Abstract
The use of poly(ADP-ribose) polymerase inhibitors (PARPi) exploits synthetic lethality in solid tumors with homologous recombination repair (HRR) defects. Significant clinical benefit has been established in breast and ovarian cancers harboring BRCA1/2 mutations, as well as tumors harboring characteristics of “BRCAness”. However, the durability of treatment responses is limited and emerging data have demonstrated the clinical challenge of PARPi resistance. With the expanding use of PARPi, the significance of PARP therapy in patients pre-treated with PARPi remains in need of significant further investigation. Molecular mechanisms contributing to this phenomenon include restoration of HRR function, replication fork stabilization, BRCA1/2 reversion mutations and epigenetic changes. Current studies are evaluating the utility of combination therapies of PARPi with cell cycle checkpoint inhibitors, anti-angiogenic agents, PI3K/AKT pathway inhibitors, MEK inhibitors, and epigenetic modifiers to overcome this resistance. In this review, we address the mechanisms of PARPi resistance supported by preclinical models, examine current clinical trials applying combination therapy to overcome PARPi resistance, and discuss future directions to enhance the clinical efficacy of PARPi.
Keywords: PARPi, homologous recombination, PARPi resistance, PARPi combination therapy
The significance of PARP inhibition, synthetic lethality and beyond
Poly(ADP-ribose) polymerase (PARP) 1 is a member of a family of enzymes with a critical role in the base excision repair (BER) mechanism of single stranded DNA breaks (SSB), a component of the larger DNA damage repair (DDR) machinery (1–3). Upon sensing and binding to SSB, the catalytic domain of PARP1 undergoes “auto-PARylation”, forming poly(ADP-ribose) complexes that ultimately recruit SSB repair proteins, promote chromatin remodeling, and induce repair of DNA damage (1,2,4,5). Dysfunction in PARylation leads to the accumulation of SSB causing replication-dependent DNA double strand breaks (DSB), as well as “PARP trapping”, whereby PARP1 is unable to dissociate from sites of DNA damage (6–8). PARP inhibitor (PARPi)-induced PARP trapping causes replication fork collapse and an insurmountable accumulation of DSB for cellular viability, leading to clinically relevant cytotoxicity (8–10).
To restore normal DNA architecture at sites of DSB, cells rely on one of two mechanisms: homologous recombination (HR) or non-homologous end joining (NHEJ). While healthy cells can depend on BRCA1/2 proteins and the error-free HR mechanism of DSB repair, cancer cells with germline BRCA1/2 (gBRCA1/2) mutations or HR deficiency (HRD) must resort to the lower fidelity NHEJ repair mechanisms, leading to gene alterations and deletions. The subsequent chromosomal instability promotes overwhelming levels of DNA damage and replication stress (1,11) In BRCA1/2 mutant tumors, defective DSB mechanisms force a reliance on PARP-induced SSB repair mechanisms. The opportune use of PARPi in the setting of synthetic lethality is thus the rationale supporting the clinical application and success of PARPi in this subset of exquisitely sensitive tumor cells compared to wild-type counterparts (4,12).
Clinically, patients with gBRCA1/2 mutations have demonstrated an increased propensity for breast, ovarian, and prostate cancers (13,14). Among patients with ovarian cancer, 20% harbor BRCA1/2 mutations, the majority of which are germline mutations (15,16). With more granular analysis, half of patients with high grade serous ovarian cancer have HRD tumors, exhibiting sensitivity to PARPi comparable to their BRCA1/2-mutated counterparts (17). While this “BRCAness” phenotype has been explained by hypermethylation of the BRCA1 gene promotor, loss of heterozygosity of BRCA1/2, and loss of function mutations or methylation of other HRR genes (BRCA1/2, RAD51C, RAD51D, PALB2), HRD is a dynamic process and there are likely additional relevant mechanisms that have yet to be defined (18).
A number of clinical trials have led to FDA approval for the use of olaparib, niraparib and rucaparib with specific clinical indications (Table 1). FDA approval for PARPi use in the maintenance setting of ovarian cancer following platinum-based chemotherapy, for example, stems from the Phase III PAOLA-1 trial demonstrating progression free survival (PFS) benefit of 22.1 months with olaparib and bevacizumab compared to 16.6 months with bevacizumab alone (HR 0.59; 95% CI 0.49–0.72; p<0.001) (19). Furthermore, 12 BRCA2-mutated ovarian cancer patients had a preliminary disease control rate of 92% with use of niraparib and bevacizumab compared to niraparib alone in the phase 1 ENGOT-ov24 clinical trial (20).
Table 1:
PARP Inhibitor Use by FDA Designation and Associated Clinical Trial
| Primary Disease | |||
|---|---|---|---|
| Agent | Treatment Indication | Upfront Indication | Clinical Trial |
| Olaparib (Lynparza) | gBRCA/sBRCAm ovarian cancer | Ovarian, fallopian tube, peritoneal cancer after partial or complete response to initial platinum-based therapy | SOLO-1 (NCT01844986) |
| HRD ovarian cancer(in combination with bevacizumab) | Ovarian, fallopian tube, peritoneal cancer after partial or complete response to initial platinum-based therapy | PAOLA-1 (NCT03737643) | |
| gBRCAm metastatic pancreatic adenocarcinoma | Maintenance treatment for disease without progression on at least 16 weeks of first-line platinum-based chemotherapy | POLO (NCT02184195) | |
| Niraparib (Zejula) | Any advanced ovarian, fallopian tube, peritoneal cancer | Ovarian, fallopian tube, peritoneal cancer after partial or complete response to initial platinum-based therapy | PRIMA (NCT02655016) |
| Secondary Disease | |||
| Agent | Treatment Indication | Recurrence Indication | Clinical Trial |
| Olaparib (Lynparza) | gBRCAm ovarian cancer | Ovarian cancer treated with three or more prior lines of chemotherapy | Study 42 (NCT01078662) |
| gBRCAm, HER2-negative metastatic breast cancer | Metastatic breast cancer treated with chemotherapy in the neoadjuvant, adjuvant or metastatic setting | OlympiAD (NCT02000622) | |
| Recurrent HRD mCRPC | mCRPC with progression following prior treatment with enzalutamide or abiraterone | PROFOUND (NCT02987543) | |
| Rucaparib (Rubraca) | gBRCA/sBRCA mutant ovarian cancer | Ovarian, fallopian tube, peritoneal cancer treated with two or more prior lines of chemotherapy | ARIEL2 (NCT01891344) STUDY 10 (NCT01482715) |
| BRCA1/2-mutatant mCRPC | Prostate cancer treated with both androgen deprivation therapy and chemotherapy | TRITON2 (NCT02952534) | |
| Niraparib (Zejula) | gBRCA/sBRCAm ovarian cancer OR genomic instability and who have progressed more than six months after response to the last platinum-based chemotherapy | Ovarian, fallopian tube, peritoneal cancer treated with three or more prior lines ofchemotherapy | QUADRA (NCT02354586) |
| Talazoparib (Talzenna) | gBRCAm, HER2-negative locally advanced or metastatic breast cancer | EMBRACA (NCT01945775) | |
| Agent | Treatment Indication | Second Line Maintenance Indication | Clinical Trial |
| Olaparib (Lynparza) | Any patient with recurrent ovarian, fallopian tube, peritoneal cancer | Platinum-sensitive ovarian, fallopian tube, peritoneal cancer after partial or complete response to platinum-based therapy | SOLO-2 (NCT1874353) |
| Rucaparib (Rubraca) | Any patient with recurrent ovarian, fallopian tube, peritoneal cancer | Platinum-sensitive ovarian, fallopian tube, peritoneal cancer after partial or complete response to platinum-based therapy | ARIEL3 (NCT01968213) |
| Niraparib (Zejula) | Any recurrent ovarian, fallopian tube, peritoneal cancer | Platinum-sensitive ovarian, fallopian tube, peritoneal cancer after partial or complete response to platinum-based therapy | ENGOT-OV16/NOVA (NCT01847274) |
Mechanisms of de novo and adaptive PARPi resistance
Clinical trials have demonstrated an acquired drug resistance to PARPi after prolonged exposure, likely contributing to the observed treatment failure rate of 40% of patients (21). Proposed mechanisms of resistance include restoration of the HRR machinery and stabilization of the replication fork, components of which contribute to the DNA replication stress response necessary to maintain genomic integrity (22). Additionally, BRCA1/2 reversion mutations and epigenetic modifications have also been associated with PARPi resistance. Given FDA approval of olaparib, niraparib and rucaparib in the treatment and maintenance setting of recurrent ovarian cancer, as well as current studies exploring the benefit of PARPi use in the frontline setting, understanding the mechanisms of PARPi resistance has become critical for effective patient selection and to produce durable clinical responses (23).
