Introduction
There have been tremendous advances in the treatment of metastatic cancer patients with the widespread incorporation of immune checkpoint inhibitors into clinical practice [1-7]. Unfortunately, in pancreatic adenocarcinoma, checkpoint inhibitors only appear to be effective in patients with microsatellite instability-high tumors, which comprises only 1% of patients with metastatic disease [8••, 9]. Five-year survival rates remain dismal in patients with metastatic disease at < 10% while the incidence of pancreatic cancer continues to increase [10]. Despite this, the treatment landscape for metastatic pancreatic adenocarcinoma (mPDA) is being redefined through novel targets which have emerged with the widespread incorporation of next-generation sequencing (NGS) tumor testing. Among the most promising of these targets are genes encoding for instrumental proteins which orchestrate DNA damage repair (DDR) and are responsible for ensuring genomic integrity in tumor cells in the face of replication stress and cytotoxic insults [11••]. Double-strand break repair is the most critical DDR pathway given unregulated double-strand breaks result in mitotic catastrophe [12]. Homologous recombination (HR) and non-homologous end joining (NHEJ) are the two double-strand break repair pathways, with HR most commonly utilized by eukaryotic cells because of its error-free nature [13]. Based upon the crucial role HR plays in maintaining cellular integrity, it is unsurprising why defects in HR machinery render tumor cells sensitive to DNA damage-inducing agents. Beyond novel targets, NGS has led to insights about optimal therapeutic sequencing for patients through identification of specific tumor signatures (e.g., choosing platinum chemotherapy in the first-line setting for patients with DDR-deficient tumors) [11••]. Inthesub-sequent paragraphs, we will discuss DDR targets and the therapeutics which have demonstrated promise in targeting them.
DDR
The prevalence of somatic and germline mutations in DDR genes in mPDA is higher than previously reported with 17–25% of patients having tumors with these mutations [12, 13]. HR defects comprise most of these mutations in pancreatic tumors from aggregate data presented by Pishvaian et al. [11••].
Patients with mPDA who have DDR mutations demonstrate markedly improved survival when exposed to platinum chemotherapy when compared to patients with DDR intact tumors (2.37 years versus 1.45 years, p < .0001). In the absence of exposure to platinum chemotherapy, patients with DDR-deficient tumors demonstrate no difference in survival compared to patients with DDR intact tumors.
Although it was previously believed that only patients with germline DDR mutations benefit from DNA damaging agents such as platinum chemotherapy and poly (ADP-ribose) polymerase (PARP) inhibitors, several studies suggest this benefit also extends to patients with somatic mutations [14, 15••, 16••].
Among DDR inhibitors, PARP inhibitors have the most clinical data in mPDA. There are, however, other DDR inhibitors targeting ataxia-telangiectasia and Rad3 related (ATR), ataxia-telangiectasia mutated (ATM), DNA-dependent polymerase kinase (DNA-PK), and WEE1 which have demonstrated preclinical and early clinical promise [17-19]. Tables 1 and 2 are a list of completed and ongoing PARP inhibitor studies in mPDA patients, respectively. Figure 1 illustrates the concept of synthetic lethality in HR-deficient cancer cell lines when both PARP and ATR are targeted.
Table 1.
Completed trials of PARP inhibitors in PDA patients
| Trial number |
Stage of study |
PARP inhibitor (monotherapy or combination) |
Number of PDA patients |
Selection criteria (germline or somatic DDR mutations) |
Primary outcome and key secondary outcome findings |
|---|---|---|---|---|---|
| NCT02184195 | III | Olaparib | 154 | Germline BRCA | PFS 7.4 months PARP maintenance arm versus 3.8 months in placebo arm (HR .53, P = .004). |
| NCT01078662 | II | Olaparib | 23 | Germline BRCA | RR 21.7%. No significant difference in RR between BRCA1 or BRCA2 mutants. |
| NCT02042378 | II | Rucaparib | 19 | Somatic or Germline BRCA | RR 15.8%, with all responses observed in patients with platinum-sensitive disease. |
| NCT02184195 | II | Veliparib | 16 | Germline BRCA and PALB2 | RR 0%, with SD in 5 patients, all with platinum-sensitive disease. The tria was stopped early. |
| NCT01585805 | II | Cisplatin and gemcitabine ± veliparib | 50 | Germline BRCA and PALB2 | RR 74% in the triplet arm and 62.5% in the doublet arm. No significant difference in PFS and OS between the triplet and doublet arms |
| NCT01585805 | I | Cisplatin, gemcitabine, and veliparib | 17 | Germline BRCA and BRCA WT patients | MTD at DL2. RR 77.8% in germline BRCA cohort and 0% in BRCA WT cohort. |
| NCT01296763 | I | Olaparib, irinotecan, cisplatin, and mitomycin | 18 | Unselected | Addition of mitomycin was too toxic (3 DLTs) with 89% of the cohort experiencing G3/G4 AEs. The RR of all patients was 23%. |
| NCT01489865 | I | FOLFOX plus veliparib | 22 | Germline BRCA, PALB2 or FANC mutation | RP2D of veLiparib 300 mg BID in addition to standard mFOLFOX 6 doses. RR of 14%, PFS 2.6 months and OS 5.4 months. |
Abbreviations: Pancreatic adenocarcinoma—PDA; poly (ADP-ribose) polymerase—PARP; DNA damage repair—DDR; progression-free survival—PFS; hazard ration—HR; response rate—RR; recommended phase 2 dose—RP2D; maximum tolerated dose—MTD; dose level 2—DL2; overall survival—OS; grade—G; dose-limiting toxicity—DLT; adverse events—AEs
Table 2.
