Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Jan 1.
Published in final edited form as: Cancer J. 2021 Nov-Dec;27(6):465–475. doi: 10.1097/PPO.0000000000000554

PARP Inhibitors in Pancreatic Cancer

Timothy J Brown 1, Kim A Reiss 1
PMCID: PMC8682800  NIHMSID: NIHMS1748347  PMID: 34904809

Abstract

Despite representing only 5% of all annual cancer diagnoses in the United States, pancreatic cancer is projected to become the second-leading cause of cancer-related death within the next ten years. Progress in the treatment of advanced pancreatic cancer has been slow. Systemic therapies rely on combination cytotoxic agents, with limited options at progression. Recently, poly adenosine diphosphate (ADP)-ribose polymerase (PARP) inhibitors have demonstrated clinical activity in patients with advanced pancreatic cancer and pathogenic variants in BRCA1, BRCA2 and PALB2. In this review, we discuss the development of PARP inhibitors in pancreatic cancer, relevant clinical trials, and future directions.

Keywords: Pancreatic Cancer, PARP inhibitors, Homologous Recombination Repair, BRCA, DNA Damage Repair


Pancreatic cancer is an aggressive disease characterized by genomic instability and a high rate of activating mutations in oncogenes and tumor suppressor genes1. Although it accounts for approximately 5% of all cancer diagnoses in the United States, pancreatic cancer is a leading cause of cancer-related mortality and is projected to become the second leading cause of cancer death in the United States by 20302,3. Due to a combination of late symptom onset and aggressive pathophysiology, the majority of pancreas cancer diagnoses are made in the advanced setting. Treatment in such cases is aimed at reducing symptom burden and extending life. Until very recently, the only available systemic options were cytotoxic chemotherapies delivered in a “one-size-fits-all” approach until disease progression, clinical decline or death4,5.

A clearly defined, inherited genetic basis for the development of pancreatic cancer is identified in an estimated 5–15% of those with the disease, with variations in frequency depending on local population demographics6. National Comprehensive Cancer Network (NCCN) guidelines recommend that all patients with a diagnosis of pancreatic cancer undergo germline genetic panel testing, both to identify pathogenic variants that may influence treatment as well as to inform family members who may be offered cascade testing based on positive results7. The development of effective, targeted therapeutic strategies for those with particular genomic variants is an area of active scientific and clinical study.

Indeed, it has been shown in retrospective and prospective studies that platinum chemotherapies such as cisplatin and oxaliplatin are particularly effective against pancreatic cancers that harbor pathogenic variants in BRCA1, BRCA2 and PALB2, with sustained responses lasting months or even years811. However, although maintenance (de-escalation) chemotherapy strategies are being tested and are recommended by the NCCN, perpetual treatment with cytotoxic agents leads to cumulative toxicity in patients, resulting in progressive fatigue and organ dysfunction4,5,7,12.

Over the past decade, poly adenosine diphosphate (ADP)-ribose polymerase (PARP) inhibitors (PARPi) have shown efficacy in solid tumors driven by defects in homologous recombination (HR), particularly in the platinum-sensitive setting1323. PARP inhibitors are oral agents with a manageable toxicity profile. The landmark phase 3 POLO study tested the PARPi olaparib against placebo as maintenance therapy for patients with metastatic, platinum-sensitive pancreatic cancer and a germline BRCA variant24. The study met its primary endpoint of progression-free survival (PFS), demonstrating a median PFS of 7.4 months versus 3.8 months in the experimental and control groups, respectively (hazard ratio 0.54)24. This resulted in the FDA approval of olaparib in the maintenance setting for this group of patients.

The FDA approval of olaparib as maintenance therapy for patients with metastatic pancreatic cancer and germline BRCA mutations represents a step forward in the development of targeted treatments for patients with pancreatic cancer. This preliminary success opens the door for further development of PARP inhibitors in a variety of clinical scenarios and in combinations with other therapies. By the same token, the high variability in efficacy, even in a narrow group of germline BRCA carriers, pushes the scientific community to better identify predictive biomarkers for response to PARP inhibition and to evaluate resistance mechanisms for next generation targeting. In this review, we will explore the development of PARPi in pancreatic cancer, prior key trials of PARPi in pancreatic cancer, and ongoing and future directions for PARPi in pancreatic cancer.

Biological Rationale and Preclinical Evidence for PARP Inhibition in Pancreatic Cancer

Up to 20% of pancreatic cancers possess defects in HR25. Most frequently, these are due to pathogenic variants in BRCA225. Carcinogenesis results when cells containing a pathogenic BRCA mutation lose the remaining BRCA allele, resulting in DNA instability and an accumulation of mutations in other genes, a process termed “loss of heterozygosity”26.The resulting tumors are particularly vulnerable to therapies that cause double stranded DNA breaks, as the tumor cells lack the ability to repair them via the high-fidelity HR pathway.

The PARP enzyme is involved in base-excision repair (BER), a pathway that is critical for fixing single-strand breaks in the DNA, and assists with DNA repair27,28. In HR proficient cells, PARP inhibition results in single strand breaks and collapse of the DNA replication fork that are subsequently repaired via HR27,28. In cells that are homologous recombination deficient (HRD), the double stranded breaks that result from the DNA replication fork collapse cannot be repaired, leading to cumulative DNA damage and, ultimately, cell death27,28. PARP inhibitors (PARPi) have two primary mechanisms of action. First, is catalytic disruption of PARP’s enzymatic activity whereby PARP is no longer able to participate in BER, resulting in an accumulation of single strand breaks (that otherwise would be repaired via HR), chromosomal instability, and apoptosis28. Second, and even more critical, is that PARPi’s increase the affinity of PARP for the DNA strand, promoting DNA-protein crosslinks and thus “trapping” PARP on the DNA29. As a result, DNA replication during S-phase fails, as the replication fork cannot bypass the trapped PARP-DNA complex. The result is an accumulation of strand breaks that ultimately leads to cell death30. While the catalytic activity of most clinically used PARPi is similar, efficacy and toxicity seem linked to the ability of the drug to “trap”29. Veliparib, which does not trap, is the least effective of the class and the PARPi that can be most easily combined with other cytotoxic agents. Rucaparib, olaparib, and niraparib trap at similar levels, while talazoparib has the greatest ability to trap. As a result, talazoparib is the most potent and toxic of this drug class29,31,32.

The observation that PARPi are effective against HRD cells was first demonstrated in Chinese hamster ovary cell lines that were HR deficient27. At low concentrations, the cells were exquisitely sensitive to PARP inhibition. However, when the HR deficiency was reversed, the cells were no longer sensitive to PARP inhibition27. Mice that were xenografted with BRCA2-deficient cancer cells and were treated with PARPi demonstrated less tumor formation compared to BRCA2-proficient counterparts28. Treatment of BRCA-mutated cancer cells with PARPi results in selective tumor toxicity with relative sparing of the normal, heterozygous cells that have intact HR27,28. The concept of selective toxicity of PARP inhibition for BRCA-mutated cells illustrates the concept of “synthetic lethality”, the situation where two deficits have little individual effect on the cellular phenotype but their combination leads to cellular death33,34. This has been illustrated in pre-clinical models - knockout models of Parp1 −/− mice are both viable and fertile, and mice without PARP do not develop early onset tumors compared to those with intact PARP27. However, absence of PARP activity results in increased reliance on HR27. In vitro deletion of BRCA2 in human cell lines results in exquisite sensitivity to PARPi at even low concentrations27. A preclinical model of BRCA-deficient pancreatic cancer showed similar findings; treatment with PARPi reduced cellular viability in vitro and slowed the formation of tumors in vivo35.

Phase 1 Trials of PARP inhibitors in Pancreatic Cancer

On the basis of this preclinical evidence, PARP inhibitors were subsequently trialed in humans. Table 1 lists PARPi’s that have been trialed in patients with pancreatic cancer, including their specific pharmacologic properties and dosing. The initial phase 1 studies allowed enrollment of patients with advanced cancers, with mechanisms to enrich the population for BRCA1 and BRCA2 mutation carriers (Table 2) given the preclinical data that this drug class may be efficacious in the HRD setting.

Table 1.

PARPi that have been trialed in patients with pancreatic cancer. Dosing and common toxicities information obtain from FDA prescribing information. For drugs that are not FDA approved, dosing information was obtained from the most recent clinical trial report.

