abiraterone versus placebo/abiraterone as the first-line treatment of mCRPC with a defined HRR status. Patients with HRR alteration (423 patients) and without HRR alteration (247 patients) were randomized 1:1 to receive niraparib or placebo. rPFS, assessed by central review, was the primary endpoint and was assessed first in the BRCA1/2 mutated cohort and then in the HRR deficient cohort. Median rPFS in the BRCA1/2 subgroup was significantly longer in the niraparib combination arm compared to the placebo control arm compared with the placebo group (16.6 vs 10.9 months; HR: 0.53; 95% CI, 0.36 to 0.79; p=0.001). When considering the entire HRR-deficient cohort, again rPFS was significantly longer in the niraparib than in with the placebo group (16.5 vs 13.7 months; HR, 0.73; 95% CI, 0.56 to 0.96; p=0.022). The OS data were immature in this first initial analysis.

The TALAPRO-275 trial compared the combination of talazoparib/enzalutamide (402 patients) was compared to placebo/enzalutamide (403 patients) as the first-line treatment of mCRPC (regardless of HRR gene alterations). Patients were stratified at randomization by HRR gene alteration status and prior treatment with life-prolonging therapy. A statistically significant improvement in rPFS was observed in the talazoparib and enzalutamide at the planned primary analysis, with a median rPFS not reached in the PARPi combination versus 21.9 months in the placebo group (HR: 0.63, 95% CI: 0.51–0.78; p < 0.0001). In addition, subgroup analysis using HRR gene alteration status showed an HR for rPFS of 0.46 (95% CI 0.30–0.70; p=0.0003) in patients with HRD and of 0.70 (0.54–0.89; p=0.0039) in patients with a status of non-deficient or unknown in favor of talazoparib/enzalutamide combination versus placebo.

In all these studies, the combination treatments were well tolerated with a manageable safety profile consistent with the reported side effects of the individual drugs.

Given these very positive results, evaluating the synergistic effect of ARSi and PARPi in early-stage disease will be of great value, and indeed clinical trials are ongoing to evaluate evaluating the combination of niraparib/abiraterone (ARSiAMPLITUDE)80 and talazoparib/enzalutamide (TALAPRO-3)81 in metastatic hormone-sensitive PC patients with HRR alterations to potentially prolong the duration of hormone sensitivity and modify the disease course.

PARPi are also being studied in combination with other drugs in metastatic PC.

The combination of PARPi with immune checkpoint inhibitors (ICI) has been proposed based on the genomic instability caused by the HR and DDR defects, which may trigger neoantigen production and T-cell activation.82 However, the phase III Keylink-010 trial83 showed no significant improvement in outcomes with the combination of pembrolizumab and olaparib in biomarker-unselected, heavily pretreated mCRPC, suggesting that further patient selection is critical for this approach. The combination of PARPi with radioligand therapies, such as lutetium-177 (177Lu)-PSMA-617 and radium-223 dichloride, shows potential due to the synergy between radiation-induced DNA damage and PARPi. Early studies have tested these combinations with ARSi with positive safety profiles, suggesting that they could be further explored also in combination with PARPi.84–86

Other combinations of PARPi with targeted therapies, such as AKT inhibitors and VEGFR inhibitors, are also being studied.87,88 While some studies have shown good safety, others have shown improved PFS but with increased adverse events, highlighting the need for further research to identify biomarkers for treatment tailoring.

Summing up, enzalutamide/talazoparib combination is a valid first-line option in mCRPC patients with HRR mutations; abiraterone/olaparib and abiraterone/niraparib are available in patients with BRCA1/2 mutations. In patients with advanced relapses, disease and mutations in any of the prespecified HRR gene olaparib can be used, while rucaparib is available for patients with BRCA1/2 alterations after therapy with ARPi and docetaxel.

Pancreatic Cancer

Pancreatic cancer, and in particular, pancreatic ductal adenocarcinoma (PDAC), remains one of the deadliest tumor with a 5-year survival rate of 10% in the metastatic setting. In the recent years, with the increasing number of molecular characterizations, a significant percentage of PDAC tumors have shown HRD defects, thus offering new potential therapeutic patients in selected population. It is estimated that approximately 15–20% of PDAC are HRD (mostly due to mutations in BRCA1/2 and PALB2 genes).89,90 These patients represent a subset for whom the use of PARPi may help improve their outcomes. Back in 2019, olaparib was approved in PDAC following the results of Phase III trials that evaluated the use of this PARPi versus placebo in PDAC patients who had a response or stabilization to prior platinum therapy. The primary endpoint (PFS) was met with a PFS of 7.4 months in the olaparib arm vs 3.8 months in the placebo arm (resulting in a statistically significant HR of 0.53).91,92 The overall impact of the study was somewhat limited by the fact that the comparison was to placebo and most importantly because the secondary endpoint of the study (OS) did not show an advantage in the olaparib group.

The efficacy of rucaparib in PDAC in two studies has been evaluated. In one study, the drug was tested in patients with defined mutations in BRCA1/2 and PALB2 genes as maintenance after platinum, while in the second study, the prior response to platinum was not mandatory.93,94 The first maintenance study after response to platinum-based therapy gave positive results in terms of ORR (42%) PFS and OS (13 and 23.5 months, respectively).93 Interestingly, of the 42 patients evaluable in the phase II trial, 3 had complete response and 12 had a partial response. Enrollment in the second study was stopped due to a low response rate in the first patients.94 In this study, prior response to platinum was not mandatory, and patients with platinum-resistant tumor did not achieve an objective response. This suggested that platinum sensitivity may be a useful biomarker for PARPI efficacy.

Interestingly, trials are testing PARPi in the early setting of pancreatic cancer; in particular, the APOLLO trial is comparing olaparib versus placebo in patients with no evidence of recurrent disease following perioperative chemotherapy in patients with mutations in the BRCA1/2 or PALB2 genes.95

Finally, new PARPi such as veliparib have been tested in phase II trials with no positive results.96 In this Phase II, the safety and efficacy of velarapib were evaluated in BRCAm PDAC patients, with progressive disease with 1–2 prior chemotherapy regimens; no confirmed radiographic responses were observed, although a stable disease >8 weeks was seen in one-fourth of patients. One possible explanation for the lack of positive results could be that, as mentioned above, several enrolled patients were resistant to platinum.