Ongoing trials with DDR-targeting agents in PDA patients
| Trial number |
Stage of study |
DDR agent (monotherapy or combination) |
Accrual target (patient number) |
Selection criteria (germline or somatic DDR mutations) |
Primary outcome |
|---|---|---|---|---|---|
| NCT03140670 | II | Maintenance rucaparib | 42 | Germline or somatic BRCA and PALB2 mPDA patients who do not progress on platinum chemotherapy | 6-month PFS |
| NCT02890355 | II | FOLFIRI ± veliparib | 143 | 2nd-line mPDA patients, unselected by DDR status | OS |
| NCT03553004 | II | Niraparib | 18 | 2nd-line mPDA patients with germline or somatic DDR (unspecified) mutations | RR |
| NCT03337087 | I/II | Flurouracil, nanoliposomal irinotecan, and rucaparib | 110 (not all mPDA patient) | Later-line mPDA patients for phase I, untreated mPDA with HR mutations (somatic or germline) | |
| RP2D of phase I, RR in PDA patients with HR mutations in phase II | |||||
| NCT02677038 NCT02511223 |
II | Olaparib | 34 | 2nd-line and beyond germline BRCA-negative PDA patients with DDR mutations or family history of BRCA-associated malignancies | RR |
Abbreviations: DNA damage repair—DDR; pancreatic adenocarcinoma—PDA; response rate—RR; recommended phase II dose—RP2D; homologous recombination—HR; overall survival—OS
Fig. 1.
Simplified figure illustrating how synthetic lethality is created in cancer cells, with background HR deficits (deleterious mutations in ATM pathway or BRCA), when treated with PARP inhibitors or ATR inhibitors. Crosses imply cells with intrinsic defects in the denoted proteins while –I denotes the target of the specific inhibitors. Abbreviations: MMB, mismatched base; PARP, poly (ADP-ribose) polymerase; DSB, double-stranded breaks; HR, homologous recombination; ATM, ataxia-telangiectasia mutated; ATR, ataxia-telangiectasia and Rad3 related.
PARP inhibitors—monotherapy
PARP1 and PARP2 are enzymes involved in base excision repair which are normally recruited to sites of single-strand DNA breaks [20]. PARP inhibitors inhibit the enzymatic activity of both enzymes; however, their differential potency appears to be defined by PARP1 trapping, which represents an additional mechanism of action [21].
Kaufman et al. presented findings from a phase II multi-cohort study of metastatic germline BRCA mutant patients treated with olaparib 400 mg twice daily [20]. Of the 298 patients who received at least one dose of olaparib, 23 patients had mPDA. Seventeen (74%) of these patients had BRCA2 mutations while one patient had both BRCA1/BRCA2 mutations; the median number of prior therapies was 2. Response rate (RR) in the pancreatic cancer patients was 21.7% compared to 26.2% in the entire cohort. Stable disease (SD) lasting ≥ 8 weeks was seen in 34.8% of mPDA patients compared to 41.8% of the entire cohort. RR between BRCA1 and BRCA2 mPDA were similar at 20% and 23.5%, respectively. There was no significant difference in RR between mPDA patients who did (20%) and did not (25%) have prior exposure to platinum-based chemotherapy. Median progression-free survival (PFS) and overall survival (OS) were 4.6 months and 9.8 months, respectively, in mPDA patients. Anemia (17.4%), fatigue (13%), abdominal pain (4.3%), and vomiting (4.3%) were the most common Grade (G) 3 or greater adverse events reported in the pancreas cohort.