PARPi Properties60 Dosing Common Toxicities
Olaparib Targets PARP1,2,3 and 4. Interacts with CYP 300 mg BID Fatigue, anemia, thrombocytopenia, neutropenia
Rucaparib^ Targets PARP1,2,3 and 4. Interacts with CYP 600 mg BID Thrombocytopenia, anemia, nausea, fatigue
Veliparib* Weakest PARP trapping abillity 80 mg BID days 1–12 of 21 day cycles Anemia, leukopenia, fatigue
Niraparib^ High CNS penetration, off-target interactions with doapnmine, serotoning, norepinephrine receptors 200–300 mg daily Nausea, thrombocytopenia, anemia, fatigue
Talazoparib^ Most potent PARP-trapping ability, long half-life 1 mg daily Anemia, neutropenia, thrombocytopenia
Fluzoparib* No CNS penetration. Trapping ability not yet defined 150 mg BID Anemia, neutropenia
*-

denotes agent is still investigational and not approved by the US FDA for any cancers.

^ -

denotes drug is not approved by the US FDA for the treatment of pancreatic cancers, but has been approved for the treatment of other cancer types

BID: twice daily

Table 2.

Sumary of completed phase 1 trials involving patients with pancreatic cancer

PARPi Author Design N (N Pancreas) Tumor types included Mutations (n with mutations) Key Findings Ref
Olaparib Fong Phase 1 dose escalation with expansion phase of only patients with BRCA mutations 60 (2) Advanced solid tumors refractory to established therapies (ovarian, breast, colorectal, melanoma, sarcoma, prostate, other) Not required in the initial cohort, expansion cohort enrolled only patients with BRCA1/2 mutations (n=22) MTD 400 mg BID
Sufficient inhibition of PARP achieved at olaparib 60 mg BID
Fatigue and nausea were common side effects.
9 of 19 evaluable patients with BRCA mutations had a response
36
Rucaparib Plummer Phase 1 dose escalation in combination with temozolomide 100 mg/m2 escalating to 200 mg/m2 daily days 1–5 of 28 day cycles 32 (1) Advanced solid tumors (sarcoma, melanoma, colorectal, other) Not reported RP2D: rucaparib 12 mg/m2 and temozolomide 200 mg/m2 days 1–5 of 28 day cycles.
Grade 3 neutropenia occurred in high doses
One patient with pancreatic cancer experienced prolonged disease stabilization
40
Kristeleit Phase 1 3+3 dose escalation 56 (2) Ovarian cancer and other solid tumors All patients had germline BRCA1/2 mutations RP2D: 600 mg BID
Fatigue, myelosuppression. Gastrointestinal upset were common but manageable.
One of the two patients with pancreatic cancer experienced a prolonged response
30
Niraparib Sandhu Phase 1 dose escalation 100 (1) Advanced solid tumors With (29) or without BRCA mutations MTD and RP2D: 300 mg/day
Anemia, thrombocytopenia, neutropenia, fatigue were common.
Single patient with pancreatic cancer possessed BRCA2 mutation and did not respond to treatment
37
Veliparib O’Reilly Single arm phase 1 dose escalation study with gemcitabine 600 mg/m2 day 3 and 10 and cisplatin 25 mg/m2 days 3 and 10 17 (17) Advanced or metastatic pancreatic cancer and were naïve to platinum-based therapies BRCA mutations (9) or family history of BRCA-related cancers
RP2D: 80 mg BID 11ay 1–12 of 21 day cycles in combination with cisplatin and gemcitabine
Grade 4 neutropenia and thrombocytopenia were common
Objective responses only seen in patients with BRCA mutations (7 of 9 patients)
One patient developed acute myeloid leukemia 2.5 years into therapy
42
Tuli Single arm phase 1 combining veliparib with gemcitabine and radiation 30 (30) Treatment-naïve locally advanced pancreatic cancer or borderline resectable pancreatic cancer No patient had BRCA1 or BRCA2 mutations
Nine patients with mutations in other DNA damage repair genes (ARID1A, ATM, CHECK2, PALB2, PTEN, and loss of MLH1).
MTD: 40 mg BID
Lymphopenia and Anemia were common Grade 3 Aes
Slightly longer median OS in patients with mutations in DDR
43
Talazoparib de Bono Dose escalation followed by expansion cohort 113 (13) Tumors predicted to be potentially sensitive to PARPi, including germline BRCA1/2 mutations With (59) or without germline BRCA mutations 4.0 mg/day RP2D
Marrow toxicity common
2/13 pancreatic cancer patients had a PR (one with BRCA2 mutation, one with PALB2 mutation)
39
Fluzoparib Li Phase 1 3+3 dose escalation 79 (2) Advanced solid tumors With (59) or without BRCA mutations MTD: 150 mg BID
Antitumor responses seen in platinum resistant and platinum-refractory BRCA mutated cancers
38

RP2D: Recommended phase 2 dose, MTD: Maximum tolerated dose, BID: twice daily, AE: adverse event DDR: DNA Damage Repair, PR: partial response.

Olaparib, rucaparib, niraparib, talazoparib, and fluzoparib have all completed monotherapy phase 1 clinical trials that included patients with pancreatic cancer. In these toxicity trials, it was strikingly obvious that patients with germline BRCA variants were more likely to benefit from PARPi treatment compared to patients with sporadic cancers30,3639. For example, in the phase 1 study of olaparib, 12 of 19 evaluable patients with germline BRCA mutations had either disease response or stabilization, while no patients with sporadic disease had such a result36.

The observation that patients with HRDs had superior outcomes to those with sporadic cancers extended to patients with pancreatic cancer. In the phase 1/2 trial of rucaparib, one of two patients with BRCA-related pancreatic cancer experienced a prolonged partial response30. Similarly, of 13 pancreatic cancer patients enrolled in the phase 1 trial of talazoparib, two had partial responses to therapy: one with a BRCA2 variant, the other with a PALB2 variant39. These findings provided support to pursue PARPi therapy specifically in HRD pancreatic cancer.

Several notable phase 1 combination studies with PARPi have been completed. First, a phase 1 study of rucaparib in combination with temozolomide was performed following pre-clinical evidence of potentiation of alkylators by PARPi40,41. A total of 32 patients were treated, including one patient with pancreatic cancer. The combination was deemed to be safe and adequate PARP inhibition could be achieved, although myelosuppression was noted at lower-than-expected doses of temozolomide. The single patient with pancreatic cancer enrolled on the study experienced disease stability for more than six months40. This combination has not been pursued further so far and there are no active clinical trials of this combination on clinicaltrials.gov (as of August 9, 2021).

Second, a phase 1 study of veliparib in combination with cisplatin and gemcitabine was performed in patients with advanced pancreatic cancer and germline BRCA mutations or a family history of BRCA-related cancers42. Nine patients with BRCA mutations were enrolled and seven patients without BRCA mutations were enrolled. Seven patients on the study (all with BRCA mutations) had responses, six partial responses and one complete response. The patient with a complete response developed acute myeloid leukemia 2.5 years into therapy that was potentially related to the treatment. An additional eight patients with BRCA mutations had stable disease. No responses were seen in patients without BRCA mutations42. Given the promising signals of efficacy, this combination was then tested in a randomized phase 2 trial, which will be discussed later in this review10.

Finally, a phase 1 study investigated the use of veliparib in combination with gemcitabine and radiotherapy for patients with locally advanced pancreatic cancer43. This single-institution trial enrolled patients with treatment-naïve locally advanced pancreatic cancer or borderline resectable pancreatic cancer. Thirty patients were enrolled, however none possessed BRCA mutations. Nine patients had tumors with mutations in other DNA damage repair genes (ARID1A, ATM, CHEK2, PALB2, PTEN, and loss of MLH1). The median OS seen in patients treated with this regimen was 14.6 months. A slightly longer median OS of 19 months was observed for patients with defects in DNA damage repair pathways, although this was not statistically significant43.

Based on the findings that PARPi therapy was well tolerated and demonstrated efficacy in patients with DNA damage repair alterations, a number of phase 2 and 3 trials were launched. Most prominent were trials in the canonical tumor types associated with BRCA, namely: breast, ovarian, prostate and pancreatic cancer.