Finally, several trials are testing the combination of PARPi and immune checkpoint inhibitors based on the positive preclinical results.97 Hopefully, this will also translate into benefits also at the clinical level.

In conclusion, patients with BRCA1/2 mutations, platinum sensitive (not progression after ≥ 16 weeks), and metastatic pancreatic cancer can receive olaparib as maintenance.

Other Tumors

PARPi have been and are currently being tested in tumors other than the four in which these drugs are approved.

In NSCLC, a systemic review analyzing 12 trials in which PARPi were used mostly in combination with chemotherapy found a slight improvement in OS (with an HR of 0.9 (0.83–0.97)) but a nonsignificant difference in PFS.98 A randomized phase II trial comparing olaparib with placebo as maintenance in patients responding to platinum therapy (PIPSeN trial) was stopped due to lack of improvement.99 Given the evidence that HR defects predict the response to immune checkpoint inhibitors in NSCLC patients,100 it is likely that there could be a potential benefit, in this subset of patients from the use of a combination of PARPi and ICI.

In HCC, there is evidence of a potential sensitization by the use of PARPi and shown in a patient with HCC presenting a mutation in the FANC A gene (belonging to HR).101 Recent results from a tumor-agnostic phase II trial showed varying degrees of response in different tumors with germline of somatic mutations in genes belonging to HR.102

Acquired or Intrinsic Resistance to PARPi

Although the introduction of PARPi has significantly improved patient survival, a significant percentage of HRD-patients do not respond to these drugs. In addition to the intrinsic resistance, as is reported for almost all drugs used in oncology, drug resistance also occurs in patients who initially respond to treatment with PARPi. Several studies have been conducted to elucidate the mechanisms of resistance and possible ways to overcome it. Based on preclinical studies, the mechanisms of resistance reported to date include some that are generally observable mechanisms (such as increased expression of P-glycoprotein mediating drug efflux from cells or activation of epithelial-to-mesenchymal transition-EMT) some that are related to alteration of the target (such as reduced levels of DNA trapped PARP1, or reduced expression of the poly-ADP-ribose glycohydrolase) and some that specifically related to the mechanism of action and/or of synthetic lethality. The latter are particularly relevant as they are among those seen clinically.

The ABCB1 (ATP-binding cassette superfamily B member 1) gene encodes a P-glycoprotein (P-gp) that belongs to a family of protein pumps (ABC proteins) found in the cells’ membrane. The ABC protein acts by removing drugs from the cells, thereby controlling their intracellular levels. It is well known that ABC protein expression is associated with resistance to those drugs that are substrates of the glycoprotein. Doxorubicin and etoposide are two examples of drugs whose intracellular levels are regulated by ABC protein. Olaparib is a substrate of P-gp, and it has been shown that repeated treatment with these drugs induces an upregulation of the ABCB1 gene conferring resistance to the drug. To confirm the role of this mechanism in the resistance to olaparib, treatments with ABC inhibitors are able to restore the sensitivity to olaparib in preclinical models.103–105

EMT is a well-known mechanism by which epithelial cells acquire a more malignant and invasive phenotype that is associated with metastasis and drug resistance.106–109 Using transgenic mouse models and patient-derived xenografts (PDX), three independent groups have found that PARPi resistance can also be associated with an enhanced mesenchymal phenotype,110–112 although other mechanisms have been found to be associated with resistance in these models (including increased drug efflux).

Another mechanism of resistance observed for several DNA damaging agents (cisplatin, etoposide, trabectedin, topotecan) is the loss of expression of the DNA/RNA helicase SLFN11. This helicase acts normally or when overexpressed by preventing tumor replication. In fact, SLFN11 is recruited early to stressed replication forks, thereby inducing replication arrest and controlling tumor growth.113 In many cancer cells, SLFN11 expression is low and, when treated with DNA damaging agents SLFN11, is further inactivated mostly due to hypermethylation of its promoter. This has been associated with resistance to many DNA damaging agents and more recently for PARPi.110,114–117 However, it should be noted, however, that, in a retrospective study of ovarian cancer patients treated with olaparib, there was a positive trend between high SLFN11 expression and better PFS, although this was not observed when overall survival was considered.118 Olaparib and other PARPi are able to exert anticancer activity not only because of their ability to inhibit the enzymatic activity of PARP but also because they trap PARP into DNA. In fact, the trapping activity is considered to be a relevant factor in the overall activity of the inhibitors.6,119 There is evidence that the basal levels of PARP correlate positively with the response to PARPi.120,121 Although this has not been clinically demonstrated, it is important to note that the different PARPi clinically available have a different ability and potency to trap PARP, and therefore, their use could potentially be selected based on PARP levels.

The most important and clinically relevant mechanisms of resistance to date are those associated with restoration of BRCA function and, more generally, to restoration of HR.

Indeed, reversion mutations in the BRCA1/2 genes have been found in different patients with different tumors that were in progression after PARPi.122,123 The importance of mutation reversion is further corroborated by the evidence that in long responders to PARPi there is an enrichment of BRCA mutations that cannot be reversed by secondary mutations.124 Additional mutations have recently been found in patients-derived xenografts derived from PARPi resistant patients and confirmed clinically in a cohort of patients with ovarian cancer.125 Interestingly, splice site mutations are able to remove the entire exon (in this case exon 11) carrying the mutation, thus resulting in the production of a truncated protein with functional (albeit reduced) activity.

Methylation of the BRCA promoter resulting in loss of protein expression is another mechanism leading to HR deficiency and PARPi activity.126,127 It has been reported that promoter demethylation in epigenetically silenced BRCA1 tumors represents an additional mechanism of resistance to PARPi by restoring HR function.128

Finally, the mechanisms proposed for mutation reversion and promoter demethylation in the BRCA genes, have also been found in other genes acting in the HR repair such as RAD51C, RAD51D and PALB2.129–134

Conclusions and Future Perspectives

The introduction of PARPi into clinical practice has significantly and positively changed the scenario for patients with defects in HR. This is true not only for breast and ovarian cancer that, which were the first tumors in which these drugs were tested but also for other solid tumors such as prostate and pancreatic cancer in which PARPi are being tested on an ongoing basis. In recent years, other clinical trials have been implemented assessing these agents in different oncologic settings, including small cell lung cancer and metastatic colon cancer. Their efficacy is associated with the presence of HRD (ie mutation in HHR genes, including BRCA1/2) and the response of a platinum-based therapy, even if hints of activity have also been observed in HRP tumors. The increasing availability of tests to determine the status of HR beyond BRCA1/2 and other known gene mutations, together with the introduction of functional tests that can easily identify defective tumors, will certainly increase the number of patients (and likely tumors) that could potentially benefit from the use of PARPi.