Shroff et al. presented results from the global RUCAPANC (NCT02042378) phase II study exploring the role for rucaparib in mPDA patients with germline or somatic BRCA1/BRCA2 mutations [14]. In this study, 19 patients, with a median of two prior lines of systemic therapy, were treated with rucaparib 600 mg twice daily. Of these patients, 16 (84.2%) had germline BRCA mutations while 3 (15.8%) had somatic mutations. Fifteen (78.9%) patients had tumors with BRCA2 mutations while four (21.1%) had tumors with BRCA1 mutations. RR was 15.8% across the entire cohort with two patients achieving a confirmed partial response (PR) and one achieving a complete response (CR). There was an additional patient who achieved SD for 72 weeks and, just prior to study discontinuation, was classified as having an unconfirmed CR. None of the patients with confirmed or unconfirmed responses had progressed on prior platinum chemotherapy. The disease control rate (DCR) was 31.6% in all patients and 44.4% in patients who had received only one prior chemotherapy regimen. The most common G3/G4 adverse events were anemia (31.6%), fatigue (15.8%), ascites (15.8%), thrombocytopenia (10.5%), vomiting (10.5%), increased transaminases (10.5%), abdominal pain (10.5%), and nausea (10.5%).
Lowery et al. presented findings from a phase II study which explored the efficacy of veliparib in pre-treated germline BRCA and PALB2 mutant (44% with 1 prior line, 56% with 2 prior lines of systemic therapy) PDA patients; 94% of patients were metastatic while 6% were locally advanced [22]. A total of 16 patients (31% BRCA1, 69% BRCA2) were enrolled and treated with veliparib 400 mg twice daily (the first 3 patients were treated at 300 mg twice daily to ensure tolerance). The RR was 0%, and DCR was 31%, with stable disease in five patients. The trial was stopped early for futility based upon its pre-specified efficacy threshold. One patient who remained progression-free for 9.5 months while on therapy had previously been platinum chemotherapy sensitive and had a PR to FOLFIRINOX for 6 months. All five of the patients with SD had prior platinum chemotherapy responsiveness. G3/G4 adverse events were observed in 69% of patients with the most common ones being fatigue (25%) and hyperbilirubinemia (19%).
Gollian et al. reported on the efficacy of olaparib in mPDA patients without BRCA mutations but with other DDR-deficient phenotypes which the authors termed “BRCAness [15••].” This is an ongoing two-arm parallel phase II study conducted in the USA and Israel, and 32 patients with at least prior line of systemic therapy have been treated at olaparib 400 mg twice daily. The phenotypes of patients who have been treated on study include 12 with tumors with DDR mutations (6 with ATM, 2 with somatic BRCA2, and 1 each with PALB2, FANC2, PTEN, and CCNE mutations), 5 with tumors with ATM loss by immunohistochemistry, and 14 with a family history of BRCA-associated malignancy without germline alterations. RR in the US cohort is 18.1% while in the Israeli cohort is 0%. DCR in the US cohort is 72.7% while in the Israeli cohort is 29.4%. Median PFS is 14 weeks and 24.7 weeks in the Israeli and US cohorts, respectively. No G3/G4 toxicities were reported in the study at the time of presentation.
Based on prior findings that rucaparib only demonstrated response in mPDA patients who were platinum chemotherapy sensitive, Reiss et al. conducted a phase II study of maintenance rucaparib in 42 patients with tumors with pathogenic mutations in BRCA1/BRCA2 or PALB2 (germline or somatic) who had not progressed on platinum chemotherapy [16••]. Participants in the study had received at least 4 months of prior platinum chemotherapy and achieved disease control prior to starting 600 mg twice daily of rucaparib. At the time of the interim analysis, 19 of 24 patients were evaluable for response. RR was 37% with 1 CR and 6 PR while DCR for at least 2 months was 90%. The median PFS in treated patients was 9.1 months. G3/G4 adverse events were not specifically reported but common adverse events included nausea, dysgeusia, and fatigue.
Golan et al. presented findings from the POLO trial (NCT02184195), a randomized phase III study in which 154 patients with germline BRCA1/BRCA2 mutant mPDA who had not progressed after at least 16 weeks first-line platinum chemotherapy were randomized in a 3:2 fashion to olaparib 300 mg twice daily or placebo [23••]. The primary endpoint of the study was PFS while secondary endpoints included OS and RR. Median PFS in the experimental arm was 7.4 months while that in the in the placebo arm was 3.8 months (hazard ratio (HR) .53; 95% confidence interval (CI), .35–.82; P = .004). At the time of interim analysis, after data maturity in 46% of the study population, median OS in the olaparib and placebo arms was 18.9 and 18.1 months, respectively (HR .91; 95% CI, 0.56 to 1.46; P = 0.68). Among patients with measurable disease at baseline, RR was 23% in the study arm and 12% in the placebo arm; median duration of response was 24.9 months in the study arm and 3.7 months in the placebo arm. Serious adverse events were experienced by 24% and 15% of patients in treatment and placebo arms, respectively; 10% of patients treated with olaparib experienced ≥ G3 anemia. Based on these study findings, olaparib received FDA approval as a maintenance therapy for germline BRCA mutant PDA patients with non-progressive disease after initial platinum chemotherapy.