Phase 2 Trials

Multiple phase 2 trials have investigated the efficacy of PARPi in pancreatic cancer (Table 3). The first of these was a multicenter phase 2 study that enrolled patients with pathogenic germline BRCA1 or BRCA2 mutations and recurrent solid tumors to receive olaparib monotherapy44. The primary endpoint was tumor response rate44. A total of 298 patients were enrolled, of whom 23 had advanced pancreatic cancer. The tumor response rate for patients with pancreatic cancer was 21.7% (95% CI 7.5–43.7%), although 65% had received prior platinum therapy. One patient had a complete response, and four patients had partial responses. The median duration of response for patients with pancreatic cancer was 4.4 months, the median PFS was 4.6 months, and the median OS was 9.8 months. Proportionally, more patients with pancreatic cancer who were not previously exposed to platinum chemotherapy had a response (two of eight patients without prior platinum use versus three of fifteen patients with prior platinum use)44. This study provided adequate rationale to continue the development of PARP inhibitors in BRCA-related pancreatic cancer.

Table 3.

Summary of published phase 2 and 3 trials of PARPi in pancreatic cancer.

Phase 2 trials
Author (Trial name) Design Stage Setting Drug Key Findings Ref
Kaufman Non-randomized phase 2 Advanced or metastatic
  • Progression after gemcitabine

  • Germline BRCA1 or BRCA2

Olaparib ORR: 21.7%
DOR: 134 days
OS: 18.4 mos
PFS: 4.6 mos
44
Shroff (RUCAPANC) Non-randomized phase 2 Locally advanced or metastatic
  • Progression after 1–2 lines of chemotherapy

  • Germline or somatic BRCA1 or BRCA2

Rucaparib ORR: 15.8%
DCR: 31.6%
45
Lowery Non-randomized phase 2 Locally advanced or Metastatic
  • Progression.

  • Germline BRCA1, BRCA2, or PALB2

Veliparib ORR: 0%
mPFS: 1.7 mo
mOS: 3.1 mo
47
O’Reilly Randomized phase 2 Locally advanced or metastatic
  • First Line

  • Germline BRCA1, BRCA2 or PALB2

Gem/Cis +/− Veliparib (veliparib vs without)
ORR: 74.1% vs 65.2% (p=0.55)
DCR: 100% vs 78.3% (p=0.02)
mOS: 15.5 mos vs 16.4 mos (p=0.6)
mPFS: 10.1 mos vs 9.7 mos (p=0.73)
G3–4 heme toxicities: 48% vs 30%
10
Javle Two parallel non-randomized phase 2 Metastatic
  • Second or Third Line DNA Damage Repair deficiencies other than BRCA mutations

Olaparib ORR:
mOS: 9.9 mos
mPFS: 3.7 mos
48
Reiss Non-randomized phase 2 Unresectable or metastatic Maintenance
  • Germline or somatic BRCA1 or BRCA2

Rucaparib mPFS: 13.1 mos
mOS: 23.5 mos
DCR: 66.7%
DOR: 17.3 mos
ORR: 41.7%
46
Phase 3 trials
Author (Trial Name) Design Stage Setting Drug Outcomes Ref
Golan (POLO) Randomized phase 3 Metastatic
  • Maintenance

  • Germline BRCA1 or BRCA2

Olaparib vs placebo mPFS 7.4 mo vs 3.8 mo (HR 0.43 95%CI 0.35–0.82, p=0.004)
mOS 18.9mo vs 18.1 mo (HR 0.91 95% CI 0.56–1.46, p=0.68)
HRQOL between group difference −2.47 (95%CI −7.27 to 2.33) on 100-point scale
24,49,50

mPFS = median progression-free survival, OS= median overall survival, HRQOL= health-related quality of life 95%CI = 95% Confidence Interval, ORR = objective response rate, DCR = disease control rate, G3–4 Heme toxicities = rates of grade 3 to grade 4 hematologic toxicity, gem= gemcitabine, cis= cisplatin. POLO HRQOL was assessed with the EORTC QLQ-C30 global health-related quality-of-life score. Javle study accepted DNA damage repair deficiencies: somatic BRCA variant, somatic or germline: ATM, PALB2, CHEK1, FANCA, BARD1, RAD50, and ARID1A.

RUCAPANC was a single arm phase 2 study testing monotherapy rucaparib in patients with locally advanced or metastatic pancreatic cancer and pathogenic germline or somatic BRCA1 or BRCA2 mutations after progression on at least one prior line of chemotherapy45. The primary endpoint was objective response rate. A total of nineteen patients received at least one dose of rucaparib. The confirmed objective response rate was 15.8%, with two confirmed partial responses, one confirmed complete response, and one unconfirmed complete response45. Notably, responses were only observed in platinum-sensitive patients and two of the responses were seen in patients with somatic BRCA2 variants45.

Based on these observations, a single arm phase 2 maintenance trial of rucaparib in patients with somatic or germline variants in BRCA or PALB2, advanced pancreatic cancer and platinum sensitive disease was performed46. The primary endpoint was PFS. A majority of the 42 evaluable patients had received at least 16 weeks of platinum-based chemotherapy for locally advanced or metastatic disease without evidence of platinum resistance; eight patients received fewer than 16 weeks due to either intolerance or allergy to platinum. The median PFS was 13.1 months, with a six-month PFS of 59.5% and a 12-month PFS of 54.8%. The median OS was 23.5 months with eight patients alive more than two years after enrollment. Importantly, of the six patients with germline PALB2 variants, three had responses, including one who had a CR. Of the two patients with somatic BRCA2 variants, one had a prolonged partial response. A post-hoc analysis of patients who received at least 16 weeks of platinum-based chemotherapy compared to the eight patients who received fewer than 16 weeks of chemotherapy showed no difference in PFS or OS, raising the question whether a full four months of platinum-based chemotherapy prior to starting maintenance PARPi therapy is required. Although limited in sample size, patients with BRCA1 pathogenic variants and those with higher disease burden at study start responded less favorably to maintenance PARPi46. Overall, this trial added credence to using PARPi for patients with germline PALB2 variants and somatic BRCA variants, expanding the group of patients for whom this therapy might be applied.

Veliparib was tested in a single-arm phase 2 trial of patients with previously treated stage III or IV pancreatic cancer with known germline BRCA1, BRCA2 or PALB2 mutations to determine the response rate, duration of response, PFS, and OS47. A total of 16 patients were enrolled, with 14 having had prior exposure to platinum-based therapy. The study was closed early due to insufficient activity. No confirmed responses were seen in this trial, although one patient who had not been previously exposed to platinum chemotherapy had an unconfirmed partial response. The best observed response was stable disease in five patients. The median PFS was 1.7 months and the median OS was 3.1 months47. Veliparib’s inability to PARP trap and the high proportion of patients with platinum-resistant cancer likely contributed to these disappointing results.

Based on phase 1 data showing tolerance and efficacy of veliparib with platinum-based chemotherapy in patients with BRCA-related pancreatic cancer, O’Reilly et al performed a randomized, multicenter, phase 2 trial investigating the combination of gemcitabine and cisplatin with or without veliparib in patients with advanced, previously untreated pancreatic cancer and a germline BRCA or PALB2 mutation10. Fifty patients were randomized 1:1 to receive cisplatin 25mg/m2 with gemcitabine 600 mg/m2 on days 3 and 10 with or without veliparib 80 mg twice daily on days 1–12 of each 21-day cycle. The primary endpoint was objective response rate (ORR). PFS, OS, safety, and disease control rate (DCR) were secondary endpoints. Twelve (24%) of the enrolled patients had BRCA1 mutations, thirty-five (70%) had BRCA2 mutations, and three (6%) had PALB2 mutations. There was no significant difference in the ORR (74.1% vs 65.2%; p = 0.55), PFS (10.1mo vs 9.7mo) or OS (15.5 mo vs 16.4 mo) between groups. Myelosuppression requiring dose reductions was more common in the experimental group10. Importantly, although this trial was negative, it demonstrated an unprecedented ORR to first line palliative cisplatin plus gemcitabine in patients with BRCA and PALB2 variants. This regimen is now considered a standard option for this patient population.

A major area of scientific interest is to expand upon the group of patients for whom PARPi therapy might be used. In an effort to explore this, two parallel phase 2 trials enrolled patients with advanced pancreatic cancer and either: (1) non-BRCA HRD variants, (2) a personal or family history of BRCA-related cancers or (3) ATM loss by IHC48. A total of 46 patients were enrolled and the primary endpoint was ORR by RECIST 1.1. Thirty-four patients were platinum-sensitive, two were platinum-naïve, and the remainder were platinum-refractory. Two patients in this combined study had a partial response and thirty-three had stable disease, with a median disease control rate of 2.9 months48. The median PFS was 3.7 months (consistent across both studies) and the median OS was 9.9 months. Patients with platinum-sensitive cancers experienced longer median PFS and OS (4.1 vs 2.2 months and 10.5 vs 5.4 months, respectively). Lastly, patients with mutations in DNA damage repair genes had longer PFS than those with a family history of BRCA-related tumors or those with ATM loss (5.7 mos [95% CI 3.6–8.8 mos] vs 2.6 mos [1.9–3.9 mos]), although this association was not formally tested48. The somewhat disappointing results of this study highlight that our ability to identify patients for PARPi therapy remains crude and that functional assays for DNA damage repair deficiency are needed.