Restoring the HRR machinery
Of the two coordinated cellular repair mechanisms for DNA DSB, the HR machinery has higher fidelity due to use of the sister chromatid as a template in the repair process. Resection of DSB ends into single-strand DNA (ssDNA) is initiated in the S/G2 phase by the MRE11-Rad50-Nbs1 (MRN) complex, C-terminal binding protein interacting protein (CtIP), and other nucleases such as EXO-1 and DNA2; this DNA end resection induces the HR pathway (24,25). The ssDNA overhanging sections are coated by replication protein A (RPA), leading to activation of gH2AX by ataxia-telangiectasia mutated (ATM) and ataxia-telangiectasia and Rad3 related (ATR) kinases, part of the phosphatidylinositol 3-kinase (PI3K) family that respond to DNA replication stress (26). Upon further signal transduction, BRCA1 and p-53 binding protein 1 (53BP1) are recruited to the sites of DSB and activate CtIP (27). Functional CtIP creates 3’ overhangs at which a complex of BRCA2, PALB2, and Rad51 works in conjunction to form a nucleoprotein filament D-loop, protecting further DNA degradation and ultimately completing HRR (28,29).
Restoring HR function through alteration of deficient HR-related proteins or inhibitor molecules, thus allowing DSB repair and decreasing replication stress, can lead to PARPi resistance (23). One proposed mechanism of PARPi resistance involves cyclin-dependent kinases (CDKs), which dictate DNA end resection of HR through phosphorylation of CtIP and nucleases, as well as activation and regulation of ATR/ATM (25,30). In vitro studies using OVCAR-8 cells demonstrate that impaired CDK12 function in ovarian cancer cells leads to decreased BRCA1 levels, compromised HR function, and sensitivity to the PARPi, veliparib (16). Furthermore, the chromatin binding protein 53BP1 is a negative regulator of DNA end resection, by promoting binding of the Shieldin effector protein complex onto ssDNA overhangs, leading to molecular alterations in the Shieldin genes (23,31). Decreased 53BP1 activity allows for DNA end resection in the absence of BRCA1, recruitment of RAD51 and, thus, HR restoration. Altered 53BP1 function is thus associated with decreased PARPi efficacy in BRCA1-mutated breast and ovarian cancers (32,33). Interestingly, HR restoration due to loss of 53BP1 does not occur in BRCA2-deficient tumors (34).
Stabilization of the replication fork
Functional PARP1 and BRCA1/2 proteins play key roles in stabilizing the DNA replication fork and maintaining genomic integrity (35). PARP1 regulates replication stress and BRCA1/2 proteins protect DNA strands from degradation by the MRN complex (36,37). Following the concept of synthetic lethality, PARPi-induced PARP trapping leads to an overwhelming accumulation of DSB and clinically significant cytotoxicity in the absence of BRCA1/2-dependent prevention of DNA degradation and replication fork protection (23). Emerging evidence points to the protection of the DNA replication fork as the driver of PARPi resistance. Decreased EZH2 activity inhibits MUS81 nuclease, which has been associated with PARPi resistance in BRCA2-mutant breast cancer cells through replication fork stabilization rather than HR (38,39). BRCA1/2-mutant breast and ovarian tumors have been shown to have FANCD2 overexpression and increased activity, also associated with fork stability and PARPi-resistance in these cell lines (40). The loss of FANCD2 in these tumors lead to increased cytotoxicity, implying a central role in replication fork protection (40). Furthermore, PTIP, a MLL3/4 recruitment protein complex that works in conjunction with 53BP1 to inhibit MRE11-initiated DNA strand degradation, is also shown to have a protective role in BRCA-deficient cells. Inactivation of PTIP in B-lymphocytes inhibited MRN complex recruitment to stalled replication forks, and thus prevented further DNA degradation without affecting HR activity (41). Replication fork stability is thus a feasible mechanism by which BRCA-deficient tumors evade cytotoxicity and acquire drug resistance.
BRCA1/2 reversion mutations and the restoration of BRCAness
Secondary BRCA1/2 reversion mutations are an established mechanism for PARP1 resistance (42). These somatic mutations may take the form of amino acid insertions or deletions to restore a functional, wild-type protein, or restore the open reading frame (ORF) of BRCA1/2 to ultimately allow DSB by HR (42,43). The exact etiology of these reversion mutations—sporadic or drug-induced—is unclear. The frequency of reversion mutations is also unknown, though the intentional use of ctDNA and liquid biopsy in clinical trials offers some insight, suggesting that reversion mutations may vary between tumor types (43). Among 24/1534 patients with metastatic castration-resistant prostate cancer (mCRPC) with gBRCA1/2 mutations and history of PARPi or platinum chemotherapy exposure, the frequency of BRCA2 reversion mutations was estimated to be 40% (44). Within a cohort of 97 patients with high grade serous ovarian cancer and either germline or somatic BRCA1/2 mutations undergoing treatment with rucaparib on clinical trial, 8.2% patients had BRCA1/2 mutation reversion prior to treatment and 10.3% exhibited reversion post-progression. The presence of BRCA1/2 reversion mutations was directly associated with decreased clinical benefit from rucaparib (45).
Interestingly, reversion mutations have also been described in other HRR genes. RAD51 has been implicated in PARPi resistance among triple negative breast cancer (TNBC) patients (46). In the ARIEL2 trial of rucaparib in platinum-sensitive, recurrent HGSOC, RAD51C/D reversion mutations were associated with PARPi resistance in cohort analysis (47,48).
Epigenetic modifications
Prolonged exposure to PARPi treatment has been associated with epigenetic modifications to PARP1, BRCA1/2, and other proteins essential to DDR—including post-translational histone modifications, chromatin remodeling, DNA methylation, and RNA-mediated processes—affecting protein function and altering sensitivity to PARPi. For example, the demethylation of BRCA1 has been shown to restore BRCA1 expression, reinstate the HR machinery, and thus impart PARPi resistance to BRCA1-mutated and BRCA1-methylated patient-derived xenograft (PDX) models of breast cancer (49). In the phase II ARIEL 2 study of rucaparib treatment for platinum-sensitive, relapsed ovarian cancer, analysis of pre-treatment and post-treatment tumor biopsy samples revealed that heterozygous demethylation of BRCA1 was specifically attributed to PARPi resistance (48,50). This mechanism of resistance has been demonstrated clinically in the recurrent setting of both PARPi and platinum-based chemotherapy treatments (48,51). Quantification of BRCA1 methylation prior to treatment has potential to determine the benefit and utility of PARPi therapy (50).
Other epigenetic modifications associated with PARPi resistance includ de-ubiquitination of the BARD1 BRCT1 domain by the USP15 enzyme (52). The functional N-terminal RING domain of BRCA1 dictates its function and forms a heterodimer with BARD1 to regulate DNA end resection, forming a single-stranded template upon which the essential recruitment of BRCA2/PALB2 and RAD51 occurs to carry out HR (53). As the BRCA1/BARD1 complex is essential to RAD51 stimulation, de-ubiquitination of the BARD1 domain and consequent retention of BRCA1/BARD1 complex at sites of DSB promotes recruitment of downstream tumor suppressor proteins, leading to PARPi resistance (52). Post-translational modifications to PARP1 have also been demonstrated to cause resistance to PARPi treatment in preclinical studies of TNBC. The phosphorylation of PARP1 protein by MET in the setting of oxidative DNA damage conferred increased PARP1 enzymatic activity, leading to reduced binding and activity of PARPi. MET inhibitors re-sensitized breast and lung cancer xenografts to PARPi therapy (23). Furthermore, the acetylation of 53BP1 creates a cellular environment analogous to 53BP1 loss, which inhibits NHEJ, restores HR function, and thus causes resistance to PARP inhibition in BRCA1-mutated tumors (54,55). This 53BP1-mediated rescue of BRCA1 deficiency has been demonstrated in BRCA-mutated, TNBC and suggests a clinically relevant mechanism of PARPi resistance (34).
Combination therapy strategies to circumvent PARPi resistance
The established association of prolonged PARPi exposure with resistance to therapy necessitates efforts to circumvent PARPi resistance, especially given increasing use of PARPi as maintenance and treatment of multiple solid tumors. Preclinical and early stage clinical trials are utilizing rational combination strategies to re-sensitize tumor cells to PARP inhibition, as well as induce sensitivity to PARPi in BRCA1/2 wildtype tumors or those with HR proficiency (23). In targeting the various pathways and functional steps of HR restoration, replication fork stabilization, and BRCA1/2 reversion mutations, these strategies may manipulate tumor cells’ ability to evade DNA damage (Table 2).