From the previously described studies, PARP inhibitors appear to have the most anti-tumor activity in mPDA patients who remain sensitive to platinum chemotherapy. Platinum-sensitive clinical phenotype may be a surrogate for a HR-deficient genotype. PARP inhibitors have mostly been tested in patients who have germline DDR mutations (BRCA and beyond) however appear to also have anti-tumor effects in patients with somatic DDR mutations. PARP inhibitor monotherapy in mPDA, whether in maintenance or later-line settings, appears to have less activity than PARP inhibitors in other disease sites where they are routinely utilized [24•, 25].
PARP inhibitors—combined therapy
Beyond PARP inhibitor monotherapy, combinations of the agents with chemotherapy have also been trialed. Yarchoan et al. enrolled 18 mPDA patients on a two-center phase I study and treated them first with the combination of olaparib (twice daily day(D)1, D8), cisplatin (D1, D8), and irinotecan (D1, D8) [26]. The maximum tolerated dose (MTD) of this combination was cisplatin 25 mg/m2 D1, D8; irinotecan 70 mg/m2 D1, D8; and olaparib 100 mg twice daily on D1, D8. After this MTD was achieved, the investigators explored the safety and tolerability of adding mitomycin 5 mg/m2 on D1. Only six patients received the mitomycin addition and tolerated it poorly with two patients developing G4 neutropenia and one patient developing G3 febrile neutropenia. Given the three observed dose-limiting toxicities (DLTs), the combination was deemed too toxic. Thirteen patients had received two or more lines of therapy while three patients had received one prior therapy line; two patients were known BRCA2 mutants. Safety data was collected for the entire cohort, and 16 (89%) patients developed G3/G4 toxicities with the most common toxicities being neutropenia (89%), lymphopenia (72%), and anemia (22%). Thirteen patients were evaluable for response while five came off therapy prior to the first evaluation. RR was 23% with three PR, and DCR was 62%. Among the two patients with BRCA2 mutations, one achieved a PR for 4 years prior to succumbing to therapy-associated acute myeloid leukemia while the other achieved SD for 3 months.
O’Reilly et al. presented findings from a phase I trial of cisplatin, gemcitabine, and veliparib in 17 advanced PDA patients (88.2% metastatic, 11.8% locally advanced) [27]. Two cohorts of patients (nine with germline mutated BRCA, seven with wildtype BRCA, and one with unknown BRCA status) were enrolled in the study and treated with gemcitabine 600 mg/m2 D3, D10; cisplatin 25 mg/m2 D3, D10; and veliparib at escalating doses on every 21-day cycle. MTD was reached at dose level 2 of veliparib at a dose of 80 mg twice daily on D1-D12. The dose level 2A (D1–21) dosing of veliparib was too toxic for patients with 4 DLTs (50% neutropenia, 50% thrombocytopenia) observed. There were two G5 events in the entire patient cohort with one patient developing therapy-associated AML and the other suffering from a colonic perforation. RR was 77.8% in BRCA mutant patients with one CR and six PR while DCR was 100%. In the BRCA wildtype patient cohort, RR was 0% and DCR was 85.7%. Median OS was 23.3 months and 11 months in BRCA mutant and wildtype patients, respectively.
Findings from the phase I study of cisplatin, gemcitabine, and veliparib established the dose for the combination used in the randomized phase II portion of the study comparing the efficacy of cisplatin and gemcitabine ± veliparib in 50 germline BRCA- and PALB2-mutated patients. O’Reilly et al. recently presented the findings from this portion of the trial [28••]. Patients were randomized to either gemcitabine 600 mg/m2 D3, D10; cisplatin 25 mg/m2 D3, D10; and veliparib 80 mg D1-D12 every 21 days or gemcitabine and cisplatin at the same schedule. The primary endpoint of the study was RR in each arm with secondary endpoints of PFS, DCR, and OS. The RR was 74% in the triplet arm and 65.2% in the doublet arm, with both arms exceeding pre-specified response thresholds to move to the subsequent stage of the trial. Median PFS was 10.1 months for the triplet arm and 9.7 months for the doublet arm (P = .73). Median OS was 15.5 months for the triplet arm and 16.4 months for the doublet arm (P = .6). Patients treated with the triplet combination experienced more than twice the number of hematologic G3/G4 toxicities (53%) compared to patients treated with the doublet combination (22%). The 2-year survival for the entire cohort was 30.6% (95% CI, 17.8–44.4%). The study results suggest that while veliparib may not have added additional PFS or OS benefit to gemcitabine plus cisplatin in BRCA- and PALB2-mutated advanced PDA patients, gemcitabine plus cisplatin is a highly active combination in PDA tumors with such germline mutations.