Phase 3 Trials

The POLO study is the sole completed phase 3 trial of a PARPi in pancreatic cancer. In this randomized, double-blind study, patients with metastatic disease and germline BRCA1 or BRCA2 mutations who had been treated with at least four months of platinum-based chemotherapy without progression were randomized 3:2 to receive maintenance olaparib or placebo. Patients who received olaparib experienced a longer median PFS compared to placebo (7.4 months vs 3.8 months; HR 0.54) while preserving quality of life24,49. Although this trial met the primary endpoint of improved PFS, there was no difference in median overall survival with olaparib compared to placebo (19.0 months vs 19.2 months, respectively)50. On the basis of these results, olaparib was approved by the United States Food and Drug Administration (FDA) for maintenance treatment of adult patients with BRCA-mutated metastatic pancreatic adenocarcinoma whose disease has not progressed after at least 16 weeks of platinum-based chemotherapy51. The approval has made olaparib a therapeutic option in clinical practice for these patients.

An ongoing phase 3 trial of fluzoparib in patients with metastatic pancreatic cancer with germline BRCA1/2 or PALB2.mutations that has not progressed on platinum-based chemotherapy hopes to further build on the success of POLO in moving PARPi into the maintenance space for patients with pancreatic cancer (NCT04300114).

Resistance to PARP Inhibition

Primary Resistance

Despite a strong biological rationale, not all patients with BRCA mutations or DNA damage repair gene alterations respond to treatment with PARPi. For example, roughly 20% of patients receiving olaparib on the POLO study progressed by the first planned assessment, despite entering the study with clinical platinum sensitivity24. In patients with pathogenic germline BRCA or PALB2 mutations treated with rucaparib, approximately 16% progressed at first assessment46. This implies that even in the setting of platinum sensitivity, some HRD tumors are primarily resistant to PARPi treatment. In a series of cases of pancreatic cancer with pathogenic germline BRCA mutations, 12% of tested samples (6/49) retained the wild-type allele and were found to be homologous recombination proficient52. Consistent with clinical experience, this finding suggests that in some cases, a patient with a germline BRCA variant and pancreatic cancer would not be expected to respond favorably to targeted therapy, such as with PARPi.

Adaptive Resistance

Secondary resistance to PARP inhibitors develops almost inevitably in the palliative setting. Identifying mechanisms of the development of resistance to PARP inhibition is an area of focus, particularly as the development of successful second generation therapies will likely depend on understanding the key mechanisms of PARPi failure.

One described mechanism of adaptive resistance is the restoration of homologous repair by reversion mutations following treatment53. A recently published study collated all published literature on reversion mutations and characterized 300 reversion mutations in 91 patients that resulted in PARPi resistance53. Other purported mechanisms of resistance include mutations or downregulation in PARP1, increased drug efflux via p-glycoprotein, independent restoration of homologous repair (such as with loss of 53BP1, REV7, RIF1, and shielden), and restoration of replication fork stability (via loss of SLFN11, RADX or SMARCAL1 depletion) among others5462,6267. Strategies to overcome PARPi resistance with combination therapies are currently under development and are discussed later in this review.

Prospectively predicting which patients with HR mutations will benefit from PARP inhibition will be crucial to advancing the field and assays for HR are in development. Currently in clinical practice, HRD is assessed using genomic methods such as next generation sequencing specifically analyzing BRCA1, BRCA2, PALB2, ATM, CHEK2, and FANC68,69. In the context of clinical trials, HRD is usually determined by whole genome sequencing, whole exome sequencing, targeted next generation sequencing, or deep exome sequencing68. Other assays remain investigational but are promising due to their high sensitivity and specificity. Substitution base signature 3 (SBS3; indels>3 base pairs in length) is strongly associated with HRD, however requires whole genome sequencing to perform7072. Myriad’s MyChoice HRD assay performs whole exome sequencing to calculate a genomic instability score (GIS) that correlates with HRD52,7375. Although this test is FDA approved in gynecological malignancies and requires formalin-fixed paraffin-embedded tissue to calculate the GIS; its use in pancreatic cancer may be challenging due to limited cellularity of tumor specimens13,52,74,75. In a series of pancreatic cancer patients with germline BRCA mutations described above, the authors used the HRDetect score, a combination score derived from mutational signatures52. HRDetect was found in this series to have 100% sensitivity and 98% specificity in identifying HRD pancreatic cancer, however it requires fresh frozen tissue for whole genome sequencing52. Measurement and detection of RAD51 foci by immunofluorescence or imunhistochemistry may serve as a marker for intact HR69. Early clinical evidence suggests this functional assay may correlate to sensitivity to PARPi or platinum65. A DNA fiber assay is also in development that seeks to measure stalled replication fork protein stability to correlate with resistance or sensitivity to PARPi or platinum69,76. Further refinement and technological availability is needed prior to these assays being employed clinically to fully realize the potential efficacy of PARP inhibition for a given patient.

Future directions

There are several ongoing trials to further define the role of PARPi in pancreatic cancer (Table 4). These include studies that move PARPi into the earlier disease setting and those combining PARPi with other agents based on synergy observed in preclinical models.

Table 4.

Ongoing phase 2 or 3 studies of PARPi in pancreatic cancer. Status as of July 21, 2021. Trials may be open to other tumor types, visit clinicaltrials.gov for the latest information.

PARPi ClinicalTrials.gov Identifier (Name) Trial Phase Setting Drugs Status
Olaparib NCT04666740
(POLAR)
Non-randomized phase 2 Maintenance after first or second-line therapy for metastatic disease.
Germline mutations in homologous recombination (BRCA 1, BRCA2, PALB2, ATM, BAP1, BARD1, BLM, BRIP1, CHEK2, FAM175A, FANCA, FANCC, NBN, RAD50, RAD51, RAD51C, RTEL1)
Platinum sensitive or resistant.
Pembrolizumab + Olaparib Recruiting
NCT04548752 Randomized phase 2 Maintenance therapy in metastatic disease with germline BRCA1 or BRCA2 mutations after first line platinum-based chemotherapy Pembrolizumab + Olaparib
Olaparib monotherapy
Recruiting
NCT02677038 Non-Randomized phase 2 Metastatic and with mutations in homologous recombination (e.g. somatic BRCA mutation, FANC1, ATM or RAD51 mutations) or in patients with close family members with BRCA-driven cancers
Negative for germline BRCA1 and BRCA2 mutations
After at least one prior line of chemotherapy for metastatic disease and not have platinum-refractory cancer
Olaparib monotherapy Active, not recruiting
NCT04858334
(APOLLO)
Randomized phase 2 Adjuvant therapy following completion of surgery and chemotherapy
Germline or somatic BRCA1, BRCA2 or PALB2 mutation
Olaparib monotherapy
Placebo
Recruiting
NCT02498613 Non-randomized phase 2 Maintenance therapy in advanced and metastatic pancreatic cancer Cediranib + Olaparib Recruiting
NCT03682289 Non-randomized phase 2 Maintenance therapy in locally advanced or metastatic pancreatic cancer following at least one previous line of therapy AZD6738 + Olaparib
AZD6738
Recruiting
Rucaparib NCT04171700
(LODESTAR)
Non-randomized phase 2 Maintenance therapy in unresectable, locally advanced, or metastatic solid tumors with deleterious mutations in BRCA1, BRCA2, PALB2, RAD51C or RAD51D. An expansion cohort is planned for patients with deleterious mutations in BARD1, BRIP1, FANCA, NBN, RAD51 or RAD51B.
Following at least one but not more than three prior lines of systemic therapy
Rucaparib monotherapy Recruiting
Niraparib NCT03601923 Non-randomized phase 2 Maintenance therapy in metastatic or locally advanced pancreatic cancer with germline or somatic mutations in BRCA1, BRCA2, PALB2, CHEK2 or ATM.
No progression on prior oxaliplatin-containing regimens
Niraparib monotherapy Recruiting
NCT04409002 Non-randomized phase 2 Metastatic pancreatic cancer Niraparib + Dostarlimab + Radiation Recruiting
NCT03553004
(NIRA-PANC)
Non-randomized phase 2 Metastatic pancreatic cancer with germline or somatic mutations in genes involved in DNA repair following progression after 1–2 lines of chemotherapy Niraparib Monotherapy Recruiting
NCT04493060 Non-randomized phase 2 Metastatic pancreatic cancer with germline or somatic BRCA1, BRCA2, or PALB2 mutations.
Following progression on 1–2 prior lines of chemotherapy for metastatic disease, with prior exposure to a platinum agent.
Niraparib + Dostarlimab Recruiting
NCT03404960
(PARPVAX)
Randomized Phase 1b/2 Maintenance therapy for locally advanced or metastatic pancreatic cancer with stable disease after at least 16 weeks of platinum-based treatment Niraparib + Nivolumab
Niraparib + Ipilimumab
Recruiting
Talazoparib No identified phase 2 or 3 trials
Veliparib NCT02890355 Randomized phase 2 Metastatic pancreatic cancer with recurrence following one prior systemic regimen
BRCA1, BRCA2, or other defects in homologous recombination repair
Veliparib + mFOlFIRI
FOLFIRI
Active, Not Recruiting
NCT01585805 Randomized phase 2 Locally advanced or metastatic pancreatic cancer with BRCA1, BRCA2, or PALB2 mutation.
No prior treatment allowed for Arms A and B (GCV vs GC), up to two prior treatments allowed for Arm C (single agent veliparib)
Gemcitabine + Cisplatin and veliparib
Gemcitabine + cisplatin
Veliparib monotherapy
Active, not recruiting
Fluzoparib NCT04300114 Randomized Double-Blind Phase 3 Maintenance therapy in patients with BRCA1/2 or PALB2-mutated pancreatic cancer that has not progressed on first-line platinum-based chemotherapy Fluzoparib
Placebo
Recruiting