Given the positive results of PARPi in different settings, they have been introduced earlier and earlier in the management of cancer patients and still open is the optimal duration of adjuvant PARPi. While in the OC setting PARPi have been used for up to 2 years, in the OlympiA trial olaparib has been administered for one year and this could be different for the different PARPi and in early stage cancers (ie gBRCAm carriers with early-stage breast cancer). Another important issue relates to the safety of PARPi, especially the long-term effects, including their impact of the quality of life in cancer patients considering the long treatment duration. Hematological toxicities and higher risk of developing myelodysplastic syndrome and acute myeloid leukemia are among the reported adverse effects of these agents. As most of these effects appear to be due to the inhibition of PARP2, as new PARPi more selective and less toxic are being studied at the preclinical and clinical levels. Some are already in phase III trial (iniparib, veliparib, senaparib), some in phase II (INO-1001, nesuparib, saruparib, stenoparib, atamparib, vendaparib, CEP-9722, BGP-15, TSL-1502SC10914, HWH340) and several others in early clinical trials.

Further research should be focused not only on a better patient stratification using more biomarkers as outlined above but also to explore through biologically driven approach to implement new combination of PARPi and both cytotoxic and non-cytotoxic agents, including immunotherapy.

Lastly, as for other anticancer agents, also for this class of agents, resistance to therapy has been observed in the clinic. There is the strong need for preclinical models that recapitulate human tumors in which to evaluate the resistance mechanism and ways to overcome it. Resistance to treatment is still one of the reasons for treatment failure despite an initial response. Although some mechanisms have been highlighted (such as reversion mutations in BRCA genes), additional studies on well-defined models are urgently needed to unleash the full potential of this class of drugs to induce durable responses and thus a strong benefit for patients.

Acknowledgments

Part of this work has been done under the institutional “Ricerca Corrente” granted by the Italian Ministry of Health; Ministry of University and Research-MUR under PNRR M4C2I1.3 heal Italia project PE00000019 CUP B43D22000710006 of Istituto di Ricerche Farmacologiche Mario Negri IRCCS – P.I. Giovanna Damia.

Disclosure

The authors report no conflicts of interest in this work.

References

1. Lord CJ, Ashworth A. PARP inhibitors: synthetic lethality in the clinic. Science. 2017;355(6330):1152–1158. doi:10.1126/science.aam7344

2. Konstantinopoulos PA, Ceccaldi R, Shapiro GI, D’Andrea AD. Homologous recombination deficiency: exploiting the fundamental vulnerability of ovarian cancer. Cancer Discov. 2015;5(11):1137–1154. doi:10.1158/2159-8290.CD-15-0714

3. Ceccaldi R, Rondinelli B, D’Andrea AD. Repair pathway choices and consequences at the double-strand break. Trends Cell Biol. 2016;26(1):52–64. doi:10.1016/j.tcb.2015.07.009

4. Bryant HE, Schultz N, Thomas HD, et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature. 2005;434(7035):913–917. doi:10.1038/nature03443

5. Farmer H, McCabe N, Lord CJ, et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature. 2005;434(7035):917–921. doi:10.1038/nature03445

6. Murai J, Yin N HS, Das BB, et al. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res. 2012;72(21):5588–5599. doi:10.1158/0008-5472.CAN-12-2753

7. D’Andrea AD. Mechanisms of PARP inhibitor sensitivity and resistance. DNA Repair. 2018;71:172–176. doi:10.1016/j.dnarep.2018.08.021

8. Cong K, Cantor SB. Exploiting replication gaps for cancer therapy. Mol Cell. 2022;82(13):2363–2369. doi:10.1016/j.molcel.2022.04.023

9. King MC, Marks JH, Mandell JB, New York Breast Cancer Study Group. Breast and ovarian cancer risks due to inherited mutations in BRCA1 and BRCA2. Science. 2003;302(5645):643–646. doi:10.1126/science.1088759

10. Klatte DCF, Wallace MB, Löhr M, Bruno MJ, van Leerdam ME. Hereditary pancreatic cancer. Best Pract Res Clin Gastroenterol. 2022;58-59:101783. doi:10.1016/j.bpg.2021.101783

11. Szentmartoni G, Mühl D, Csanda R, Szasz AM, Herold Z, Dank M. Predictive value and therapeutic significance of somatic BRCA mutation in solid tumors. Biomedicines. 2024;12(3):593. doi:10.3390/biomedicines12030593

12. Venkitaraman AR. Cancer susceptibility and the functions of BRCA1 and BRCA2. Cell. 2002;108(2):171–182. doi:10.1016/S0092-8674(02)00615-3

13. Li X, Zou L. BRCAness, DNA gaps, and gain and loss of PARP inhibitor-induced synthetic lethality. J Clin Invest. 2024;134(14):e181062. doi:10.1172/JCI181062

14. Vanacker H, Harter P, Labidi-Galy SI, et al. PARP-inhibitors in epithelial ovarian cancer: actual positioning and future expectations. Cancer Treat Rev. 2021;99:102255. doi:10.1016/j.ctrv.2021.102255

15. Guffanti F, Mengoli I, Damia G. Current HRD assays in ovarian cancer: differences, pitfalls, limitations, and novel approaches. Front Oncol. 2024;14:1405361. doi:10.3389/fonc.2024.1405361

16. Goldlust IS, Guidice E, Lee JM. PARP inhibitors in ovarian cancer. Semin Oncol. 2024;51(1–2):45–57. doi:10.1053/j.seminoncol.2024.01.001

17. Moore K, Colombo N, Scambia G, et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 2018;379(26):2495–2505. doi:10.1056/NEJMoa1810858