Pishvaian et al. reported the final findings from the phase I/II study of modified FOLFOX 6 (D1–D3) plus veliparib (twice daily on D1–D7) every 14 days in advanced PDA patients at ASCO 2019 [29]. The recommended phase 2 dose (RP2D) of the combination was veliparib 200 mg twice daily along with standard doses of modified FOLFOX. No DLTs were observed in the phase I portion of the study. In the phase II portion of the study, two cohorts of patients were enrolled (treatment naive and pre-treated) and were pre-selected if their tumors carried germline or somatic mutations in BRCA1/2, PALB2, and ATM and if they had family histories suggestive of a breast or ovarian cancer syndrome. Of 57 patients evaluable for response, RR was 26%, median PFS was 3.7 months, and median OS was 8.5 months. In patients with a family history suggestive of breast and ovarian cancer syndrome (N = 43), median PFS and OS were 4.3 and 10.1 months, respectively. In patients with tumors with DDR mutations (N = 16), median PFS and OS were 7.2 and 11.1 months, respectively. Intriguingly, in treatment naive patients with positive family history and DDR mutant tumors, ORR of the combination was 58%.
PARP inhibitors in combination with platinum chemotherapy elicit a higher RR in advanced PDA patients than chemotherapy alone however whether that translates to a PFS or OS benefit is unclear. Patients with tumors with DDR mutations, either germline or somatic, appear to derive a disproportionate benefit from the treatment combinations compared to patients with tumors with intact DDR. PDA patients with germline mutations in BRCA or other HR genes may be especially sensitive to platinum chemotherapy, even as a monotherapy.
Therapies beyond PARP inhibitors
ATM is one of the two most upstream kinases which coordinates the DDR response and is stimulated by double-strand breaks. It activates downstream effectors such as Chk2 and TP53 which play crucial roles in directly orchestrating DNA repair and cell cycle arrest [30•]. Perkhofer et al. reported findings from cell line models of ATM-deficient PDA (AKC) suggesting these cell lines were particularly sensitive to ATR inhibition [17]. The AKC cell lines were more sensitive to ATR inhibition than the KC (ATM intact PDA) cell lines by IC50. The authors then investigated the effect of the ATR inhibitor VE-822 in a subcutaneous transplantation model. VE-822 significantly reduced tumor progression in the AKC model while it had minimal effects in the KC model. Adding gemcitabine to VE-822 increased the tumor stabilizing effect in AKC models while not meaningfully slowing tumor growth in KC models.
The ATM inhibitor AZD0156 has been trialed preclinically in combination with cisplatin in patient-derived pancreatic cancer cell lines [31]. Even in cell lines devoid of DDR deficiencies, AZD0156 sensitized the tumor cells to cisplatin, suggesting the ability of the inhibitor to create a DDR-deficient tumor phenotype.
ATR is a primary DDR kinase which is stimulated by single-strand intermediates and replication stress [30•]. Wallez et al. reported findings from experiments with the ATR inhibitor AZD6738 in combination with gemcitabine in mouse PDA cell lines, human PDA cell lines, and a TP53- deficient subcutaneous PDA model [18]. All four cell lines from the KPC mouse model (KRAS G12D, TP53 R172H) were very sensitive to AZD6738 monotherapy alone. Several human PDA cell lines which demonstrated intrinsic resistance to the ATR inhibitor included MiaPaCa-2 and Panc-1. Upon further interrogation, the investigators found these cell lines upregulated ATM and DNA-PK when exposed to AZD6738. In clonogenic survival assays of both cell lines, this resistance was able to be overcome with the addition of gemcitabine. AZD6738 plus gemcitabine was tested in vivo in K8484 allograft models. For this part of the experiment, allograft mice were treated in each of the four arms, AZD6738 monotherapy at 25 mg/kg 5 days per week, gemcitabine monotherapy 100 mg/kg twice per week, placebo, and the combination. In the AZD6738 monotherapy arm, there was no tumor shrinkage or slowing of growth while in the gemcitabine monotherapy arm there was slowing of growth without shrinkage. In the combination arm, 7 of 10 mice demonstrated tumor shrinkage by the end of week 3 of treatment. There was no significant change in mouse body weight in the combination arm compared to the monotherapy arms.