GCV = Gemcitabine, cisplatin, veliparib. GC = Gemcitabine, Cisplatin, mFOLFIRI = modified fluorouracil, leucovorin, irinotecan.

Curative Intent

The APOLLO study (ECOG-ACRIN 2192; NCT04858334) is a randomized phase 2 double blind study of adjuvant olaparib vs placebo for one year in patients with germline or somatic pathogenic mutations in BRCA or PALB2 who have completed all curative intent treatment77. Patients who are within eight weeks of completing all standard therapy are randomized 2:1 to receive olaparib or a placebo pill. The primary endpoint is PFS. Information gleaned from this trial will further inform the use of adjuvant PARPi, a strategy that was recently shown to be effective in improving invasive disease-free survival in early stage, high-risk, BRCA-mutated breast cancer78.

PARPi plus Immune Checkpoint Blockade

Combining PARPi with immunotherapy is a novel concept that is under investigation. The addition of PARPi to HRD cells results in cytosolic accumulation of DNA and leads to activation of the stimulator of interferon genes (STING) pathway. In turn, this triggers an inflammatory cascade that leads to enhanced tumor infiltration by lymphocytes and increased PDL1 expression. These observations provide an intriguing biological rationale for combining PARPi with immunotherapy and several studies are currently exploring this strategy7982. The phase 1b/2 randomized PARPVAX study is enrolling patients with pancreatic cancer that have not progressed on platinum chemotherapy to receive niraparib with either ipilimumab or nivolumab (NCT03404960). The POLAR study, another non-randomized phase 2 trial, is testing the combination of pembrolizumab with olaparib in patients with metastatic pancreatic cancer and HRR deficiency in the maintenance setting (NCT04666740). The Southwest Oncology Group is running a similar trial, testing olaparib with or without pembrolizumab as maintenance therapy in patients with metastatic pancreatic cancer and germline BRCA1/2 mutations (NCT04548752). If successful, these trials will open a potentially new combination of historically well-tolerated treatments to patients with metastatic pancreatic cancer and will further the development of maintenance combination therapies, both for patients with HRD and those without.

PARPi plus Other Agents

Several combinations with PARPi are in clinical development based on preclinical data showing synergy in the HRD and non-HRD settings. For example, preclinical data have shown that treatment of pancreatic cancer cells with bromodomain and extraterminal (BET) inhibitors results in cellular sensitivity to PARP inhibition by blunting DDR signaling and reducing transcription of BRCA1 and RAD5183,84. Together, this produces a phenotype similar to the BRCA-mutated pancreatic cancers. A first in-human study testing this combination has been completed, however results have not yet been published (NCT03205176). Combining PARPi with vascular endothelial growth factor inhibitor bevacizumab is also of interest and has been approved for use in ovarian cancer following the results of the phase 3 PAOLA-1 trial demonstrating a 6 month median PFS improvement in patients with platinum-sensitive advanced ovarian cancer randomized to receive olaparin-bevacizumab compared to placebo-bevacizumab (22.1 vs 16.6 mos)13. It is suspected that the anti-tumor activity is derived from the relative tissue hypoxia produced by inhibiting angiogenesis. This results in decreased transcription of homologous recombination repair genes and may increase the efficacy of concurrent PARP inhibition8587. Clinical trials exploring this combination are underway in other cancer types88. A third combination that shows synergy in preclinical studies is PARPi plus inhibitors of ataxia telangiectasia and Rad3-related protein (ATR). This combination was first hypothesized to be effective following the observation that lung cancer cells lacking ATM were more sensitive to PARPi than expected89. Coinhibitory studies on biliary tract cancers support a synergistic antitumor effect from combination PARPi and ATR inhibition and early phase clinical trials are underway (NCT04298021)90. Another combination of interest is PARPi and AKT inhibition following preclinical evidence showing inhibition of the PI3K-AKT pathway results in suppression of BRCA1, promoting HR deficiency and PARPi sensitivity91. A recent phase 1 trial of PARP inhibition with olaparib with the AKT inhibitor capivasertib in advanced solid tumors recently showed preliminary signs of anti-tumor activity, particularly in BRCA-mutated cancers and even in patients with prior PARPi exposure92.

Because primary and secondary resistance to PARPi remain a major hindrance for the clinical use of PARPi, rational mechanisms for overcoming resistance are needed. Co-inhibition of RAD52 and PARP may be a mechanism for suppressing alternative HR pathways. Pre-clinical data suggest a synergy between RAD52 inhibitors and PARPi in BRCA-deficient tumors, introducing the concept of “dual synthetic lethality”60,93. Altering cellular energy metabolism may also have a role in overcoming PARPi resistance. Preclinical inhibition of nicotinamide phosphoribosyltransferase (PAMPT) in combination with olaparib results in cellular depletion of NAD+ and slows the growth of tumors in a triple negative breast cancer model60,94. Suppression of microhomology-mediated end-joining via POL Ø inhibition may also prevent the emergence of PARPi resistance by preventing the hypermutating phenotype that results from reliance on microhomology-mediated end-joining95. In HR deficient cells, POL Ø deficiency is also synthetically lethal and co-inhibition with PARP inhibition results in effective cell killing9597.

Beyond BRCA

Finally, the study and treatment of patients with DDR alterations beyond those with BRCA and PALB2 variants is of tremendous scientific interest, as this would expand the group of patients for whom PARPi might be used. Mounting data suggests that selecting therapies based solely on genotype is an insufficient strategy, as (1) not all patients with BRCA or PALB2 mutations are truly DDR deficient, (2) patients without mutations may be DDR deficient when tested by functional assays and (3) there remains relatively little clarity around which variants outside the core DDR genes truly result in a PARPi responsive phenotype52. Highlighting this last point, when evaluated by the whole genome sequencing assay HRDetect, pancreatic tissue samples from patients with ATM or CHEK2 variants do not appear to have an HRD, which would suggest that PARP inhibitors might not be an effective strategy for this group52. To date, multiple PARPi clinical trials have enrolled patients with one of a variety of mutations (Tables 24), but this “lumping” strategy is likely to dilute efficacy signals due to variability in HRD among variants. Therefore, there is substantial interest in developing clinically useful functional assays to more precisely identify the right patients for PARP inhibitor treatment.