Table 2.
A Summary of PARPi combination Clinical Trials
| Combination therapy | Trial | Phase | Study popoulation | |
|---|---|---|---|---|
| PARPi + WEE1 inhibitor | Olaparib + AZD1775 | NCT03579316 | II | Recurrent fallopian tube, ovarian and primary peritoneal cancers |
| NCT04197713 | I | Advanced solid tumors with selected mutations and PARP resistance | ||
| NCT02576444 | II | Tumors harboring TP53 or KRAS mutations | ||
| NCT02511795 | I | Refractory solid tumors, Relapsed SCLC | ||
| PARPi + ATR inhibitor | Olaparib + AZD6738 | NCT02576444 (OLAPCO) | II | Solid tumors haboring mutations leading to dysregulation of PI3K/AKT pathway |
| NCT04065269 (ATARI) | II | Gynecologic cancers | ||
| NCT03787680 | II | Prostate cancer | ||
| NCT03878095 | II | IDH1 and IDH2 mutant tumors | ||
| NCT03462342 (CAPRI) | II | High grade serous ovarian cancers | ||
| NCT03428607 | II | SCLC | ||
| NCT03682289 | II | Clear cell renal cell cancer; metastatic renal cell cancer; metastatic urothelial cancer; metastatic pancreatic cancer; locally advanced pancreatic cancer | ||
| Niraparib + BAY1895344 | NCT04267939 | Ib | Advanced solid tumors and ovarian cancer | |
| PARPi + CHK1 inhibitor | Olaparib + prexasertib | NCT03057145 | I | Advanced solid tumors |
| PARPi + anti-angiogenics | Olaparib + bevacizumab (PAOLA-1) | NCT02477644 | III | High grade serous ovarian cancers |
| Niraparib + bevacizumab | NCT02354131 | II | Platinum-sensitive recurrent ovarian cancer | |
| Olaparib + cediranib (EVOLVE) | NCT02681237 | II | HGSOC with progression post-PARPi | |
| Olaparib + cediranib v olaparib alone | NCT0111648 | II | Relapsed platinum-sensitive ovarian cancer of high grade serous or endometrioid histology or deleterious gBRCA1/2 mutation | |
| Olaparib + cediranib v olaparib alone v standard platinum-based chemo | NCT02446600 | III | Recurrent platinum-sensitive ovarian cancer | |
| PARPi + PI3K inhibitor | Olaparib + BKM120 or BYL719 | NCT01623349 | Ib | Recurrent TNBC or HGSOC |
| Olaparib + vistusertib (AZD 2014) or olaparib + capivasertib (AZD5363) | NCT02208375 | Ib/II | Recurrent endometrial, TNBC, ovarian, primary peritoneal or fallopain tube cancer | |
| Olaparib + capivasertib | NCT02338622 | Ib | BRCA1/2 and non-BRCA1/2 mutant cancers | |
| Niraparib + copanlisib | NCT03586661 | Ib | Recurrent endometrial, ovarian, primary peritoneal or fallopain tube cancer | |
| PARPi + MAPK pathway inhibitor | Olaparib + selumetinib | NCT03162627 | I/II | Endometrial, ovarian and other solid tumors with Ras pathway alterations and ovarian tumors with PARP resistance |
| PARPi + IO | Olaparib + durvalumab | NCT03594396 | I/II | Resectable stage II/III TNBC |
| NCT03772561 (MEDIPAC) | I | Advanced or metastatic solid tumors | ||
| NCT03842228 | I | DDR-mutated unresectable, advanced or metastatic solid tumors | ||
| NCT03775486 | II | Metastatic NSCLC | ||
| NCT03579784 | II | Unresectable or recurrent gastric carcinoma | ||
| NCT03810105 | II | Recurrent prostate cancer | ||
| NCT04053322 (DOLAF) | II | Locally advanced or metastatic ER+ HER2- breast cancer | ||
| NCT03851614 (DAPPER) | II | Locally advanced or metastatic MMR proficient CRC, pancreatic adenocarcinoma or leiomyosarcoma | ||
| NCT03951415 (DOMEC) | II | Recurrent, refractory or metastatic endometrial cancer or carcinosarcoma of the endometrium | ||
| NCT03167619 (DORA) | II | Locally advanced or metastatic platinum-treated TNBC | ||
| NCT02734004 (MEDIOLA) | II | gBRCAm platinum-sensitive ovarian cancer; gBRCAm HER2- breast cancer; platinum-resistant relapsed gastric cancer | ||
| NCT03801369 | II | Metastatic TNBC | ||
| NCT03459846 (BAYOU) | II | Advanced or metastatic platinum-ineligible urothelial carcinoma | ||
| NCT02484404 | II | Advanced, recurrent or metastatic ovarian, TNBC, lung, prostate, CRC or solid tumors | ||
| NCT03534492 (NEODURVARIB) | II | Resectable urothelial cancer | ||
| NCT03737643 (DUO-O) | III | Newly diagnosed advanced ovarian, fallopian tube or primary peritoneal carcinoma or carcinosarcoma | ||
| NCT04269200 (DUO-E) | III | Newly diagnosed advanced or recurrent endometrial carcinoma | ||
| olaparib + pembrolizumab | NCT02861573 (KEYNOTE-365) | Umb | Previously treated mCRPC | |
| olaparib + tremelimumab | NCT02571725 | I | BRCA1/2 mutated recurrent ovarian cancer | |
| olaparib + tremelimumab + durvalumab | NCT02953457 | I/II | DDR mutated recurrent ovarian, fallopian tube or primary peritoneal cancer | |
| niraparib + pembrolizumab | NCT02657889 (TOPACIO-KEYNOTE 162) | II | Advanced or metastatic TNBC or platinum-resistant ovarian cancer | |
| niraparib + PD-1 inhibitor | NCT03308942 | II | Locally advanced or metastatic NSCLC | |
| niraparib + TSR-042 | NCT03307785 | I/II | Solid tumors | |
| niraparib + TSR-042 + platinum-based therapy | NCT03602859 | III | Nonmucinous epithelial ovarian cancer | |
| niraparib + atezolizumab | NCT03522246 (ANITA) | III | Recurrent ovarian, fallopian tube, or primary peritoneal carcinoma | |
| rucaparib + nivolumab | NCT03572478 | I/II | mCRPC or recurrent endometrial cancer | |
| NCT03522246 (ATHENA) | III | Front line ovarian cancer maintenance | ||
| NCT03338790 (CheckMate 9KD) | III | mCRPC | ||
| rucaparib + atezolizumab | NCT04276376 (ARIANES) | II | DDR-deficient or platinum sensitive solid tumors | |
| NCT03694262 (EndoBARR) | II | Recurrent progressive endometrial carcinoma | ||
| talazoparib + avelumab | NCT03330405 (Javelin PARP Medley) | Ib/II | Previously treated advanced solid tumors | |
| NCT03565991 (Javelin BRCA/ATM) | BRCA/ATM-mutant solid tumors | |||
| NCT03637491 (Javelin BRCA/ATM) | Ras-mutant solid tumors | |||
| NCT03642132 (Javelin Ovarian PARP 100) | III | Front line ovarian cancer | ||
| BGB-A317 + BGB-290 | NCT02660034 | Basket | Ovarian cancer, TNBC, mCRPC, bladder cancer, SCLC, HER2 (−) gastric cancer, pancreatic cancer, other solid tumors | |
| veliparib + atezolizumab | NCT02849496 | HER2 (−), BRCA-mutant TNBC | ||
| pamiparib + tislelizumab | NCT02660034 | I | Previously treated advanced solid tumors |
Combining PARPi + ATR, CHK1, and Wee1 inhibitors
Targeting the downstream effector molecules and signaling pathways of HR has yielded promising clinical benefit regardless of HR status. To ensure cellular survival amidst DNA damage, cells activate cell cycle checkpoints to halt the cell cycle and stimulate DNA damage repair (DDR) mechanisms to repair the SSB and accumulated DSB through PARPi and replication fork collapse in S phase. Due to reliance on p-53 dependent S phase checkpoint, tumors harboring p53 mutations, namely ovarian cancers, must rely instead on G2 checkpoints to induce DDR (56). Specifically targeting ATR, Chk1, and Wee1 has been shown to restore HR and replication fork stability, thereby re-sensitizing BRCA1/2 deficient tumors to PARP inhibition (23,57).