Estrada-Bernal et al. explored the radiosensitizing ability of the MEK1/2 inhibitor trametinib (GSK212) in KRAS mutant PDA cell lines MIAPaCa-2 and AsPC-1 [19]. In clonogenic survival assay experiments, the authors found pre-treating MIAPaCa-2 for 24 h or 48 h with 10 nm of trametinib sensitized the cells the ionizing radiation. This was not observed when the cells were pre-treated just prior to radiation with the MEK inhibitor or if the drug was administered 1-h post-radiation, suggesting that sustained MEK1/2 pathway inhibition is what creates radiosensitivity in these cell lines. Because non-homologous end joining (NHEJ) and HR are the DDR mechanisms triggered by radiation, the investigators wanted to assess the specific mechanism by which trametinib radiosensitized the PDA cell lines. The authors demonstrated this by pre-treating MiaPACa-2 cells with trametinib 24 h prior to administered ionizing radiation. The MEK inhibitor blunted NHEJ through decreasing expression of DNA-PK and blunted HR through downregulating BRCA1 and RAD51 expression.
TP53 is mutated in > 70% of PDA, leading to loss of the G1/S checkpoint and dependence on the G2/M to maintain genomic integrity [31, 32]. WEE1 is a key regulator of the G2/M checkpoint through its inactivation of CDK1, and thus, WEE1 inhibitors may play a therapeutic role in this disease [33].
A phase I study in 34 locally advanced PDA patients tested the WEE1 inhibitor AZD1775 (adavosertib) in combination with gemcitabine and radiation therapy [34]. Patients in the study received 4 cycles of oral adavosertib D1, D2 and D8, D9 (100–175 mg), IV gemcitabine 1000 mg/m2 D1, D8, and radiation during cycles 2 and 3. Each cycle was 21 days. The RP2D of adavosertib was chosen to be 150 mg in the setting of the DLTs (anorexia, nausea, and fatigue) experienced by 33% of study patients. Median OS in the study patients was 21.4 months while median PFS was 9.4 months.
Summary
DDR mutations may represent an Achilles’ heel of advanced PDA and are present in 17–25% of tumors. Beyond patients with germline BRCA- and PALB2-associated tumors, patients with tumors that harbor somatic DDR mutations appear to also derive differential benefit from agents such as PARP inhibitors and platinum chemotherapy. Patients who appear to derive the greatest benefit from PARP inhibitors, from existing clinical trials, appear to be those who possess platinum-sensitive tumors and are given the agents in combination with DNA damaging cytotoxic therapy. The role for maintenance therapy with PARP inhibitors after platinum chemotherapy has been established in patients with germline BRCA mutations however needs more definitive exploration in patients with somatic DDR mutations. Furthermore, whether PARP inhibitors should be administered with other agents in the maintenance setting to maximize efficacy needs prospective evaluation. Beyond PARP inhibitors, an array of DDR-targeting agents have demonstrated preclinical promise in DDR-deficient PDA models and include ATM, ATR, DNA-PK, and WEE1 inhibitors; clinical validation remains pending. We still have much work to do in order to improve outcomes for patients with PDA; however, the future appears promising given our improved mechanistic understanding about the biology of the disease and development of potent therapeutics which seek to exploit some of these mechanisms.
Opinion statement.
Metastatic (and locally advanced) pancreatic adenocarcinoma (mPDA) represents a major challenge for the oncology community given the rising mortality burden from the disease and the preponderance of patients diagnosed with unresectable disease. Although systemic therapies have become more potent with the development of fluorouracil, irinotecan, and oxaliplatin (FOLFIRINOX) and gemcitabine plus nab-paclitaxel as first-line treatments, the median overall survival for patients treated with either of these regimens remains just above 1 year. A significant need exists to build upon the effectiveness of first-line regimens, incorporate tolerable maintenance treatments, and add effective later-line options for patients with this disease. We believe every newly diagnosed mPDA patient should undergo next-generation sequencing (NGS) testing, preferably from tumor tissue, to assess for the presence of DNA damage repair (DDR) defects, microsatellite instability, and other possible actionable molecular alterations (such as neurotrophic tropomysin receptor kinase (NTRK) fusions, anaplastic lymphoma kinase (ALK) rearrangements, or human epidermal growth factor receptor 2 (HER2) amplification). Existing clinical data suggests that patients, whose tumors harbor DDR defects, benefit from treatment with platinum-based chemotherapy and poly (ADP-ribose) polymerase (PARP) inhibitors. Preclinically, inhibitors of other critical players in DDR such as ataxia-telangiectasia and Rad3 related (ATR), ataxia-telangiectasia mutated (ATM), DNA-dependent protein kinase (DNA-PK), and WEE1 have demonstrated promising anti-tumor activity in PDA cell lines and xenografts. How to move forward the preclinical promise of these newer DDR-targeting therapies into rational clinical trial combinations and sequence PARP inhibitors in relation to platinum chemotherapy remain areas of tremendous clinical research interest. We believe clinical trials should be considered early for mPDA patients, in all treatment Lines, so that novel therapies may be added to the treatment armamentarium for patients with this disease. Beyond NGS testing from tumor tissue, we believe it is important to consider germline genetic testing for all patients diagnosed with PDA given recent data suggesting a much stronger hereditary component of the disease than previously understood, and the potential screening implications for family members.