Conclusions

Locally advanced and metastatic pancreatic cancer continues to have a poor prognosis. However, the recognition that a portion of patients with this disease possesses a uniquely targetable biology has changed the landscape of treatment and scientific research. PARP inhibitors are a molecularly targeted therapy that are well tolerated with sensitivity noted in patients with tumors possessing BRCA1 and BRCA2 mutations. The success of olaparib in the monotherapy setting for patients with metastatic disease has made this class of drugs an option for patients in clinical practice and has opened the doors for additional research that is focused on moving these agents into the curative intent setting, more precisely identifying and expanding the population for which they could be used, identifying and thwarting resistance mechanisms, and improving efficacy with combination therapies.

Funding:

NIH T32CA009679 (TJB)

Disclosures:

TJB: none. KAR: Research funding from Clovis Oncology, Bristol-Myers-Squibb, GlaxoSmithKline.

References

  • 1.Mizrahi JD, Surana R, Valle JW, Shroff RT. Pancreatic cancer. The Lancet Elsevier Ltd, 2020;395:2008–2020. [DOI] [PubMed] [Google Scholar]
  • 2.Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the united states. Cancer Research. 2014. [DOI] [PubMed] [Google Scholar]
  • 3.Noone A, Howlader N, Krapcho M, Miller D, Brest A, Yu M, et al. SEER Cancer Statistics Review (CSR) 1975–2015. SEER Cancer Statistics Review, 1975–2015, National Cancer Institute based on November 2017 SEER data submission, posted to the SEER web site, April 2018. Available from: https://seer.cancer.gov/csr/1975_2015/.
  • 4.Conroy T, Desseigne F, Ychou M, Bouché O, Guimbaud R, Bécouarn Y, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. The New England journal of medicine 2011;364:1817–1825. [DOI] [PubMed] [Google Scholar]
  • 5.Von Hoff DD, Ervin T, Arena FP, Chiorean EG, Infante J, Moore M, et al. Increased Survival in Pancreatic Cancer with nab-Paclitaxel plus Gemcitabine. New England Journal of Medicine 2013; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Solomon S, Das S, Brand R, Whitcomb DC. Inherited pancreatic cancer syndromes. Cancer Journal (United States). 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Pancreatic Adenocarcinoma - Version 2.2021. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). Available from: https://www.nccn.org/professionals/physician_gls/pdf/pancreatic.pdf. [DOI] [PubMed]
  • 8.Yu S, Agarwal P, Mamtani R, Symecko H, Spielman K, O’Hara M, et al. Retrospective Survival Analysis of Patients With Resected Pancreatic Ductal Adenocarcinoma and a Germline BRCA or PALB2 Mutation . JCO Precision Oncology 2019;1–11. [DOI] [PubMed] [Google Scholar]
  • 9.Wattenberg MM, Asch D, Yu S, O’Dwyer PJ, Domchek SM, Nathanson KL, et al. Platinum response characteristics of patients with pancreatic ductal adenocarcinoma and a germline BRCA1, BRCA2 or PALB2 mutation. British Journal of Cancer Springer US, 2020;122:333–339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.O’Reilly EM, Lee JW, Zalupski M, Capanu M, Park J, Golan T, et al. Randomized, multicenter, phase II trial of gemcitabine and cisplatin with or without veliparib in patients with pancreas adenocarcinoma and a germline BRCA/ PALB2 mutation. Journal of Clinical Oncology 2020;38:1378–1388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Golan T, Kanji ZS, Epelbaum R, Devaud N, Dagan E, Holter S, et al. Overall survival and clinical characteristics of pancreatic cancer in BRCA mutation carriers. British journal of cancer 2014; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Chevalier H, Vienot A, Lièvre A, Edeline J, El Hajbi F, Peugniez C, et al. FOLFIRINOX De‐Escalation in Advanced Pancreatic Cancer: A Multicenter Real‐Life Study. The Oncologist 2020;25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.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. New England Journal of Medicine 2019;381:2416–2428. [DOI] [PubMed] [Google Scholar]
  • 14.Mateo J, Carreira S, Sandhu S, Miranda S, Mossop H, Perez-Lopez R, et al. DNA-Repair Defects and Olaparib in Metastatic Prostate Cancer. New England Journal of Medicine 2015;373:1697–1708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hussain M, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, et al. Survival with Olaparib in Metastatic Castration-Resistant Prostate Cancer. New England Journal of Medicine 2020;383:2345–2357. [DOI] [PubMed] [Google Scholar]
  • 16.Robson M, Im S-A, Senkus E, Xu B, Domchek SM, Masuda N, et al. Olaparib for Metastatic Breast Cancer in Patients with a Germline BRCA Mutation. New England Journal of Medicine 2017;377:523–533. [DOI] [PubMed] [Google Scholar]
  • 17.Penson RT, Valencia RV, Cibula D, Colombo N, Leath CA, Bidzinski M, et al. Olaparib versus nonplatinum chemotherapy in patients with platinum-sensitive relapsed ovarian cancer and a germline BRCA1/2 mutation (SOLO3): A randomized phase III trial. Journal of Clinical Oncology 2020;38:1164–1174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Mirza MR, Monk BJ, Herrstedt J, Oza AM, Mahner S, Redondo A, et al. Niraparib Maintenance Therapy in Platinum-Sensitive, Recurrent Ovarian Cancer. New England Journal of Medicine 2016;375:2154–2164. [DOI] [PubMed] [Google Scholar]
  • 19.Del Campo JM, Matulonis UA, Malander S, Provencher D, Mahner S, Follana P, et al. Niraparib maintenance therapy in patients with recurrent ovarian cancer after a partial response to the last platinum-based chemotherapy in the ENGOT-OV16/NOVA trial. Journal of Clinical Oncology 2019;37:2968–2973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Coleman RL, Oza AM, Lorusso D, Aghajanian C, Oaknin A, Dean A, et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet 2017;390:1949–1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Litton JK, Rugo HS, Ettl J, Hurvitz SA, Gonçalves A, Lee K-H, et al. Talazoparib in Patients with Advanced Breast Cancer and a Germline BRCA Mutation. New England Journal of Medicine 2018;379:753–763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Abida W, Patnaik A, Campbell D, Shapiro J, Bryce AH, McDermott R, et al. Rucaparib in Men with Metastatic Castration-Resistant Prostate Cancer Harboring a BRCA1 or BRCA2 Gene Alteration. Journal of Clinical Oncology 2020;38:3763–3772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.de Bono J, Mateo J, Fizazi K, Saad F, Shore N, Sandhu S, et al. Olaparib for Metastatic Castration-Resistant Prostate Cancer. New England Journal of Medicine 2020;382:2091–2102. [DOI] [PubMed] [Google Scholar]
  • 24.Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, et al. Maintenance Olaparib for Germline BRCA -Mutated Metastatic Pancreatic Cancer. New England Journal of Medicine 2019;381:317–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ghiorzo P Genetic predisposition to pancreatic cancer. World journal of gastroenterology 2014;20:10778–10789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Brody LC. Treating Cancer by Targeting a Weakness. New England Journal of Medicine 2005; [DOI] [PubMed] [Google Scholar]
  • 27.Bryant HE, Schultz N, Thomas HD, Parker KM, Flower D, Lopez E, et al. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 2005; [DOI] [PubMed] [Google Scholar]
  • 28.Farmer H, McCabe H, Lord CJ, Tutt AHJ, Johnson DA, Richardson TB, et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005; [DOI] [PubMed] [Google Scholar]
  • 29.Shen Y, Aoyagi-Scharber M, Wang B. Trapping Poly(ADP-Ribose) Polymerase. The Journal of pharmacology and experimental therapeutics 2015;353:446–457. [DOI] [PubMed] [Google Scholar]
  • 30.Kristeleit R, Shapiro GI, Burris HA, Oza AM, LoRusso P, Patel MR, et al. A phase I–II study of the oral PARP inhibitor rucaparib in patients with germline BRCA1/2-mutated ovarian carcinoma or other solid tumors. Clinical Cancer Research 2017; [DOI] [PubMed] [Google Scholar]