18. González-Martín A, Pothuri B, Vergote I, et al. Niraparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 2019;381(25):2391–2402. doi:10.1056/NEJMoa1910962

19. Monk BJ, Parkinson C, Lim MC, et al. A randomized, phase III trial to evaluate rucaparib monotherapy as maintenance treatment in patients with newly diagnosed ovarian cancer (ATHENA-MONO/GOG-3020/ENGOT-ov45). J Clin Oncol off J Am Soc Clin Oncol. 2022;40(34):3952–3964. doi:10.1200/JCO.22.01003

20. DiSilvestro P, Banerjee S, Colombo N, et al. Overall survival with maintenance olaparib at a 7-year follow-up in patients With newly diagnosed advanced ovarian cancer and a BRCA mutation: the SOLO1/GOG 3004 trial. Obstet Gynecol Surv. 2023;78:25–27. doi:10.1097/ogx.0000000000001120

21. Banerjee S, Moore KN, Colombo N, et al. Maintenance olaparib for patients with newly diagnosed advanced ovarian cancer and a BRCA mutation (SOLO1/GOG 3004): 5-year follow-up of a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol. 2021;22(12):1721–1731. doi:10.1016/S1470-2045(21)00531-3

22. Ray-Coquard I, Leary A, Pignata S, et al. Olaparib plus bevacizumab first-line maintenance in ovarian cancer: final overall survival results from the PAOLA-1/ENGOT-ov25 trial. Ann Oncol Off J Eur Soc Med Oncol. 2023;34(8):681–692. doi:10.1016/j.annonc.2023.05.005

23. González-Martín A, Desauw C, Heitz F, et al. Maintenance olaparib plus bevacizumab in patients with newly diagnosed advanced high-grade ovarian cancer: main analysis of second progression-free survival in the phase III PAOLA-1/ENGOT-ov25 trial. Eur J Cancer Oxf Engl 1990. 2022;174:221–231. doi:10.1016/j.ejca.2022.07.022

24. Ray-Coquard I, Pautier P, Pignata S, et al. Olaparib plus bevacizumab as first-line maintenance in ovarian cancer. N Engl J Med. 2019;381(25):2416–2428. doi:10.1056/NEJMoa1911361

25. González-Martín A, Pothuri B, Vergote I, et al. Progression-free survival and safety at 3.5 years of follow-up: results from the randomised phase 3 PRIMA/ENGOT-OV26/GOG-3012 trial of niraparib maintenance treatment in patients with newly diagnosed ovarian cancer. Eur J Cancer Oxf Engl 1990. 2023;189:112908. doi:10.1016/j.ejca.2023.04.024

26. Poveda A, Floquet A, Ledermann JA, et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a final analysis of a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2021;22(5):620–631. doi:10.1016/S1470-2045(21)00073-5

27. Pujade-Lauraine E, Ledermann JA, Selle F, et al. Olaparib tablets as maintenance therapy in patients with platinum-sensitive, relapsed ovarian cancer and a BRCA1/2 mutation (SOLO2/ENGOT-Ov21): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2017;18(9):1274–1284. doi:10.1016/S1470-2045(17)30469-2

28. Mirza MR, Monk BJ, Herrstedt J, et al. Niraparib maintenance therapy in platinum-sensitive, recurrent ovarian cancer. N Engl J Med. 2016;375(22):2154–2164. doi:10.1056/NEJMoa1611310

29. Coleman RL, Oza AM, Lorusso D, et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Lond Engl. 2017;390(10106):1949–1961. doi:10.1016/S0140-6736(17)32440-6

30. Coleman RL, Oza AM, Lorusso D, et al. 2022-RA-249-ESGO overall survival results from ariel3: a phase 3 randomised, double-blind study of rucaparib vs placebo following response to platinum-based chemotherapy for recurrent ovarian carcinoma. Ovarian Cancer. BMJ Publishing Group Ltd; 2022:A2261.A226. doi:10.1136/ijgc-2022-ESGO.488

31. Ledermann J, Harter P, Gourley C, et al. Olaparib maintenance therapy in patients with platinum-sensitive relapsed serous ovarian cancer: a preplanned retrospective analysis of outcomes by BRCA status in a randomised Phase 2 trial. Lancet Oncol. 2014;15(8):852–861. doi:10.1016/S1470-2045(14)70228-1

32. Pujade-Lauraine E, Ledermann J, Penson RT, et al. Treatment with olaparib monotherapy in the maintenance setting significantly improves progression-free survival in patients with platinum-sensitive relapsed ovarian cancer: results from the phase III SOLO2 study. Gynecol Oncol. 2017;145:219–220. doi:10.1016/j.ygyno.2017.03.505

33. Coleman RL, Oza A, Lorusso D, et al. O003/#557 overall survival results from ARIEL3: a phase 3 randomized, double-blind study of rucaparib vs placebo following response to platinum-based chemotherapy for recurrent ovarian carcinoma. Int J Gynecol Cancer. 2022;32(Suppl 3). doi:10.1136/ijgc-2022-igcs.5

34. Oza AM, Matulonis UA, Malander S, et al. Quality of life in patients with recurrent ovarian cancer treated with niraparib versus placebo (ENGOT-OV16/NOVA): results from a double-blind, phase 3, randomised controlled trial. Lancet Oncol. 2018;19(8):1117–1125. doi:10.1016/S1470-2045(18)30333-4

35. Penson R, Valencia RV, Colombo N, et al. Final overall survival results from SOLO3: phase III trial assessing olaparib monotherapy versus non-platinum chemotherapy in heavily pretreated patients with germline BRCA1 - and/or BRCA2-mutated platinum-sensitive relapsed ovarian cancer (026). Gynecol Oncol. 2022;166:S19–S20. doi:10.1016/S0090-8258(22)01244-6

36. Kristeleit R, Lisyanskaya A, Fedenko A, et al. Rucaparib versus standard-of-care chemotherapy in patients with relapsed ovarian cancer and a deleterious BRCA1 or BRCA2 mutation (ARIEL4): an international, open-label, randomised, phase 3 trial. Lancet Oncol. 2022;23(4):465–478. doi:10.1016/S1470-2045(22)00122-X

37. Chiappa M, Guffanti F, Bertoni F, Colombo I, Damia G. Overcoming PARPi resistance: preclinical and clinical evidence in ovarian cancer. Drug Resist Updat Rev Comment Antimicrob Anticancer Chemother. 2021;55:100744. doi:10.1016/j.drup.2021.100744

38. Vanderstichele A, Van Nieuwenhuysen E, Han S, et al. Randomized phase II CLIO study on olaparib monotherapy versus chemotherapy in platinum-resistant ovarian cancer. J Clin Oncol. 2019;37(15_suppl):5507. doi:10.1200/JCO.2019.37.15_suppl.5507

39. Moore KN, Secord AA, Geller MA, et al. Niraparib monotherapy for late-line treatment of ovarian cancer (QUADRA): a multicentre, open-label, single-arm, phase 2 trial. Lancet Oncol. 2019;20(5):636–648. doi:10.1016/S1470-2045(19)30029-4

40. Domchek SM, Aghajanian C, Shapira-Frommer R, et al. Efficacy and safety of olaparib monotherapy in germline BRCA1/2 mutation carriers with advanced ovarian cancer and three or more lines of prior therapy. Gynecol Oncol. 2016;140(2):199–203. doi:10.1016/j.ygyno.2015.12.020

41. Matulonis U, Herrstedt J, Oza A, et al. Long-term safety and secondary efficacy endpoints in the ENGOT-OV16/NOVA phase III trial of niraparib in recurrent ovarian cancer. Gynecol Oncol. 2021;162:S24–S25. doi:10.1016/S0090-8258(21)00693-4

42. Xiao F, Wang Z, Qiao L, et al. Application of PARP inhibitors combined with immune checkpoint inhibitors in ovarian cancer. J Transl Med. 2024;22(1):778. doi:10.1186/s12967-024-05583-z

43. Drew Y, Kim JW, Penson RT, et al. Olaparib plus durvalumab, with or without bevacizumab, as treatment in PARP inhibitor-naïve platinum-sensitive relapsed ovarian cancer: a phase II multi-cohort study. Clin Cancer Res Off J Am Assoc Cancer Res. 2024;30(1):50–62. doi:10.1158/1078-0432.CCR-23-2249

44. Harter P, Wimberger P, Okamoto A, et al. Durvalumab plus paclitaxel/carboplatin plus bevacizumab followed by durvalumab, bevacizumab plus olaparib maintenance among patients with newly-diagnosed advanced ovarian cancer without a tumor BRCA1/BRCA2 mutation: updated results from DUO-O/ENGOT-OV46/GOG-3025 Trial. Gynecol Oncol. 2024;190:S65–S66. doi:10.1016/j.ygyno.2024.07.096

45. Konstantinopoulos PA, Waggoner S, Vidal GA, et al. Single-arm phases 1 and 2 trial of niraparib in combination with pembrolizumab in patients with recurrent platinum-resistant ovarian carcinoma. JAMA Oncol. 2019;5(8):1141–1149. doi:10.1001/jamaoncol.2019.1048

46. Baretta Z, Mocellin S, Goldin E, Olopade OI, Huo D. Effect of BRCA germline mutations on breast cancer prognosis: a systematic review and meta-analysis. Medicine. 2016;95(40):e4975. doi:10.1097/MD.0000000000004975

47. Robson ME, Tung N, Conte P, et al. OlympiAD final overall survival and tolerability results: olaparib versus chemotherapy treatment of physician’s choice in patients with a germline BRCA mutation and HER2-negative metastatic breast cancer. Ann Oncol Off J Eur Soc Med Oncol. 2019;30(4):558–566. doi:10.1093/annonc/mdz012

48. Tutt ANJ, Garber JE, Kaufman B, et al. Adjuvant olaparib for patients with BRCA1- or BRCA2-mutated breast cancer. N Engl J Med. 2021;384(25):2394–2405. doi:10.1056/NEJMoa2105215

49. Geyer CE, Garber JE, Gelber RD, et al. Overall survival in the OlympiA phase III trial of adjuvant olaparib in patients with germline pathogenic variants in BRCA1/2 and high-risk, early breast cancer. Ann Oncol Off J Eur Soc Med Oncol. 2022;33(12):1250–1268. doi:10.1016/j.annonc.2022.09.159

50. Robson M, Im SA, Senkus E, et al. Olaparib for metastatic breast cancer in patients with a germline BRCA mutation. N Engl J Med. 2017;377(6):523–533. doi:10.1056/NEJMoa1706450

51. Litton JK, Hurvitz SA, Mina LA, et al. Talazoparib versus chemotherapy in patients with germline BRCA1/2-mutated HER2-negative advanced breast cancer: final overall survival results from the EMBRACA trial. Ann Oncol Off J Eur Soc Med Oncol. 2020;31(11):1526–1535. doi:10.1016/j.annonc.2020.08.2098

52. Litton JK, Rugo HS, Ettl J, et al. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N Engl J Med. 2018;379(8):753–763. doi:10.1056/NEJMoa1802905

53. Robson ME, Im SA, Senkus E, et al. OlympiAD extended follow-up for overall survival and safety: olaparib versus chemotherapy treatment of physician’s choice in patients with a germline BRCA mutation and HER2-negative metastatic breast cancer. Eur J Cancer Oxf Engl 1990. 2023;184:39–47. doi:10.1016/j.ejca.2023.01.031

54. Ettl J, Quek RGW, Lee KH, et al. Quality of life with talazoparib versus physician’s choice of chemotherapy in patients with advanced breast cancer and germline BRCA1/2 mutation: patient-reported outcomes from the EMBRACA phase III trial. Ann Oncol Off J Eur Soc Med Oncol. 2018;29(9):1939–1947. doi:10.1093/annonc/mdy257

55. Daly GR, AlRawashdeh MM, McGrath J, et al. PARP inhibitors in breast cancer: a short communication. Curr Oncol Rep. 2024;26(2):103–113. doi:10.1007/s11912-023-01488-0

56. Bray F, Laversanne M, Sung H, et al. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 2024;74(3):229–263. doi:10.3322/caac.21834

57. de la Maza MD F, Pérez Gracia JL, Miñana B, Castro E. PARP inhibitors alone or in combination for prostate cancer. Ther Adv Urol. 2024;16:17562872241272928. doi:10.1177/17562872241272929

58. Armstrong AJ, Taylor A, Haffner MC, et al. Germline and somatic testing for homologous repair deficiency in patients with prostate cancer (part 1 of 2). Prostate Cancer Prostatic Dis. 2024;27:1–10. doi:10.1038/s41391-024-00901-4

59. Armenia J, Wankowicz SAM, Liu D, et al. The long tail of oncogenic drivers in prostate cancer. Nat Genet. 2018;50(5):645–651. doi:10.1038/s41588-018-0078-z

60. de Bono J, Mateo J, Fizazi K, et al. Olaparib for metastatic castration-resistant prostate cancer. N Engl J Med. 2020;382(22):2091–2102. doi:10.1056/NEJMoa1911440

61. Thapa B, De Sarkar N, Giri S, Sharma K, Kim M, Kilari D. Integrating PARP Inhibitors in mCRPC therapy: current strategies and emerging trends. Cancer Manag Res. 2024;16:1267–1283. doi:10.2147/CMAR.S411023

62. 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 Oncol. 2020;21(1):162–174. doi:10.1016/S1470-2045(19)30684-9

63. 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(32):3763–3772. doi:10.1200/JCO.20.01035

64. 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(9):1250–1264. doi:10.1016/S1470-2045(21)00376-4

65. 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(3):362–373. doi:10.1016/S1470-2045(21)00757-9

66. Fizazi K, Piulats JM, Reaume MN, et al. Rucaparib or physician’s choice in metastatic prostate cancer. N Engl J Med. 2023;388(8):719–732. doi:10.1056/NEJMoa2214676

67. Loriot Y, Bianchini D, Ileana E, et al. Antitumour activity of Abiraterone acetate against metastatic castration-resistant prostate cancer progressing after docetaxel and enzalutamide (MDV3100). Ann Oncol Off J Eur Soc Med Oncol. 2013;24(7):1807–1812. doi:10.1093/annonc/mdt136

68. Oh WK, Cheng WY, Miao R, et al. Real-world outcomes in patients with metastatic castration-resistant prostate cancer receiving second-line chemotherapy versus an alternative androgen receptor-targeted agent (ARTA) following early progression on a first-line ARTA in a US community oncology setting. Urol Oncol. 2018;36(11):500.e1–500.e9. doi:10.1016/j.urolonc.2018.08.002

69. 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(480):eaam7479. doi:10.1126/scisignal.aam7479

70. Schiewer MJ, Goodwin JF, Han S, et al. Dual roles of PARP-1 promote cancer growth and progression. Cancer Discov. 2012;2(12):1134–1149. doi:10.1158/2159-8290.CD-12-0120

71. Schiewer MJ, Knudsen KE. Transcriptional roles of PARP1 in cancer. Mol Cancer Res. 2014;12(8):1069–1080. doi:10.1158/1541-7786.MCR-13-0672

72. 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(7):975–986. doi:10.1016/S1470-2045(18)30365-6

73. Clarke NW, Armstrong AJ, Thiery-Vuillemin A, et al. Abiraterone and olaparib for metastatic castration-resistant prostate cancer. NEJM Evid. 2022;1(9):EVIDoa2200043. doi:10.1056/EVIDoa2200043

74. Chi KN, Rathkopf D, Smith MR, et al. Niraparib and abiraterone acetate for metastatic castration-resistant prostate cancer. J Clin Oncol Off J Am Soc Clin Oncol. 2023;41(18):3339–3351. doi:10.1200/JCO.22.01649

75. 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 Lond Engl. 2023;402(10398):291–303. doi:10.1016/S0140-6736(23)01055-3

76. Hussain M, Mateo J, Fizazi K, et al. Survival with olaparib in metastatic castration-resistant prostate cancer. N Engl J Med. 2020;383(24):2345–2357. doi:10.1056/NEJMoa2022485

77. Saad F, Clarke NW, Oya M, et al. Olaparib plus abiraterone versus placebo plus abiraterone in metastatic castration-resistant prostate cancer (PROpel): final prespecified overall survival results of a randomised, double-blind, phase 3 trial. Lancet Oncol. 2023;24(10):1094–1108. doi:10.1016/S1470-2045(23)00382-0

78. 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 Off J Eur Soc Med Oncol. 2023;34(9):772–782. doi:10.1016/j.annonc.2023.06.009

79. Chi KN, Canil C, Alimohamed N, Soulières D, Breau RH. 2023 European society for medical oncology (ESMO) congress meeting highlights. Can Urol Assoc J J Assoc Urol Can. 2024;18(1):E46–E52. doi:10.5489/cuaj.8688

80. 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(6_suppl):TPS176. doi:10.1200/JCO.2021.39.6_suppl.TPS176

81. 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. 2023;41(6_suppl):TPS279. doi:10.1200/JCO.2023.41.6_suppl.TPS279

82. Mateo J, Lord CJ, Serra V, et al. A decade of clinical development of PARP inhibitors in perspective. Ann Oncol Off J Eur Soc Med Oncol. 2019;30(9):1437–1447. doi:10.1093/annonc/mdz192

83. 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 Off J Am Soc Clin Oncol. 2023;41(22):3839–3850. doi:10.1200/JCO.23.00233

84. 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(16_suppl):5005. doi:10.1200/JCO.2023.41.16_suppl.5005

85. Quinn Z, Leiby B, Sonpavde G, et al. Phase I study of niraparib in combination with radium-223 for the treatment of metastatic castrate-resistant prostate cancer. Clin Cancer Res Off J Am Assoc Cancer Res. 2023;29(1):50–59. doi:10.1158/1078-0432.CCR-22-2526

86. 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(4):511–518. doi:10.1158/1535-7163.MCT-22-0583

87. 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 Off J Am Soc Clin Oncol. 2023;41(4):871–880. doi:10.1200/JCO.21.02947

88. Pook D, Geynisman DM, Carles J, et al. A phase Ib, open-label study evaluating the safety and efficacy of ipatasertib plus rucaparib in patients with metastatic castration-resistant prostate cancer. Clin Cancer Res Off J Am Assoc Cancer Res. 2023;29(17):3292–3300. doi:10.1158/1078-0432.CCR-22-2585