Footnotes
Conflict of Interest
Satya Das and Dana Cardin declare they have no conflict of interest.
Human and Animal Rights and Informed Consent
This article does not contain any studies with human or animal subjects performed by any of the authors.
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
References and Recommended Reading
Papers of particular interest, published recently, have been highlighted as:
• Of importance
•• Of major importance
- 1.Gettinger S, Horn L, Jackman D, Spigel D, Antonia S, Hellmann M, et al. Five-year follow-up of nivolumab in previously treated advanced non–small-cell lung cancer: results from the CA209-003 study. J Clin Oncol. 2018;36(17):1675–84. [DOI] [PubMed] [Google Scholar]
- 2.Hamid O, Robert C, Daud A, et al. Five-year survival outcomes for patients with advanced melanoma treated with pembrolizumab in KEYNOTE-001. Ann Oncol. 2019. 10.1093/annonc/mdz011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Ascieto PA, Long GV, Robert C, et al. Survival outcomes in patients with previously untreated BRAF wild-type advanced melanoma treated with nivolumab therapy: three-year follow-up of a randomized phase 3 trial. JAMA Oncol. 2018;5(2):187–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Motzer R, Escudier B, McDermott D, George S, Hammers HJ, Srinivas S, et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med. 2015;373:1803–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Rini B, Plimack E, Stus V, Gafanov R, Hawkins R, Nosov D, et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380:1116–27. [DOI] [PubMed] [Google Scholar]
- 6.Nghiem P, Bhatia S, Lipson E, et al. PD-1 blockade with pembrolizumab in advanced Merkel-cell carcinoma. N Engl J Med. 2016;374:2542–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Reck M, Rodriguez-Abreu D, Robinson A, et al. Pembrolizumab versus chemotherapy for PD-L1–positive non–small-cell lung cancer. N Engl J Med. 2016;375:1823–33. [DOI] [PubMed] [Google Scholar]
- 8.••.Le D, Durham J, Smith K, et al. Mismatch-repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;57(6349):409–1.A seminal article which suggested the predictive power of microsatellite instability-high tumor status, across tumor histology, to predict benefit from the checkpoint inhibitor pembrolizumab.
- 9.Royal RE, Levy C, Turner K, Mathur A, Hughes M, Kammula US, et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother. 2010;33:828–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.SEER Database 2019. https://seer.cancer.gov/statfacts/html/pancreas.html.
- 11.••.Pishvaian M, Blais E, Brody J, et al. Outcomes in pancreatic adenocarcinoma (PDA) patients (pts) with genetic alterations in DNA damage repair (DDR) pathways: results from the Know Your Tumor (KYT) program. J Clin Oncol. 2019;37(4):S19.An important publication suggesting that DDR mutations are more prevalent in PDA than previously thought and furthermore, that DDR mutations in tumors predict sensitivity to platinum chemotherapy.
- 12.Heeke A, Pishvaian M, Lynce F, et al. Prevalence of homologous recombination–related gene mutations across multiple cancer types. JCO Precis Oncol. 2018. 10.1200/PO.17.00286. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Dreyer S, Paulus-Hock V, Upstill-Goddard R, et al. Defining DNA damage repair deficiency and replication stress in pancreatic cancer. J Clin Oncol. 2019;37(4):S285. [Google Scholar]
- 14.Schroff R, Hendifar A, McWilliams R, et al. Rucaparib monotherapy in patients with pancreatic cancer and a known deleterious BRCA mutation. J Prescis Oncol. 2018. 10.1200/PO.17.00316. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.••.Golan T, Varadhachary G, Sela T, et al. Phase II study of olaparib for BRCAness phenotype in pancreatic cancer. J Clin Oncol. 2018;36(4):S29.An important study which sought to expand the role of PARP inhibitors to PDA patients beyond those with germline BRCA mutations. Preliminary results from this study suggest that PARP inhibitors have a role in PDA tumors with somatic mutations in other DDR genes.
- 16.••.Reiss Binder K, Mick R, O’Hara M, et al. CT234 - a phase II, single arm study of maintenance rucaparib in patients with platinum-sensitive advanced pancreatic cancer and a pathogenic germline or somatic mutation in BRCA1, BRCA2 or PALB2. Cancer Discov. 2019. 10.1158/2159-8290.CD-NB2019-043An important study which suggested the biggest benefit of PARP inhibitors in mPDA patients may be in the platinum chemotherapy sensitive setting. The study also included patients beyond those with just germline BRCA mutations.
- 17.Perkhofer L, Illing A, Gout J, et al. Precision medicine meets the DNA damage response in pancreatic cancer. Oncoscience. 2018;5(1–2):6–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Wallez Y, Dunlop CR, Johnson TI, Koh SB, Fornari C, Yates JWT, et al. The ATR inhibitor AZD6738 synergizes with gemcitabine in vitro and in vivo to induce pancreatic ductal adenocarcinoma regression. Mol Cancer Ther. 2018;17(8):1670–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Estrada-Bernal A, Chatterjee M, Hague SJ, et al. MEK inhibitor GSK1120212-mediated radiosensitization of pancreatic cancer cells involves inhibition of DNA double-strand break repair pathways. Cell Cycle. 2015;14(23):3713–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kaufman B, Shapira-Frommer R, Schmutzler R, et al. Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol. 2015;33(3):244–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hopkins T, Ainsworth W, Ellis P, et al. PARP1 Trapping by PARP inhibitors drives cytotoxicity in both cancer cells and healthy bone marrow. Mol Cancer Res. 2019. 10.1158/1541-7786.MCR-18-0138. [DOI] [PubMed] [Google Scholar]
- 22.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. Eur J Cancer. 2018;89:19–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.••.Golan T, Hammel P, Reni M, et al. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med. 2019. 10.1056/NEJMoa1903387A practice-changing study which demonstrated the benefit of PARP inhibitors in the platinum sensitive maintenance setting for germline BRCA mutant mPDA patients in a randomized clinical trial. This study garnered olaparib an FDA indication for maintenance therapy.
- 24.•.Moore K, Colombo N, Scambia G, et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Eng J Med. 2018;379:2495–50.An important paper describing the activity of PARP inhibitors in ovarian cancer. The activity of olaparib in this study should be contrasted with the activity of the agent in pancreatic adenocarcinoma.
- 25.Litton J, Rugo H, Ettl J, et al. Talazoparib in patients with advanced breast cancer and a germline BRCA mutation. N Engl J Med. 2018;379:753–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Yarchoan M, Myzak MC, Johnson BA, et al. Olaparib in combination with irinotecan, cisplatin, and mitomycin C in patients with advanced pancreatic cancer. Oncotarget. 2017;8(27):44073–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.O’Reilly EM, Lee JW, Lowery MA, 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(7):1374–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.••.O’Reilly EM, Lee jW, Zalupski M, 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. J Clin Oncol. 2020. 10.1200/JCO.19.02931An important study demonstrating the viability of gemcitabine plus cisplatin in PDA patients with germline deficits in BRCA and PALB2. This study also suggested that though veliparib increased RR when combined with gemcitabine plus cisplatin in patients, it did not improve survival outcomes.
- 29.Pishvaian M, Wang H, Parenti S, et al. A phase I/II study of veliparib (Vel) in combination with 5-FU and oxaliplatin (FOLFOX) in patients with metastatic pancreatic cancer (mPDAC). J Clin Oncol. 2019;37(15):S4015. [Google Scholar]
- 30.•.Marechal A, Zou L. DNA damage sensing by the ATM and ATR kinases. Cold Spring Harb Perspect Biol. 5(9):a012716.A review which highlights the critical stimuli which trigger ATR and ATM along with the mechanisms and targets of the DDR-coordinating kinases.
- 31.Dreyer S, Paulus-Hock V, Lampraki EM, et al. Defining DDR defectiveness in pancreatic cancer. J Clin Oncol. 2017;35(suppl;abstr 4115). [Google Scholar]
- 32.Morton J, Timpson P, Karim S, et al. Mutant p53 drives metastasis and overcomes growth arrest/senescence in pancreatic cancer. Proc Natl Acad Sci U S A. 2010;107(1):246–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Lal S, Zarei M, Chand S, et al. WEE1 inhibition in pancreatic cancer cells is dependent on DNA repair status in a context dependent manner. Sci Rep. 2016;6:33323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Burki T AZD1775 plus chemoradiotherapy for pancreatic cancer. Lancet Oncol. 2019;20(9):e472. [DOI] [PubMed] [Google Scholar]