  • 31.Shen Y, Rehman FL, Feng Y, Boshuizen J, Bajrami I, Elliott R, et al. BMN673, a novel and highly potent PARP1/2 inhibitor for the treatment of human cancers with DNA repair deficiency. Clinical Cancer Research 2013;19:5003–5015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Murai J, Huang SYN, Renaud A, Zhang Y, Ji J, Takeda S, et al. Stereospecific PARP trapping by BMN 673 and comparison with olaparib and rucaparib. Molecular Cancer Therapeutics 2014;13:433–443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.DOBZHANSKY T Genetics of natural populations; recombination and variability in populations of Drosophila pseudoobscura. Genetics 1946; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Ashworth A A synthetic lethal therapeutic approach: Poly(ADP) ribose polymerase inhibitors for the treatment of cancers deficient in DNA double-strand break repair. Journal of Clinical Oncology 2008;26:3785–3790. [DOI] [PubMed] [Google Scholar]
  • 35.Yang X, Ndawula C, Zhou H, Gong X, Jin J. JF-305, a pancreatic cancer cell line is highly sensitive to the PARP inhibitor olaparib. Oncology letters 2015;9:757–761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Mergui-Roelvink M, et al. Inhibition of Poly(ADP-Ribose) Polymerase in Tumors from BRCA Mutation Carriers. New England Journal of Medicine 2009; [DOI] [PubMed] [Google Scholar]
  • 37.Sandhu SK, Schelman WR, Wilding G, Moreno V, Baird RD, Miranda S, et al. The poly(ADP-ribose) polymerase inhibitor niraparib (MK4827) in BRCA mutation carriers and patients with sporadic cancer: A phase 1 dose-escalation trial. The Lancet Oncology Elsevier Ltd, 2013;14:882–892. [DOI] [PubMed] [Google Scholar]
  • 38.Li H, Liu R, Shao B, Ran R, Song G, Wang K, et al. Phase I dose-escalation and expansion study of PARP inhibitor, fluzoparib (SHR3162), in patients with advanced solid tumors. Chinese Journal of Cancer Research 2020;32:370–382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.de Bono J, Ramanathan RK, Mina L, Chugh R, Glaspy J, Rafii S, et al. Phase I, dose-escalation, two-part trial of the PARP inhibitor talazoparib in patients with advanced germline BRCA1/2 mutations and selected sporadic cancers. Cancer Discovery 2017;7:620–629. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Plummer R, Jones C, Middleton M, Wilson R, Evans J, Olsen A, et al. Phase i study of the poly(ADP-Ribose) polymerase inhibitor, AG014699, in combination with temozolomide in patients with advanced solid tumors. Clinical Cancer Research 2008;14:7917–7923. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Calabrese CR, Almassy R, Barton S, Batey MA, Calvert AH, Canan-Koch S, et al. Anticancer chemosensitization and radiosensitization by the novel poly(ADP-ribose) polymerase-1 inhibitor AG14361. Journal of the National Cancer Institute 2004;96:56–67. [DOI] [PubMed] [Google Scholar]
  • 42.O’Reilly EM, Lee JW, Lowery MA, Capanu M, Stadler ZK, Moore MJ, et al. Phase 1 trial evaluating cisplatin, gemcitabine, and veliparib in 2 patient cohorts: Germline BRCA mutation carriers and wild-type BRCA pancreatic ductal adenocarcinoma. Cancer 2018;124:1374–1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Tuli R, Shiao SL, Nissen N, Tighiouart M, Kim S, Osipov A, et al. A phase 1 study of veliparib, a PARP-1/2 inhibitor, with gemcitabine and radiotherapy in locally advanced pancreatic cancer. EBioMedicine The Authors, 2019;40:375–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kaufman B, Shapira-Frommer R, Schmutzler RK, Audeh MW, Friedlander M, Balmaña J, et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. Journal of Clinical Oncology 2015;33:244–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Shroff RT, Hendifar A, McWilliams RR, Geva R, Epelbaum R, Rolfe L, et al. Rucaparib Monotherapy in Patients With Pancreatic Cancer and a Known Deleterious BRCA Mutation. JCO Precision Oncology 2018; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Reiss KA, Mick R, O’Hara MH, Teitelbaum U, Karasic TB, Schneider C, 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. Journal of Clinical Oncology 2021;JCO.21.00003. [DOI] [PubMed] [Google Scholar]
  • 47.Lowery MA, Kelsen DP, Capanu M, Smith SC, Lee JW, Stadler ZK, et al. Phase II trial of veliparib in patients with previously treated BRCA-mutated pancreas ductal adenocarcinoma. European Journal of Cancer Elsevier Ltd, 2018;89:19–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Javle M, Shacham-Shmueli E, Xiao L, Varadhachary G, Halpern N, Fogelman D, et al. Olaparib Monotherapy for Previously Treated Pancreatic Cancer With DNA Damage Repair Genetic Alterations Other Than Germline BRCA Variants. JAMA Oncology 2021;7:693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Hammel P, Kindler HL, Reni M, Van Cutsem E, MacArulla T, Hall MJ, et al. Health-related quality of life in patients with a germline BRCA mutation and metastatic pancreatic cancer receiving maintenance olaparib. Annals of Oncology 2019; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Golan T, Hammel P, Reni M, Van Cutsem E, Macarulla T, Hall MJ, et al. Overall survival from the phase 3 POLO trial: Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. Journal of Clinical Oncology 2021; [Google Scholar]
  • 51.FDA approves olaparib for gBRCAm metastatic pancreatic adenocarcinoma.. Available from: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-olaparib-gbrcam-metastatic-pancreatic-adenocarcinoma.
  • 52.Golan T, O’Kane GM, Denroche RE, Raitses-Gurevich M, Grant RC, Holter S, et al. Genomic Features and Classification of Homologous Recombination Deficient Pancreatic Ductal Adenocarcinoma. Gastroenterology 2021;160:2119–2132.e9. [DOI] [PubMed] [Google Scholar]
  • 53.Pettitt SJ, Frankum JR, Punta M, Lise S, Alexander J, Chen Y, et al. Clinical BRCA1/2 Reversion Analysis Identifies Hotspot Mutations and Predicted Neoantigens Associated with Therapy Resistance. Cancer discovery 2020;10:1475–1488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Taglialatela A, Alvarez S, Leuzzi G, Sannino V, Ranjha L, Huang JW, et al. Restoration of Replication Fork Stability in BRCA1- and BRCA2-Deficient Cells by Inactivation of SNF2-Family Fork Remodelers. Molecular Cell Elsevier Inc., 2017;68:414–430.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Dungrawala H, Bhat KP, Le Meur R, Chazin WJ, Ding X, Sharan SK, et al. RADX Promotes Genome Stability and Modulates Chemosensitivity by Regulating RAD51 at Replication Forks. Molecular Cell 2017;67:374–386.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Gupta R, Somyajit K, Narita T, Maskey E, Stanlie A, Kremer M, et al. DNA Repair Network Analysis Reveals Shieldin as a Key Regulator of NHEJ and PARP Inhibitor Sensitivity. Cell Elsevier Inc., 2018;173:972–988.e23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Dev H, Chiang TWW, Lescale C, de Krijger I, Martin AG, Pilger D, et al. Shieldin complex promotes DNA end-joining and counters homologous recombination in BRCA1-null cells. Nature Cell Biology Springer US, 2018;20:954–965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Vaidyanathan A, Sawers L, Gannon AL, Chakravarty P, Scott AL, Bray SE, et al. ABCB1 (MDR1) induction defines a common resistance mechanism in paclitaxel- and olaparib-resistant ovarian cancer cells. British Journal of Cancer Nature Publishing Group, 2016;115:431–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Rottenberg S, Jaspers JE, Kersbergen A, Van Der Burg E, Nygren AOH, Zander SAL, et al. High sensitivity of BRCA1-deficient mammary tumors to the PARP inhibitor AZD2281 alone and in combination with platinum drugs. Proceedings of the National Academy of Sciences of the United States of America 2008;105:17079–17084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Dias MP, Moser SC. Understanding and overcoming resistance to PARP inhibitors in cancer therapy. Nature Reviews Clinical Oncology Springer US, 2021;0123456789. [DOI] [PubMed] [Google Scholar]
  • 61.Murai J, Feng Y, Yu GK, Ru Y, Tang SW, Shen Y, et al. Resistance to PARP inhibitors by SLFN11 inactivation can be overcome by ATR inhibition. Oncotarget 2016;7:76534–76550. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Jaspers JE, Sol W, Kersbergen A, Schlicker A, Guyader C, Xu G, et al. BRCA2-deficient sarcomatoid mammary tumors exhibit multidrug resistance. Cancer Research 2015;75:732–741. [DOI] [PubMed] [Google Scholar]