89. Park W, Chen J, Chou JF, et al. Genomic methods identify homologous recombination deficiency in pancreas adenocarcinoma and optimize treatment selection. Clin Cancer Res Off J Am Assoc Cancer Res. 2020;26(13):3239–3247. doi:10.1158/1078-0432.CCR-20-0418

90. Heeke AL, Pishvaian MJ, Lynce F, et al. Prevalence of homologous recombination-related gene mutations across multiple cancer types. JCO Precis Oncol. 2018;2018:PO.17.00286. doi:10.1200/PO.17.00286

91. Golan T, Hammel P, Reni M, et al. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med. 2019;381(4):317–327. doi:10.1056/NEJMoa1903387

92. Kindler HL, Hammel P, Reni M, et al. Overall survival results from the POLO trial: a phase III study of active maintenance olaparib versus placebo for germline BRCA-mutated metastatic pancreatic cancer. J Clin Oncol Off J Am Soc Clin Oncol. 2022;40(34):3929–3939. doi:10.1200/JCO.21.01604

93. Reiss KA, Mick R, O’Hara MH, et al. Phase II study of maintenance rucaparib in patients with platinum-sensitive advanced pancreatic cancer and a pathogenic germline or somatic variant in BRCA1, BRCA2, or PALB2. J Clin Oncol Off J Am Soc Clin Oncol. 2021;39(22):2497–2505. doi:10.1200/JCO.21.00003

94. Shroff RT, Hendifar A, McWilliams RR, et al. Rucaparib monotherapy in patients with pancreatic cancer and a known deleterious BRCA Mutation. JCO Precis Oncol. 2018;2018:PO.17.00316. doi:10.1200/PO.17.00316

95. Teke ME, Saif A, Ryan CE, Lux SC, Hernandez JM, Reiss KA. A randomized study of olaparib or placebo in patients with surgically removed pancreatic cancer who have a BRCA1, BRCA2 or PALB2 mutation (The APOLLO Trial). Ann Surg Oncol. 2022;29(9):5375–5376. doi:10.1245/s10434-022-11917-2

96. Lowery MA, Kelsen DP, Capanu M, et al. Phase II trial of veliparib in patients with previously treated BRCA-mutated pancreas ductal adenocarcinoma. Eur J Cancer Oxf Engl 1990. 2018;89:19–26. doi:10.1016/j.ejca.2017.11.004

97. Keane F, O’Connor CA, Park W, Seufferlein T, O’Reilly EM. Pancreatic cancer: BRCA targeted therapy and beyond. Cancers. 2023;15(11):2955. doi:10.3390/cancers15112955

98. Tang M, Wang Y, Li P, Han R, Wang R. Assessing the benefits and safety profile of incorporating poly ADP-ribose polymerase (PARP) inhibitors in the treatment of advanced lung cancer: a thorough systematic review and meta-analysis. Front Pharmacol. 2024;15:1338442. doi:10.3389/fphar.2024.1338442

99. Postel-Vinay S, Coves J, Texier M, et al. Olaparib maintenance versus placebo in platinum-sensitive non-small cell lung cancer: the Phase 2 randomized PIPSeN trial. Br J Cancer. 2024;130(3):417–424. doi:10.1038/s41416-023-02514-5

100. Gao A, Wang X, Wang J, Zhong D, Zhang L. Homologous recombination deficiency status predicts response to immunotherapy-based treatment in non-small cell lung cancer patients. Thorac Cancer. 2024;15(25):1842–1853. doi:10.1111/1759-7714.15408

101. Paturel A, Hall J, Chemin I. Poly(ADP-Ribose) polymerase inhibition as a promising approach for hepatocellular carcinoma therapy. Cancers. 2022;14(15):3806. doi:10.3390/cancers14153806

102. Joris S, Denys H, Collignon J, et al. Efficacy of olaparib in advanced cancers with germline or somatic mutations in BRCA1, BRCA2, CHEK2 and ATM, a Belgian precision tumor-agnostic phase II study. ESMO Open. 2023;8(6):102041. doi:10.1016/j.esmoop.2023.102041

103. Vaidyanathan A, Sawers L, Gannon AL, et al. ABCB1 (MDR1) induction defines a common resistance mechanism in paclitaxel- and olaparib-resistant ovarian cancer cells. Br J Cancer. 2016;115(4):431–441. doi:10.1038/bjc.2016.203

104. Lawlor D, Martin P, Busschots S, et al. PARP inhibitors as P-glyoprotein substrates. J Pharm Sci. 2014;103(6):1913–1920. doi:10.1002/jps.23952

105. Leitner I, Nemeth J, Feurstein T, et al. The third-generation P-glycoprotein inhibitor tariquidar may overcome bacterial multidrug resistance by increasing intracellular drug concentration. J Antimicrob Chemother. 2011;66(4):834–839. doi:10.1093/jac/dkq526

106. Singh A, Settleman J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene. 2010;29(34):4741–4751. doi:10.1038/onc.2010.215

107. Foroni C, Broggini M, Generali D, Damia G. Epithelial-mesenchymal transition and breast cancer: role, molecular mechanisms and clinical impact. Cancer Treat Rev. 2012;38(6):689–697. doi:10.1016/j.ctrv.2011.11.001

108. Xu Z, Zhang Y, Dai H, Han B. Epithelial-mesenchymal transition-mediated tumor therapeutic resistance. Mol. 2022;27(15):4750. doi:10.3390/molecules27154750

109. Yang J, Weinberg RA. Epithelial-mesenchymal transition: at the crossroads of development and tumor metastasis. Dev Cell. 2008;14(6):818–829. doi:10.1016/j.devcel.2008.05.009

110. Allison Stewart C, Tong P, Cardnell RJ, et al. Dynamic variations in epithelial-to-mesenchymal transition (EMT), ATM, and SLFN11 govern response to PARP inhibitors and cisplatin in small cell lung cancer. Oncotarget. 2017;8(17):28575–28587. doi:10.18632/oncotarget.15338

111. Jaspers JE, Sol W, Kersbergen A, et al. BRCA2-deficient sarcomatoid mammary tumors exhibit multidrug resistance. Cancer Res. 2015;75(4):732–741. doi:10.1158/0008-5472.CAN-14-0839

112. Ordonez LD, Hay T, McEwen R, et al. Rapid activation of epithelial-mesenchymal transition drives PARP inhibitor resistance in Brca2-mutant mammary tumours. Oncotarget. 2019;10(27):2586–2606. doi:10.18632/oncotarget.26830

113. Murai J, Tang SW, Leo E, et al. SLFN11 blocks stressed replication forks independently of ATR. Mol Cell. 2018;69(3):371–384.e6. doi:10.1016/j.molcel.2018.01.012

114. Nogales V, Reinhold WC, Varma S, et al. Epigenetic inactivation of the putative DNA/RNA helicase SLFN11 in human cancer confers resistance to platinum drugs. Oncotarget. 2015;7(3):3084–3097. doi:10.18632/oncotarget.6413

115. Scattolin D, Maso AD, Ferro A, et al. The emerging role of Schlafen-11 (SLFN11) in predicting response to anticancer treatments: focus on small cell lung cancer. Cancer Treat Rev. 2024;128:102768. doi:10.1016/j.ctrv.2024.102768

116. Zoppoli G, Regairaz M, Leo E, et al. Putative DNA/RNA helicase Schlafen-11 (SLFN11) sensitizes cancer cells to DNA-damaging agents. Proc Natl Acad Sci. 2012;109(37):15030–15035. doi:10.1073/pnas.1205943109

117. Lok BH, Gardner EE, Schneeberger VE, et al. PARP inhibitor activity correlates with SLFN11 expression and demonstrates synergy with temozolomide in small cell lung cancer. Clin Cancer Res Off J Am Assoc Cancer Res. 2017;23(2):523–535. doi:10.1158/1078-0432.CCR-16-1040

118. Willis SE, Winkler C, Roudier MP, et al. Retrospective analysis of Schlafen11 (SLFN11) to predict the outcomes to therapies affecting the DNA damage response. Br J Cancer. 2021;125(12):1666–1676. doi:10.1038/s41416-021-01560-1

119. Pommier Y, O’Connor MJ, de Bono J. Laying a trap to kill cancer cells: PARP inhibitors and their mechanisms of action. Sci Transl Med. 2016;8(362):362ps17. doi:10.1126/scitranslmed.aaf9246

120. Pettitt SJ, Rehman FL, Bajrami I, et al. A genetic screen using the PiggyBac transposon in haploid cells identifies Parp1 as a mediator of olaparib toxicity. PLoS One. 2013;8(4):e61520. doi:10.1371/journal.pone.0061520

121. Oplustilova L, Wolanin K, Mistrik M, et al. Evaluation of candidate biomarkers to predict cancer cell sensitivity or resistance to PARP-1 inhibitor treatment. Cell Cycle. 2012;11(20):3837–3850. doi:10.4161/cc.22026

122. Tobalina L, Armenia J, Irving E, O’Connor MJ, Forment JV. A meta-analysis of reversion mutations in BRCA genes identifies signatures of DNA end-joining repair mechanisms driving therapy resistance. Ann Oncol. 2021;32(1):103–112. doi:10.1016/j.annonc.2020.10.470

123. Pettitt SJ, Frankum JR, Punta M, et al. Clinical BRCA1/2 reversion analysis identifies hotspot mutations and predicted neoantigens associated with therapy resistance. Cancer Discov. 2020;10(10):1475–1488. doi:10.1158/2159-8290.CD-19-1485

124. Swisher EM, Kristeleit RS, Oza AM, et al. Characterization of patients with long-term responses to rucaparib treatment in recurrent ovarian cancer. Gynecol Oncol. 2021;163(3):490–497. doi:10.1016/j.ygyno.2021.08.030

125. Nesic K, Krais JJ, Wang Y, et al. BRCA1 secondary splice-site mutations drive exon-skipping and PARP inhibitor resistance. mol Cancer. 2024;23(1):158. doi:10.1186/s12943-024-02048-1

126. Kondrashova O, Topp M, Nesic K, et al. Methylation of all BRCA1 copies predicts response to the PARP inhibitor rucaparib in ovarian carcinoma. Nat Commun. 2018;9(1):1–16. doi:10.1038/s41467-018-05564-z

127. Wu S, Yao X, Sun W, Jiang K, Hao J. Exploration of poly (ADP-ribose) polymerase inhibitor resistance in the treatment of BRCA1/2-mutated cancer. Genes Chromosomes Cancer. 2024;63(5):e23243. doi:10.1002/gcc.23243

128. ter Brugge P, Kristel P, van der Burg E, et al. Mechanisms of therapy resistance in patient-derived xenograft models of BRCA1-deficient breast cancer. JNCI J Natl Cancer Inst. 2016;108(11):djw148. doi:10.1093/jnci/djw148

129. Belotserkovskaya R, Raga Gil E, Lawrence N, et al. PALB2 chromatin recruitment restores homologous recombination in BRCA1-deficient cells depleted of 53BP1. Nat Commun. 2020;11(1):1–11. doi:10.1038/s41467-020-14563-y

130. Kondrashova O, Nguyen M, Shield-Artin K, et al. Secondary somatic mutations restoring RAD51C and RAD51D associated with acquired resistance to the PARP inhibitor rucaparib in high-grade ovarian carcinoma. Cancer Discov. 2017;7(9):984–998. doi:10.1158/2159-8290.CD-17-0419

131. Lord CJ, Ashworth A. Mechanisms of resistance to therapies targeting BRCA-mutant cancers. Nat Med. 2013;19(11):1381–1388. doi:10.1038/nm.3369

132. Min A, Im SA, Yoon YK, et al. RAD51C-deficient cancer cells are highly sensitive to the PARP inhibitor olaparib. Mol Cancer Ther. 2013;12(6):865–877. doi:10.1158/1535-7163.MCT-12-0950

133. Mitri Z, Goodyear SM, Mills G. Strategies for the prevention or reversal of PARP inhibitor resistance. Expert Rev Anticancer Ther. 2024;24:1–17. doi:10.1080/14737140.2024.2393251

134. Nesic K, Kondrashova O, Hurley RM, et al. Acquired RAD51C promoter methylation loss causes PARP inhibitor resistance in high-grade serous ovarian carcinoma. Cancer Res. 2021;81(18):4709–4722. doi:10.1158/0008-5472.CAN-21-0774

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.