Inhibition of ATR prevents G2/M cycle arrest, disrupts the recruitment of PALB2/BRCA2 and RAD51 onto sites of DNA damage, and thus creates an environment of HRD and replication fork instability (58). The synergism of DDR and cell cycle regulation in the combination of PARPi and ATRi has resulted in tumor suppression BRCA2-mutant PDX models (59). The CAPRI phase II single arm study combining olaparib with ceralasertib in HGSOC is currently recruiting and includes cohorts based on platinum sensitivity and prior PARPi exposure (NCT03462342). Another phase II study is also currently recruiting patients with gynecologic cancers, including clear cell ovarian or endometrial cancers with ARID1A mutations, and is analyzing the effects of single-agent berzosertib versus combination with olaparib (NCT04065269). Additionally, a phase Ib trial combines ATRi (Bay 1895344) with niraparib and includes three experimental cohorts: 1) HRD solid tumors 2) platinum resistant, HRD, PARPi naïve HGSOC 3) PARPi-resistant HGSOC (NCT04267939). These studies will not only provide further information on mechanisms of PARPi resistance, but also help to delineate which populations of patients will optimally benefit from combination treatment of ATRi and PARPi.
Activated by ATR and regulated by transcription factor E2F7, Chk1 interacts with downstream Cdc25a and Cdc25c molecules and ultimately regulates RAD51 expression and recruitment (60). Chk1 inhibition in combination with PARPi has shown similar benefit to ATRi in combination with PARPi in preclinical models, regardless of BRCA1/2 status (59,61). In a phase 1 study of the Chk1 inhibitor prexasertib combined with olaparib in HGSOC and other solid tumors, four of the 18 treated patients with BRCA1-mutant, PARPi resistant HGSOC achieved partial response (PR). Moreover, increased expression of phosphorylated histone-2AX (γH2AX), a molecular marker of DNA damage and replication stress, and pRPA, as well as reduction in RAD51 was noted in tumor biopsy samples post-treatment, further supporting mechanisms of PARPi resistance and potential markers for response (62).
Wee1 is an additional modulator of the G2/M checkpoint and is activated by Chk1 (57). There are several phase I and II trials evaluating the combination of PARPi with Wee1 inhibitors in ovarian cancer, solid tumors with PARPi resistance, and tumors harboring TP53 or KRAS mutations. The phase II EFFORT trial combining olaparib and adavosertib has shown clinical benefit in PARPi-resistant ovarian cancers (NCT03579316). The ORR for single agent adavosertib versus combination treatment was 23% and 29%, respectively, with CBR 63% and 89%, respectively (63).
Combining PARPi + Immunotherapy
Preclinical studies have provided evidence for synergy between PARPi and immunotherapy (IO) agents via two mechanisms: 1) PARPi-induced accumulation of overwhelming DNA damage and cytosolic DNA, and 2) PARPi-induced T cell modulation and PD-L1 upregulation (64–67). These molecular changes ultimately prime the tumor microenvironment and, in setting of HR deficiency, create an opportune role for the use of immune checkpoint blockade (ICB) in combination with PARP inhibition to enhance the clinical efficacy of each agents (68–71).
As demonstrated in HR deficient and/or BRCA1/2-mutated tumors, the inhibition of PARP1 leads to a catastrophic accumulation of cytotoxic DNA DSB and collapse of the replication fork (1). This overwhelming tumor mutational burden (TMB) is recognized by the immune microenvironment through damage-associated molecular patterns (DAMP), specifically cytosolic DNA-cycling GMP-AMP synthase complex (cGAS) (72). Subsequent recruitment of stimulator interferon genes (STING) and activation of the cGAS-STING pathway leads to downstream infiltration of T cells, increased type 1 IFN expression and signaling, and paracrine stimulation of dendritic cells (73,74). Both talazoparib and olaparib have shown efficacy in activating the cGAS-STING pathway and inducing T cell infiltration in BRCA1-deficient murine models (70,75). Preclinical models using breast cancer lines also demonstrate PARPi-induced upregulation of PD-L1 expression through the inactivation of GSK3β (76). Murine models of breast, ovarian and colorectal murine models have demonstrated promising antitumor activity with the combination of anti-PD-1/L1 with olaparib, talazoparib and rucaparib (67). Downstream from PARP inhibition, there is further evidence that CTLA-4 suppresses the antitumor immune via increased T-cell infiltration and IFN-y production; preclinical models of BRCA1-deficient mice with ovarian cancer demonstrate improved survival with the combination of veliparib and anti-CTLA-4 (77).
Currently, two phase II trials combining PARPi and IO have shown promising results. The phase II MEDIOLA basket trial of olaparib and durvalumab in advanced solid cancers, including TNBC, ovarian, and uterine cancers (NCT02734004), demonstrated objective response rate (ORR) and disease control rate (DCR) of 63% and 81% at 12 weeks for patients with germline BRCA1/2 mutations and platinum-sensitive disease, respectively. Patients pre-treated with prior lines of chemotherapy had an ORR of 68% (68,78). In the open-label, phase II TOPACIO trial (NCT02657889), the effects of niraparib in combination with pembrolizumab were evaluated in women with advanced or metastatic TNBC or recurrent ovarian carcinoma, irrespective of BRCA1/2 status. Those with BRCA1/2 mutations had improved ORR (45%) and disease control rates (73%) compared to the entire study cohort (25% and 68%, respectively). Interestingly, there was also clinical benefit in BRCA1/2 wildtype and HR-proficient patients (ORR 24% and 27%, respectively), suggesting benefit beyond mutational status (69). Additionally, with preclinical and phase 1 results suggesting therapeutic effect with the combination of PARPi and anti-CTLA-4 therapy in heavily-pretreated patients with gBRCA recurrent ovarian cancer (NCT02571725), a phase II study is evaluating olaparib versus the combination of olaparib and tremelimumab in platinum-sensitive, recurrent ovarian cancer (NCT04034927) (79).
Combining PARPi + anti-angiogenic agents
Preclinical data suggest a synergistic role between PARP inhibition and anti-angiogenic agents through two mechanisms: 1) inhibition of BRCA1/2 expression and 2) exacerbation of cellular hypoxia leading to genetic instability (80). In a model of BRCA2-mutated ovarian cancer stemlike cancer cells with confirmed BRCA2 reversion, chemosensitivity was restored with inhibition of VEGFR3 (81). While it is established that tumoral hypoxia promotes genetic instability with altered HR protein expression, the use of anti-angiogenics has been associated with increased hypoxia, impaired HR, and sensitivity to PARPi treatment (82,83).
Clinical trials combining the anti-VEGF agent, bevacizumab, with PARPi present contradictory findings based on HRD status. In exploratory subgroup analysis of olaparib in combination with bevacizumab in ovarian cancer patients enrolled in the PAOLA-1 trial (NCT02477644), HR deficiency was associated with improved progression free survival (PFS) with HR 0.33 compared to 0.92 in the HR proficient patient group (19). Conversely, in the randomized phase II ENGOT-ov24 trial (NCT02354131) of bevacizumab and niraparib in combination versus niraparib alone in the treatment of platinum-sensitive recurrent ovarian cancer, clinical benefit was demonstrated irrespective of HR status. In subgroup analysis, the HR proficient cohort had a HR of 0.4 (20).
A randomized phase II study of olaparib in combination with cediranib, an oral VEGF inhibitor (VEGFi), versus niraparib alone demonstrated improved PFS in recurrent platinum-sensitive ovarian cancers with combination therapy (16.5 v 8.2 months, HR 0.50; p=0.007). In post-hoc exploratory analyses, while PFS and OS were similar in the two arms of gBRCAm patients, significant PFS and OS benefit was observed in the gBRCA1/2 wild-type cohort with combination therapy (23.7 v 5.7 months, HR 0.31; p=0.0013; 37.8 v 23 months, HR 0.44; p=0.047) (84). In the subsequent phase III trial comparing combination therapy to standard platinum-based chemotherapy in patients with recurrent, platinum-sensitive HGSOC, there was no clinical benefit to olaparib + cediranib compared to standard of care (SOC) therapy and the primary endpoint of PFS was not met (85). Patients with gBRCA1/2 mutations accounted for 23.7% of the total study population and HR for PFS was 0.55 with combination therapy and 0.63 for single-agent olaparib versus SOC; for gBRCA1/2 wild-type patients, a HR of 0.97 and 1.41 was observed for combination therapy and SOC therapy, respectively (85). An open-label single arm clinical and translational phase II trial (EVOLVE, NCT02681237) exploring the use of cediranib and olaparib for HGSOC in the setting of post-PARPi progression demonstrated varying ORR and clinical benefit according to platinum-sensitivity—0% in the platinum sensitive (55% PFS rate at 16 weeks), 20% in the platinum-resistant (50% PFS), and 8% in the exploratory (39% PFS) cohorts, respectively—and mechanism of PARPi resistance. Whole exome and RNA sequencing analysis reported 19% of the acquired genomic alterations were BRCA1, BRCA2, or RAD51B reversion mutations (86). Further investigation is needed regarding clinical benefit of combination therapy not only according to overall HRD status, but within cohorts of different PARPi resistance mechanisms.
Combining PARPi + PI3K/AKT pathway inhibitors
The PI3K/AKT pathway is responsible for cell growth, survival, and regulation of metabolism, as well as serving as a sensor for DNA DSB within the DDR machinery (87). Aberrant activation is found in several tumor types, including TNBC. In preclinical models of BRCA wildtype, PTEN-mutant TNBC, silencing of PIK3CA ultimately led to an accumulation of phosphorylated histone-2AX (γ-H2AX), a molecular marker of DNA damage and replication stress. High levels of γ-H2AX support the concept that PIK3CA loss offers a potential synergy with PARPi (88,89). Investigators further demonstrated that the inhibition of PI3K impairs BRCA1/2 expression and can sensitize BRCA1/2-proficient TNBC to PARPi (90). Phase 1 trials combining PI3Ki and PARPi have also suggested that altering the PI3K pathway can sensitize a population of HR-proficient and BRCA1/2 wildtype ovarian cancer patients to PARPi (91,92). In a phase I dose escalation trial of buparlisib and olaparib (NCT01623349), PR was observed in 12/46 BRCA1/2 wildtype ovarian cancer patients (91). Similarly, the phase Ib trial combining alpelisib and olaparib showed similar rates of PR in both gBRCA1/2 wildtype and gBRCA1/2m ovarian cancer patients (92). A phase I trial combining olaparib with AKT inhibitor capivasertib further suggested that combination therapy may re-sensitize tumors to PARPi treatment. Of the 64 patients with solid tumors, 25 patients had epithelial ovarian cancer and 11 of these patients achieved clinical benefit. Four of the patients with clinical benefit were considered PARPi resistant (93). It has been suggested that decreased mTOR activity is associated with response in ovarian and TNBC, while high tyrosine kinase receptor activity and mTOR activation is associated with resistance (94). However further exploration should refine markers of treatment response and the specific populations that may benefit from combination therapy.
Combining PARPi + Ras/Raf/MEK/MAPK pathway inhibitors
While KRAS mutations are an established driver of low grade rather than high grade ovarian tumors, the Ras/Raf/MEK/MAPK (MAPK) pathway is activated in ~25% of HGSOC due to high mutational burden and gene copy number variation (CNV) (95,96). This signaling pathway is ultimately responsible for tumor cell proliferation, differentiation, and survival (95). Preclinical models of ovarian cancer cells lines treated with PARPi demonstrated enhanced activation of the MAPK pathway and downregulation of its targets, including FOXO3a and BIM1; these targets were found to be restored in cell lines with adaptive PARPi resistance. The use of MEK inhibitors (MEKi) was found to re-sensitize RAS-mutant pancreatic and ovarian cancer cell lines to PARPi with induction of FOXO3a, leading to alteration of PARP1 expression and function, as well as decreased expression of the MRN complex and BRCA1/2 (97). The downregulation of these key components of HR in RAS-mutant tumors by PARP inhibition ultimately lead to impaired DDR and genomic instability, thereby suggesting another potential synergistic mechanism of overcoming resistance to treatment (96). There is currently a phase I/II trial (NCT03162627) evaluating the use of olaparib with selumetinib in RAS-mutated solid tumors and ovarian tumors with PARPi resistance (23).
Combining PARPi + Epigenetic modifiers (BET and HDAC inhibitors)
Targets involved in transcriptional regulation and/or chromatin remodeling represent another class of drugs in development to induce HRD in solid tumors. Preclinical data including cell line and xenograft ovarian cancer models suggest that inhibition of bromodomains (BDs) and histone deacetylases (HDACs) may enhance the activity of PARPi (98–100). Mechanisms proposed for this synergy include enhanced PARP-related DNA damage, decreased BRCA1, and increased genomic instability via disruption of replication fork progression (100). However, the clinical promise of these agents has been limited by toxicity. Initial dose-finding studies performed in patients with hematologic malignancy demonstrated that BET inhibition was strongly correlated to thrombocytopenia (101–103). A recent Phase I trial of ODM-207 (a BETi) in select solid tumors demonstrated comparable toxicities and narrow therapeutic window (104).
HDAC inibitors as single agents have shown limited efficacy in solid tumors, and, given the parallel involvement at the transcriptional level, HDACi results in similar side effect profile to BETi (105). Therefore, there is significant interest in combination strategies. Ongoing clinical work investigating HDACi with PARPi includes early phase trials, such as a phase I/II trial of entinostat in combination with olaparib for women with recurrent ovarian cancer (NCT03924245), and belinostat in combination with talazoparib for multiple solid tumor types (NCT04703920).
Summary and future directions
With FDA approval of several PARP inhibitors both in the frontline and secondline maintenance and treatment settings, as well as proven clinical benefit across several solid tumor types, the increasing use of PARP inhibitors has yielded the clinical challenge of PARPi resistance. Restoration of the HR machinery, stabilization of the DNA replication fork, BRCA reversion mutations, and epigenetic molecular modifications are established mechanisms contributing to PARPi resistance. Preclinical models have offered insight into these various mechanisms yet analyses of paired tumor samples and clinical response in early phase clinical trials have suggested that multiple mechanisms may be inherent to progression on PARPi treatment. The use of cfDNA has allowed for more in-depth investigation into the adaptive genetic changes that may occur while on treatment and should be expanded to future clinical trial analyses.
Early clinical trials are utilization rational combination strategies to evade PARPi resistance, including: PARPi + ATR/Chk1/Wee1 inhibitors, PARPi + IO, PARPi + anti-angiogenic agents, PARPi + PI3K/AKT pathway inhibitors, PARPi + Ras/Raf/MEK/MAPK pathway inhibitors, and PARPi + epigenetic modifiers. While preliminary analyses have suggested promising response and clinical benefit, durability of treatment response is unclear and combination treatment is not without additive toxicity profiles. Further exploration of meaningful molecular markers of response are needed to delineate which patient cohorts will benefit most from each combination therapy. Consideration of employing ctDNA analyses to assess for BRCA and other HRR reversion mutations may be beneficial to help tailor both treatment choice, as well as optimize treatment duration. Dosing schedules must also be further investigated to reduce toxicity while maintaining benefit beyond that expected with each monotherapy.
Acknowledgments
Conflicts of Interest and Source of Funding: S Westin has clinical research funding from ArQule, AstraZeneca, Bayer, Clovis Oncology, Cotinga Pharmaceuticals, Novartis, Roche/Genentech, and Tesaro. S. Westin receives consulting fees from AstraZeneca, Clovis Oncology, MediVation, Merck, Ovation, Pfizer, Takeda, and Tesaro. This work was also supported in part by the MD Anderson Cancer Center Support Grant from the National Cancer Institute of the National Institutes of Health (NIH/NCI P30 CA016672), Ovarian SPORE NIH 1P50CA217685–01, the GOG Foundation and the T32 training grant (NIH/NCI CA101642).
References
- 1.Cerrato A, Morra F, Celetti A. Use of poly ADP-ribose polymerase [PARP] inhibitors in cancer cells bearing DDR defects: The rationale for their inclusion in the clinic. J Exp Clin Cancer Res [Internet]. 2016;35(1):1–13. Available from: 10.1186/s13046-016-0456-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Satoh M, Lindahl T. Role of poly(ADP-ribose) formation in DNA repair. Nature. 1992;356(6367):356–8. [DOI] [PubMed] [Google Scholar]
- 3.Howard S, Yanez D, Stark J. DNA damage response factors from diverse pathways, including DNA crosslink repair, mediate alternative end joining. PLoS Genet. 2015;11(1):e1004943. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Brown JS, O’Carrigan B, Jackson SP, Yap TA. Targeting DNA repair in cancer: Beyond PARP inhibitors. Cancer Discov. 2017;7(1):20–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Huarte E, Cubillos-Ruiz JR, Nesbeth YC, Scarlett UK, Martinez DG, Buckanovich RJ, et al. Depletion of dendritic cells delays ovarian cancer progression by boosting antitumor immunity. Cancer Res [Internet]. 2008/09/05. 2008;68(18):7684–91. Available from: https://www.ncbi.nlm.nih.gov/pubmed/18768667 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Barkauskaite E, Jankevicius G, Ahel I. Structures and Mechanisms of Enzymes Employed in the Synthesis and Degradation of PARP-Dependent Protein ADP-Ribosylation. Mol Cell [Internet]. 2015;58(6):935–46. Available from: 10.1016/j.molcel.2015.05.007 [DOI] [PubMed] [Google Scholar]
- 7.Lord CJ, Ashworth A. PARP Inhibitors: The First Synthetic Lethal Targeted Therapy. Science (80- ) [Internet]. 2017;355(6330):1152–8. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6175050/pdf/emss-79613.pdf [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Murai J, Huang SYN, Das BB, Renaud A, Zhang Y, Doroshow JH, et al. Trapping of PARP1 and PARP2 by clinical PARP inhibitors. Cancer Res. 2012;72(21):5588–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Shen Y, Rehman FL, Feng Y, Boshuizen J, Bajrami I. BMN 673, a novel and highly potent PARP1 / 2 inhibitor for the treatment of human cancers with DNA repair deficiency. 2019;19(18):5003–15. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6485449/pdf/emss-54129.pdf [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Pommier Y, O’Connor M, 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] [PubMed] [Google Scholar]
- 11.Roy R, Chun J, Powell S. BRCA1 and BRCA2: different roles in a common pathway of genome protection. Nat Rev Cancer. 2011;12(1):68–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Moynahan ME, Cui TY, Jasin M. Homology-directed DNA repair, mitomycin-C resistance, and chromosome stability is restored with correction of a Brca1 mutation. Cancer Res. 2001;61(12):4842–50. [PubMed] [Google Scholar]
- 13.Rouleau M, Patel A, Hendzel M, Kaufmann S, Poirier G. PARP inhibition: PARP1 and beyond. Nat Rev Cacner. 2010;10:293–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Chaudhuri A, Nussenzweig A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling Arnab. Nat Rev Mol Cell Biol. 2017;18(10):610–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Nielsen F, van Overeem H, Sorensen C. Hereditary breast and ovarian cancer: new genes in confined pathways. Nat Rev Cancer. 2016;16(599–612). [DOI] [PubMed] [Google Scholar]
- 16.Joshi PM, Sutor SL, Huntoon CJ, Karnitz LM. Ovarian cancer-associated mutations disable catalytic activity of CDK12, a kinase that promotes homologous recombination repair and resistance to cisplatin and poly(ADP-ribose) polymerase inhibitors. J Biol Chem. 2014;289(13):9247–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Cancer Genome Atlas Research N. Integrated genomic analyses of ovarian carcinoma. Nature. 2011;474(609–15). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Miller R, Leary A, Scott C, et al. ESMO recommendations on predictive biomarker testing for homologous recombination deficiency and PARP inhibitor benefit in ovarian cancer. Ann Oncol. 2020;31(12):1606–22. [DOI] [PubMed] [Google Scholar]
- 19.Ray-Coquard I, Pautier P, Pignata S, Pérol D, González-Martín A, Berger R, et al. Olaparib plus Bevacizumab as First-Line Maintenance in Ovarian Cancer. N Engl J Med. 2019;381(25):2416–28. [DOI] [PubMed] [Google Scholar]
- 20.Mirza M, Lundqvist E, Birrer M, Christensen RD, Nyvang G, Malander S, et al. Niraparib plus bevacizumab versus niraparib alone for platinum-sensitive recurrent ovarian cancer (NSGO-AVANOVA2/ENGOT-ov24): a randomised, phase 2, superiority trial. Lancet Oncol. 2019;20(10):1409–19. [DOI] [PubMed] [Google Scholar]
- 21.Li H, Liu ZY, Wu N, Chen YC, Cheng Q, Wang J. PARP inhibitor resistance: The underlying mechanisms and clinical implications. Mol Cancer. 2020;19(1):1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Ngoi NY, Pham M, Tan DS, Yap TA. Targeting the replication stress response through synthetic lethal strategies in cancer medicine. Trends Cancer. 2021;21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lee EK, Matulonis UA. Parp inhibitor resistance mechanisms and implications for post-progression combination therapies. Cancers (Basel). 2020;12(8):1–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Cejka P. DNA end resection: Nucleases team up with the right partners to initiate homologous recombination. J Biol Chem. 2015;290(38):22931–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Yun MH, Hiom K. CtIP-BRCA1 modulates the choice of DNA double-strand break repair pathway throughout the cell cycle. Nature. 2009;459(7245):460–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Weber AM, Ryan AJ. ATM and ATR as therapeutic targets in cancer. Pharmacol Ther [Internet]. 2015;149:124–38. Available from: 10.1016/j.pharmthera.2014.12.001 [DOI] [PubMed] [Google Scholar]
- 27.Isono M, Niimi A, Oike T, Hagiwara Y, Sato H, Sekine R, et al. BRCA1 directs the repair pathway to homologous recombination by promoting 53BP1 Dephosphorylation. Cell Rep. 2017;18:520–32. [DOI] [PubMed] [Google Scholar]
- 28.Sy S, Huen M, Chen J. PALB2 is an integral component of the BRCA complex required for homologous recombination repair. Proc Natl Acad Sci U S A. 2009;106:7155–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Hanenberg H, Andreassen P. PALB2 (partner and localizer of BRCA2). Atlas Genet Cytogenet Oncol Haematol. 2018;22(484–90). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Tomimatsu N, Mukherjee B, Catherine Hardebeck M, Ilcheva M, Vanessa Camacho C, Louise Harris J, et al. Phosphorylation of EXO1 by CDKs 1 and 2 regulates DNA end resection and repair pathway choice. Nat Commun. 2014;5:3561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Noordermeer SM, Adam S, Setiaputra D, Barazas M, Pettitt SJ, Ling AK, et al. The shieldin complex mediates 53BP1-dependent DNA repair. Vol. 560, Nature. 2018. 117–121 p. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hurley RM, Hendrickson AEW, Visscher DW, Ansell P, Harrell MI, Wagner JM, et al. 53BP1 as a Potential Predictor of Response in PARP Inhibitor-Treated Homologous Recombination-Deficient Ovarian Cancer. Gynecol Oncol. 2019;153(1):127–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Jaspers J, Kersbergen A, Boon U, Sol W, van Deemter L, et al. Loss of 53BP1 Causes PARP Inhibitor Resistance in Brca1-Mutated Mouse Mammary Tumors. Cancer Discov. 2013;3(1):68–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Bouwman P, Aly A, Escandell JM, Pieterse M, Bartkova J, Van Der Gulden H, et al. 53BP1 loss rescues BRCA1 deficiency and is associated with triple-negative and BRCA-mutated breast cancers. Nat Struct Mol Biol. 2010;17(6):688–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Schlacher K, Christ N, Siaud N, Egashira A, Wu H, Jasin M. Double-strand break repair-independent role for BRCA2 in blocking stalled replication fork degradation by MRE11. Cell. 2011;145(4):529–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Maya-Mendoza A, Moudry P, Merchut-Maya JM, Lee M, Strauss R, Bartek J. High speed of fork progression induces DNA replication stress and genomic instability. Nature. 2018;559(7713):279–84. [DOI] [PubMed] [Google Scholar]
- 37.Schlacher K, Wu H, Jasin M. A Distinct Replication Fork Protection Pathway Connects Fanconi Anemia Tumor Suppressors to RAD51-BRCA1/2. Cancer Cell. 2012;22(1):106–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Schlacher K. PARPi focus the spotlight on replication fork protection in cancer. Nat Cell Biol. 2017;19:1309–10. [DOI] [PubMed] [Google Scholar]
- 39.Rondinelli B, Gogola E, Yucel H, Duarte A, van de Ven M, van der Sluijs R, et al. EZH2 promotes degradation of stalled replication forks by recruiting MUS81 through histone H3 trimethylation. Nat Cell Biol. 2017;19(11):1371–8. [DOI] [PubMed] [Google Scholar]
- 40.Kais Z, Rondinelli B, Holmes A, O’Leary C, Kozono D, et al. FANCD2 maintains fork stability in BRCA1/2-deficient tumors and promotes alternative end-joining DNA repair. Cell Rep. 2016;15(11):2488–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chaudhuri AR, Callen E, Ding X, Gogola E, Duarte AA, Lee E, et al. Replication Fork Stability Confers Chemoresistance in BRCA-deficient Cells. 2017;535(7612):382–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Norquist B, Wurz KA, Pennil CC, Garcia R, Gross J, Sakai W, et al. Secondary somatic mutations restoring BRCA1/2 predict chemotherapy resistance in hereditary ovarian carcinomas. J Clin Oncol. 2011;29(22):3008–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Mayor P, Gay LM, Gornstein E, Morley S, Frampton GM, Heilmann A, et al. BRCA1/2 reversion mutations revealed in breast and gynecologic cancers sequenced during routine clinical care using tissue or liquid biopsy. J Clin Oncol [Internet]. 2017. May 20;35(15_suppl):5551. Available from: 10.1200/JCO.2017.35.15_suppl.5551 [DOI] [Google Scholar]
- 44.Carneiro BA, Collier KA, Nagy RJ, Pamarthy S, Sagar V, Fairclough S, et al. Acquired Resistance to Poly (ADP-ribose) Polymerase Inhibitor Olaparib in BRCA2 -Associated Prostate Cancer Resulting From Biallelic BRCA2 Reversion Mutations Restores Both Germline and Somatic Loss-of-Function Mutations. JCO Precis Oncol. 2018;(2):1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Lin KK, Harrell MI, Oza AM, Oaknin A, Ray-Coquard I, Tinker AV., et al. BRCA Reversion Mutations in Circulating Tumor DNA Predict Primary and Acquired Resistance to the PARP Inhibitor Rucaparib in High-Grade Ovarian Carcinoma. Cancer Discov. 2019;9(2):210–9. [DOI] [PubMed] [Google Scholar]
- 46.Liu Y, Burness M, Martin-Trevino R, Guy J, Bai S, Harouaka R, et al. RAD51 Mediates Resistance of Cancer Stem Cells to PARP Inhibition in Triple-Negative Breast Cancer. Clin Cancer Res. 2017;23(2). [DOI] [PubMed] [Google Scholar]
- 47.Swisher EM, Lin KK, Oza AM, Scott CL, Giordano H, et al. Rucaparib in relapsed, platinum-sensitive high-grade ovarian carcinoma (ARIEL2 Part 1): an international, multicentre, open-label, phase 2 trial. Lancet Oncol. 2017;18(1):75–87. [DOI] [PubMed] [Google Scholar]
- 48.Kondrashova O, Nguyen M, Shield-artin K, Tinker AV, Nelson NH, Harrell MI, 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):984–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Ter Brugge P, Kristel P, Van Der Burg E, Boon U, De Maaker M, Lips E, et al. Mechanisms of therapy resistance in patient-derived xenograft models of brca1-deficient breast cancer. J Natl Cancer Inst. 2016;108(11):1–12. [DOI] [PubMed] [Google Scholar]
- 50.Kondrashova O, Topp M, Nesic K, Lieschke E, Ho GY, Harrell MI, et al. Methylation of all BRCA1 copies predicts response to the PARP inhibitor rucaparib in ovarian carcinoma. Nat Commun. 2018;9(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Patch A, Christie E, Etemadmoghadam D, Garsed D, George J, Fereday S, et al. Whole-genome characterization of chemoresistant ovarian cancer. Nature. 2015;521(7553):489–94. [DOI] [PubMed] [Google Scholar]
- 52.Peng Y, Liao Q, Tan W, Peng C, Hu Z, Chen Y, et al. The deubiquitylating enzyme USP15 regulates homologous recombination repair and cancer cell response to PARP inhibitors. Nat Commun [Internet]. 2019;10(1). Available from: 10.1038/s41467-019-09232-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zhao W, Steinfeld JB, Liang F, Chen X, Maranon DG, Ma CJ, et al. Promotion of RAD51-mediated homologous DNA pairing by BRCA1-BARD1. Nature. 2017;47(3):549–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Guo X, Bai Y, Zhao M, Zhou M, Shen Q, Yun CH, et al. Acetylation of 53BP1 dictates the DNA double strand break repair pathway. Nucleic Acids Res. 2018;46(2):689–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kass EM, Moynahan ME, Jasin M. Loss of 53BP1 Is a Gain for BRCA1 Mutant Cells. Cancer Cell [Internet]. 2010;17(5):423–5. Available from: 10.1016/j.ccr.2010.04.021 [DOI] [PubMed] [Google Scholar]
- 56.Rundle S, Bradbury A, Drew Y, Curtin N. Targeting the ATR-CHK1 axis in cancer therapy. Cancers (Basel). 2017;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Haynes B, Murai J, Lee J. Restored replication fork stabilization, a mechanism of PARP inhibitor resistance, can be overcome by cell cycle checkpoint inhibition. Cancer Treat Rev. 2018;71:1–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Yazinski SA, Comaills V, Buisson R, Genois MM, Nguyen HD, Ho CK, et al. ATR inhibition disrupts rewired homologous recombination and fork protection pathways in PARP inhibitor-resistant BRCA-deficient cancer cells. Genes Dev. 2017;31(3):318–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Kim H, George E, Ragland R, Rafial S, Zhang R, et al. Targeting the ATR/CHK1 axis with PARP inhibition results in tumor regression in BRCA mutant models. Clin Cancer Res. 2017;23(12):3097–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Clements KE, Thakar T, Nicolae CM, Liang X, Wang HG, Moldovan GL. Loss of E2F7 confers resistance to poly-ADP-ribose polymerase (PARP) inhibitors in BRCA2-deficient cells. Nucleic Acids Res. 2018;46(17):8898–907. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Brill E, Yokoyama T, Nair J, Yu M, Ahn YR, Lee JM. Prexasertib, a cell cycle checkpoint kinases 1 and 2 inhibitor, increases in vitro toxicity of PARP inhibition by preventing Rad51 foci formation in BRCA wild type high-grade serous ovarian cancer. Oncotarget. 2017;8(67):111026–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Do K, Kochupurakkal B, Kelland S, de Jonge A, Hedglin J, et al. Phase 1 Combination Study of the CHK1 Inhibitor Prexasertib and the PARP Inhibitor Olaparib in High-grade Serous Ovarian Cancer and Other Solid Tumors. Clin Cancer Res. 2021; [DOI] [PubMed] [Google Scholar]
- 63.Westin S, Coleman R, Fellman B, Yuan Y, Sood A, et al. EFFORT: EFFicacy Of adavosertib in parp ResisTance: A randomized two-arm non-comparative phase II study of adavosertib with or without olaparib in women with PARP-resistant ovarian cancer. J Clin Oncol. 2021;39(15). [Google Scholar]
- 64.Stewart RA, Pilie PG, Yap TA. Development of PARP and immune-checkpoint inhibitor combinations. Cancer Res. 2018;78(24):6717–25. [DOI] [PubMed] [Google Scholar]
- 65.Chabanon RM, Muirhead G, Krastev DB, Adam J, Morel D, Garrido M, et al. PARP inhibition enhances tumor cell–intrinsic immunity in ERCC1-deficient non–small cell lung cancer. J Clin Invest. 2019;129(3):1211–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Pantelidou C, Sonzogni O, Taveira MDO, Mehta AK, Kothari A, Wang D, et al. Parp inhibitor efficacy depends on CD8+ T-cell recruitment via intratumoral sting pathway activation in brca-deficient models of triple-negative breast cancer. Cancer Discov. 2019;9(6):722–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Mosely SIS, Prime JE, Sainson RCA, Koopmann JO, Wang DYQ, Greenawalt DM, et al. Rational selection of syngeneic preclinical tumor models for immunotherapeutic drug discovery. Cancer Immunol Res. 2017;5(1):29–41. [DOI] [PubMed] [Google Scholar]
- 68.Drew Y, de Jonge M, Hong S, Park Y, Wolfer A, Brown J et al. An open- label, phase II basket study of olaparib and durvalumab (MEDIOLA): results in germline BRCA-mutated (gBRCAm) platinum-sensitive relapsed (PSR) ovarian cancer (OC). Gynecol Oncol. 2018;149:246–7. [Google Scholar]
- 69.Konstantinopoulos PA, Waggoner S, Vidal GA, Mita M, Moroney JW, Holloway R, 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):1141–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Boussios S, Karihtala P, Moschetta M, Karathanasi A, Sadauskaite A, Rassy E, et al. Combined strategies with poly (ADP-ribose) polymerase (PARP) inhibitors for the treatment of ovarian cancer: A literature review. Diagnostics. 2019;9(3). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Farkkila A, Gulhan D, Casado J, et al. Immunogenomic profiling determines responses to combined PARP and PD-1 inhibition in ovarian cancer. Nat Commun. 2020;11:1459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Gubin MM, Artyomov MN, Mardis ER, Schreiber RD. Tumor neoantigens: building a framework for personalized cancer immunotherapy. J Clin Invest. 2015;125(9):3413–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Chen Q, Sun L, Chen Z. Regulation and function of the cGAS-STING pathway of cytosolic DNA sensing. Nat Immunol. 2016;17:1142–9. [DOI] [PubMed] [Google Scholar]
- 74.Shen J, Zhao W, Ju Z, Wang L, Peng Y, Labrie M, et al. PARPi Triggers the STING-Dependent Immune Response and Enhances the Therapeutic Efficacy of Immune Checkpoint Blockade Independent of BRCAness. Cancer Res. 2019;79(2):311–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Barber GN. STING: Infection, inflammation and cancer. Nat Rev Immunol. 2015;15(12):760–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Peyraud F, Italiano A. Combined parp inhibition and immune checkpoint therapy in solid tumors. Cancers (Basel). 2020;12(6):1–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Higuchi T, Flies D, Marjon N, Mantia-Smaldone G, Ronner L, Gimotty P, et al. CTLA-4 Blockade Synergizes Therapeutically with PARP Inhibition in BRCA1-Deficient Ovarian Cancer. Cancer Immunol Res. 2015;3(11):1257–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Drew Y, Kaufman B, Banerjee S, Lortholary A, Hong SH, Park YH, et al. Phase II study of olaparib + durvalumab (MEDIOLA): Updated results in germline BRCA-mutated platinum-sensitive relapsed (PSR) ovarian cancer (OC). Ann Oncol [Internet]. 2019;30(October):v485–6. Available from: 10.1093/annonc/mdz253.016 [DOI] [Google Scholar]
- 79.Adams S, Rixe O, Lee J, McCance D, Westgate S, et al. Phase 1 study combining olaparib and tremelimumab for the treatment of women with BRCA-deficient recurrent ovarian cancer. J Clin Oncol. 2017;35(15). [Google Scholar]
- 80.Kaplan A, Gueble S, Liu Y, Oeck S, Kim H, Yun Z, et al. Cediranib suppresses homology-directed DNA repair through down-regulation of BRCA1/2 and RAD51. Sci Transl Med. 2019;11(492). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Lim J, Yang K, Taylor-Harding B, Ruprecht Wiedemeyer W, Buckanovich RJ. VEGFR3 inhibition chemosensitizes ovarian cancer stemlike cells through down-regulation of BRCA1 and BRCA2. Neoplasia (United States) [Internet]. 2014;16(4):343–353.e2. Available from: 10.1016/j.neo.2014.04.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Chan N, Koritzinsky M, Zhao H, Bindra R, Glazer PM, Powell S, et al. Chronic hypoxia decreases synthesis of homologous recombination proteins to offset chemoresistance and radioresistance. Cancer Res. 2008;68(2):605–14. [DOI] [PubMed] [Google Scholar]
- 83.Bristow R, Hill R. Hypoxia, DNA repair and genetic instability. Nat Rev Cancer. 2008;8:180–92. [DOI] [PubMed] [Google Scholar]
- 84.Liu JF, Barry WT, Birrer M, Lee JM, Buckanovich RJ, Fleming GF, et al. Overall survival and updated progression-free survival outcomes in a randomized phase II study of combination cediranib and olaparib versus olaparib in relapsed platinum-sensitive ovarian cancer. Ann Oncol. 2019;30(4):551–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Liu J, Brady M, Matulonis U, Miller A, Kohn E, Swisher E, et al. A phase III study comparing single-agent olaparib or the combination of cediranib and olaparib to standard platinum-based chemotherapy in recurrent platinum-sensitive ovarian cancer. J Clin Oncol. 2020;38(15). [Google Scholar]
- 86.Lheureux S, Oaknin A, Garg S, Bruce J, Madariaga A, Dhani N, et al. EVOLVE: A Multicenter Open-Label Single-Arm Clinical and Translational Phase II Trial of Cediranib Plus Olaparib for Ovarian Cancer after PARP Inhibition Progression. Clin Cancer Res. 2020;26(16):4206–15. [DOI] [PubMed] [Google Scholar]
- 87.Kumar A, Fernadez-Capetillo O, Carrera AC. Nuclear phosphoinositide 3-kinase β controls double-strand break DNA repair. Proc Natl Acad Sci U S A. 2010;107(16):7491–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Juvekar A, Burga LN, Hu H, Lunsford EP, Ibrahim YH, Rajendran A, et al. Combining a PI3K inhibitor with a PARP inhibitor provides an effective therapy for BRCA1-related breast cancer. Cancer Discov. 2012;2(11):1048–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Rehman FL, Lord CJ, Ashworth A. The promise of combining inhibition of PI3K and PARP as cancer therapy. Cancer Discov. 2012;2(11):982–4. [DOI] [PubMed] [Google Scholar]
- 90.Ibrahim YH, García-García C, Serra V, He L, Torres-Lockhart K, Prat A, et al. PI3K inhibition impairs BRCA1/2 expression and sensitizes BRCA-proficient triple-negative breast cancer to PARP inhibition. Cancer Discov. 2012;2(11):1036–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Matulonis UA, Wulf GM, Barry WT, Birrer M, Westin SN, Farooq S, et al. Phase I dose escalation study of the PI3kinase pathway inhibitor BKM120 and the oral poly (ADP ribose) polymerase (PARP) inhibitor olaparib for the treatment of high-grade serous ovarian and breast cancer. Ann Oncol. 2017;28(3):512–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Konstantinopoulos PA, Barry WT, Birrer M, Westin SN, Cadoo KA, Shapiro GI, et al. Olaparib and α-specific PI3K inhibitor alpelisib for patients with epithelial ovarian cancer: a dose-escalation and dose-expansion phase 1b trial. Lancet Oncol. 2019;20(4):570–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Yap TA, Kristeleit R, Michalarea V, Pettitt SJ, Lim JSJ, Carreira S, 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(10):1528–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Labrie M, Ju Z, Litton J, Kim T, Lee S, et al. Abstract 2070: Exploration of markers of synergistic lethality of PARP and PI3K-Akt-mTOR inhibitors in women’s cancers. Cancer Res. 2019;79. [Google Scholar]
- 95.Hew K, Miller P, El-AShry D, Sun J, Besser A, Ince T, et al. MAPK activation predicts poor outcome and the MEK inhibitor, selumetinib, reverses antiestrogen resistance in high grade serous ovarian cancer. Clin Cancer Res. 2016;22(4):935–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Sun C, Fang Y, Yin J, Chen J, Ju Z, Zhang D, et al. Rational combination therapy with PARP and MEK inhibitors capitalizes on therapeutic liabilities in RAS mutant cancers. 2017;9(392). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Vena F, Jia R, Esfandiari A, Garcia-Gomez JJ, Rodriguez-Justo M, Ma J, et al. MEK inhibition leads to BRCA2 downregulation and sensitization to DNA damaging agents in pancreas and ovarian cancer models. Oncotarget. 2018;9(14):11592–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Sun C, Yin J, Fang Y, Chen J, Jeong KJ, Vellano CP, et al. BRD4 inhibition is synthetic lethal wtih PARP inhibitors through the induction of homologous recombination deficiency. Cancer Cell. 2019;33(3):401–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Zhang B, Lyu J, Yang EJ, Liu Y, Wu C, Pardeshi L, et al. Class I histone deacetylase inhibition is synthetic lethal with BRCA1 deficiency in breast cancer cells. Acta Pharm Sin B [Internet]. 2020;10(4):615–27. Available from: 10.1016/j.apsb.2019.08.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Gupta V, Hirst J, Petersen S, et al. Entinostat, a selective HDAC1/2 inhibitor, potentiates the effects of olaparib in homologous recombination proficient ovarian cancer. Gynecol Oncol. 2021; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Abramson J, Blum K, Flinn I, Gutierrez M, Goy A, et al. BET inhibitor CPI-0610 is well tolerated and induces responses in diffuse large B-cell lymphoma and follicular lymphoma: preliminary analysis of an ongoing phase 1 study. Blood. 2015;126:1491. [Google Scholar]
- 102.Amorim S, Stathis A, Gleeson M, Iyengar S, et al. Bromodomain inhibitor OTX015 in patients with lymphoma or multiple myeloma: a dose-escalation, open-label, pharmacokinetic, phase 1 study. Lancet Haematol. 2016;3(4):196–204. [DOI] [PubMed] [Google Scholar]
- 103.Cochran A, Conery A, Sims R. Bromodomains: a new target class for drug development. Nat Rev Drug Discov. 2019;18(8):609–28. [DOI] [PubMed] [Google Scholar]
- 104.Ameratunga M, Braña I, Bono P, Postel-Vinay S, Plummer R, Aspegren J, et al. First-in-human Phase 1 open label study of the BET inhibitor ODM-207 in patients with selected solid tumours. Br J Cancer [Internet]. 2020;123(12):1730–6. Available from: 10.1038/s41416-020-01077-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Park S, Kim J. A short guide to histone deacetylases including recent progress on class II enzymes. Exp Mol Med. 2020;52(2):204–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