  • 63.Barazas M, Annunziato S, Pettitt SJ, de Krijger I, Ghezraoui H, Roobol SJ, et al. The CST Complex Mediates End Protection at Double-Strand Breaks and Promotes PARP Inhibitor Sensitivity in BRCA1-Deficient Cells. Cell Reports ElsevierCompany., 2018;23:2107–2118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Johnson N, Johnson SF, Yao W, Li YC, Choi YE, Bernhardy AJ, et al. Stabilization of mutant BRCA1 protein confers PARP inhibitor and platinum resistance. Proceedings of the National Academy of Sciences of the United States of America 2013;110:17041–17046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Waks AG, Cohen O, Kochupurakkal B, Kim D, Dunn CE, Buendia Buendia J, et al. Reversion and non-reversion mechanisms of resistance to PARP inhibitor or platinum chemotherapy in BRCA1/2-mutant metastatic breast cancer. Annals of Oncology Elsevier Ltd., 2020;31:590–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Cruz C, Castroviejo-Bermejo M, Gutiérrez-Enríquez S, Llop-Guevara A, Ibrahim YH, Gris-Oliver A, et al. RAD51 foci as a functional biomarker of homologous recombination repair and PARP inhibitor resistance in germline BRCA-mutated breast cancer. Annals of Oncology 2018;29:1203–1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Jaspers JE, Kersbergen A, Boon U, Sol W, Van Deemter L, Zander SA, et al. Loss of 53BP1 causes PARP inhibitor resistance in BRCA1-mutated mouse mammary tumors. Cancer Discovery 2013;3:68–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Casolino R, Paiella S, Azzolina D, Beer PA, Corbo V, Lorenzoni G, et al. Homologous Recombination Deficiency in Pancreatic Cancer: A Systematic Review and Prevalence Meta-Analysis. Journal of Clinical Oncology 2021;39:2617–2631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Fuh K, Mullen M, Blachut B, Stover E, Konstantinopoulos P, Liu J, et al. Homologous recombination deficiency real-time clinical assays, ready or not? Gynecologic Oncology Elsevier Inc., 2020;159:877–886. [DOI] [PubMed] [Google Scholar]
  • 70.Tung NM, Robson ME, Ventz S, Santa-Maria CA, Nanda R, Marcom PK, et al. TBCRC 048: Phase II Study of Olaparib for Metastatic Breast Cancer and Mutations in Homologous Recombination-Related Genes. Journal of Clinical Oncology 2020; [DOI] [PubMed] [Google Scholar]
  • 71.Alexandrov LB, Kim J, Haradhvala NJ, Huang MN, Tian Ng AW, Wu Y, et al. The repertoire of mutational signatures in human cancer. Nature 2020;578:94–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Nik-Zainal S, Davies H, Staaf J, Ramakrishna M, Glodzik D, Zou X, et al. Landscape of somatic mutations in 560 breast cancer whole-genome sequences. Nature Nature Publishing Group, 2016;534:47–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Abkevich V, Timms KM, Hennessy BT, Potter J, Carey MS, Meyer LA, et al. Patterns of genomic loss of heterozygosity predict homologous recombination repair defects in epithelial ovarian cancer. British Journal of Cancer 2012;107:1776–1782. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Aguirre AJ, Nowak JA, Camarda ND, Moffitt RA, Ghazani AA, Hazar-Rethinam M, et al. Real-time genomic characterization of advanced pancreatic cancer to enable precision medicine. Cancer Discovery 2018;8:1096–1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.González-Martín A, Pothuri B, Vergote I, DePont Christensen R, Graybill W, Mirza MR, et al. Niraparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. New England Journal of Medicine 2019;381:2391–2402. [DOI] [PubMed] [Google Scholar]
  • 76.Hill SJ, Decker B, Roberts EA, Horowitz NS, Muto MG, Worley MJ, et al. Prediction of DNA repair inhibitor response in short-term patient-derived ovarian cancer organoids. Cancer Discovery 2018;8:1404–1421. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.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.. [DOI] [PubMed]
  • 78.Tutt ANJ, Garber JE, Kaufman B, Viale G, Fumagalli D, Rastogi P, et al. Adjuvant Olaparib for Patients with BRCA1 - or BRCA2 -Mutated Breast Cancer. New England Journal of Medicine 2021;384:2394–2405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Stewart RA, Pilie PG, Yap TA. Development of PARP and immune-checkpoint inhibitor combinations. Cancer Research 2018;78:6717–6725. [DOI] [PubMed] [Google Scholar]
  • 80.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 Research 2019;79:311–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Huang J, Wang L, Cong Z, Amoozgar Z, Kiner E, Xing D, et al. The PARP1 inhibitor BMN 673 exhibits immunoregulatory effects in a Brca1−/− murine model of ovarian cancer. Biochemical and Biophysical Research Communications Elsevier Ltd, 2015;463:551–556. [DOI] [PubMed] [Google Scholar]
  • 82.Jiao S, Xia W, Yamaguchi H, Wei Y, Chen MK, Hsu JM, et al. PARP inhibitor upregulates PD-L1 expression and enhances cancer-associated immunosuppression. Clinical Cancer Research 2017;23:3711–3720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Miller AL, Fehling SC, Garcia PL, Gamblin TL, Council LN, van Waardenburg RCAM, et al. The BET inhibitor JQ1 attenuates double-strand break repair and sensitizes models of pancreatic ductal adenocarcinoma to PARP inhibitors. EBioMedicine Elsevier B.V., 2019;44:419–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Yang L, Zhang Y, Shan W, Hu Z, Yuan J, Pi J, et al. Repression of BET activity sensitizes homologous recombination-proficient cancers to PARP inhibition. Science translational medicine 2017;9:1–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Bindra RS, Crosby ME, Glazer PM. Regulation of DNA repair in hypoxic cancer cells. Cancer and Metastasis Reviews 2007;26:249–260. [DOI] [PubMed] [Google Scholar]
  • 86.Bindra RS, Schaffer PJ, Meng A, Woo J, Måseide K, Roth ME, et al. Down-Regulation of Rad51 and Decreased Homologous Recombination in Hypoxic Cancer Cells. Molecular and Cellular Biology 2004;24:8504–8518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.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) Neoplasia Press, Inc., 2014;16:343–353.e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Alvarez Secord A, O’Malley DM, Sood AK, Westin SN, Liu JF. Rationale for combination PARP inhibitor and antiangiogenic treatment in advanced epithelial ovarian cancer: A review. Gynecologic Oncology Elsevier Inc., 2021; [DOI] [PubMed] [Google Scholar]
  • 89.Jette NR, Radhamani S, Arthur G, Ye R, Goutam S, Bolyos A, et al. Combined poly-ADP ribose polymerase and ataxia-telangiectasia mutated/Rad3-related inhibition targets ataxia-telangiectasia mutated-deficient lung cancer cells. British Journal of Cancer Springer US, 2019;121:600–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Nam AR, Yoon J, Jin MH, Bang JH, Oh KS, Seo HR, et al. ATR inhibition amplifies antitumor effects of olaparib in biliary tract cancer. Cancer Letters Elsevier B.V., 2021;516:38–47. [DOI] [PubMed] [Google Scholar]
  • 91.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 Discovery 2012;2:1036–1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.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 Discovery 2020;10:1528–1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Sullivan-Reed K, Bolton-Gillespie E, Dasgupta Y, Langer S, Siciliano M, Nieborowska-Skorska M, et al. Simultaneous Targeting of PARP1 and RAD52 Triggers Dual Synthetic Lethality in BRCA-Deficient Tumor Cells. Cell Reports 2018;23:3127–3136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Bajrami I, Kigozi A, Van Weverwijk A, Brough R, Frankum J, Lord CJ, et al. Synthetic lethality of PARP and NAMPT inhibition in triple-negative breast cancer cells. EMBO Molecular Medicine 2012;4:1087–1096. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Zhou J, Gelot C, Pantelidou C, Li A, Yücel H, Davis RE, et al. A first-in-class polymerase theta inhibitor selectively targets homologous-recombination-deficient tumors. Nature Cancer Springer US, 2021;2:598–610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Mateos-Gomez PA, Gong F, Nair N, Miller KM, Lazzerini-Denchi E, Sfeir A. Mammalian polymerase θ promotes alternative NHEJ and suppresses recombination. Nature Nature Publishing Group, 2015;518:254–257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ceccaldi R, Liu JC, Amunugama R, Hajdu I, Primack B, Petalcorin MIR, et al. Homologous-recombination-deficient tumours are dependent on Polθ-mediated repair. Nature Nature Publishing Group, 2015;518:258–262. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES