Abstract
Metastatic pancreatic adenocarcinoma (PDAC) is a major cause of cancer-related mortality in 2021. Cytotoxic therapies are the therapeutic mainstay for PDAC. The recent approval of olaparib as maintenance therapy for germline BRCA1/2-mutated PDAC and pembrolizumab for mismatch repair deficient PDAC represent molecularly targeted approaches for this disease. Investigational therapeutic strategies include targeting the stroma, metabolism, tumor microenvironment and the immune system and selected approaches are reviewed herein.
Keywords: Pancreatic cancer, clinical trials, metastatic disease, genetics, KRAS
Introduction
Pancreatic cancer accounts for 57,600 new cancer diagnoses and 47,050 cancer deaths annually in the United States, representing a major cause of cancer associated mortality.[1] Metastatic or locally advanced disease represents 80-85% of new diagnoses,[2] and five-year overall survival (OS) in the metastatic setting is approximately 3%.[1] To address this significant unmet clinical need, many clinical trials of novel therapeutic approaches are currently ongoing.
Current Standards of Care & Emerging Combination Cytotoxic Therapies
Despite recent advances in molecular oncology and tumor immunology, multi-agent cytotoxic chemotherapy remains the mainstay of first-line therapy for metastatic pancreatic cancer. For patients with good functional status (ECOG 2 or less), two evidence-based chemotherapy options in the first line setting are FOLFIRINOX (folinic acid, 5-fluorouracil, irinotecan, and oxaliplatin) and gemcitabine with albumin-bound paclitaxel (nab-paclitaxel). FOLFIRINOX was associated with a significant OS benefit compared to single agent gemcitabine in a randomized phase III PRODIGE/ACCORD 11 study of 342 patients with ECOG performance status of 0 or 1.[3] Median overall survival (OS) in patients randomized to FOLFIRINOX was 11.1 months compared to 6.8 months in the gemcitabine arm (hazard ratio (HR) 0.57; 95% confidence interval (CI) 0.45 to 0.73; p < 0.001). Median progression-free survival (PFS) was 6.4 months in the FOLFIRINOX group and 3.3 months in the gemcitabine group (HR 0.47; 95% CI, 0.37 to 0.59, p < 0.001). The objective response rate (ORR) in the FOLIFIRNOX group was 31.6% compared to 9.4% in the gemcitabine group (p < 0.001). Neutropenia (46%), fatigue (24%), and vomiting (15%) were the most common grade 3 or 4 adverse effects observed in the FOLFIRINOX arm, also including 5.4% with febrile neutropenia. Similarly, in the phase III MPACT (Metastatic Pancreas Adenocarcinoma Clinical Trial) randomized trial of 861 patients compared combination gemcitabine and nab-paclitaxel to single agent gemcitabine; median OS in the combination arm was 8.5 months v. 6.7 months in the gemcitabine monotherapy arm (HR 0.72, 95% CI 0.62 to 0.83, p < 0.001).[4] Common grade 3-4 side effects observed with gemcitabine nab-paclitaxel included peripheral neutropenia (38%), leukopenia (31%), fatigue (17%), and peripheral neuropathy (17%). Febrile neutropenia occurred in 3% of patients.
While there are no published randomized trials in the metastatic setting directly comparing FOLFIRINOX to gemcitabine nab-paclitaxel, a recent systematic review and meta-analysis identified a weighted OS benefit of 1.15 months associated with FOLFIRINOX (95% CI 0.08-2.22, p = 0.03), though whole-population OS (HR 0.99, 95% CI 0.84-1.16; p = 0.9), PFS (HR 0.88, 95% CI 0.71-1.1, p = 0.26) and response rates (RR; 24% v 25%; odds ratio (OR) 0.93, 95% CI 0.64-1.36, p = 0.71) were not significantly different between the two regimens.[5] In the setting of resectable disease, the randomized phase II South West Oncology Group (SWOG) S1505 trial demonstrated similar disease-free survival following resection with perioperative mFOLFIRINOX and gemcitabine nab-paclitaxel (10.9 v. 14.2 months, p = 0.87).[6] The ongoing Precision Promise Trial (NCT04229004), an adaptive trial platform established by the Pancreatic Cancer Action Network, is currently randomizing patients to these two regimens in the first line metastatic setting. This and other selected pending trials in advanced PDAC are highlighted in Table 1.
Table 1.
Selected Ongoing Trials in Advanced Pancreas Cancer
| Trial | Phase | Disease Setting | Planned Enrollment |
Investigational Arm(s) | Control Arm | Primary Outcome(s) |
Expected Completion |
|---|---|---|---|---|---|---|---|
| Combination and Novel Chemotherapy | |||||||
| NCT04083235 | III | First line / untreated PDAC | 750 | Liposomal irinotecan + Oxaliplatin + 5-FU/LV | Gemcitabine + nab-paclitaxel | OS | December 2023 |
| NCT04229004 (Precision Promise) | II/III | First line / untreated PDAC | 8251 | Gemcitabine + nab-paclitaxel | mFOLFIRINOX | OS | February 2024 |
| NCT03610100 (ACELARATE) | III | First line / untreated PDAC | 328 | Acelarin (NUC-1031) | Gemcitabine | OS | December 2022 |
| NCT04469556 (PASS-01) | II | First line / untreated PDAC | 150 | Gemcitabine + nab-paclitaxel | mFOLFIRINOX | PFS | September 2023 |
| NCT04233866 | II | First line / untreated PDAC in patients over 70 years old | 184 | Liposomal irinotecan + 5-FU/LV | Gemcitabine + nab-paclitaxel Q2W | OS | December 2023 |
| NCT03126435 | III | Previously treated PDAC, after progression on FOLIFIRINOX | 218 | EndoTAG-1 + gemcitabine | Gemcitabine | OS | June 2022 |
| HRD-Mutated Cancers | |||||||
| NCT02677038 | II | Previously treated PDAC with suggestive family history or somatic/germline HRD mutation; germline BRCA1/2 excluded | 34 | Olaparib | N/A | ORR | November 2022 |
| NCT03553004 (NIRA-PANC) | II | Previously treated PDAC, with germline or somatic HRD mutations | 18 | Niraparib | N/A | ORR | February 2025 |
| NCT03601923 | II | Previously treated PDAC, with germline or somatic HRD mutations | 32 | Niraparib with small-field palliative radiation | N/A | PFS | February 2025 |
| NCT03404960 (PARPVAX) | Ib/II | Maintenance / PDAC responding to platinum-based chemotherapy | 84 | Niraparib + Nivolumab; Niraparib + Ipilimumab | N/A | PFS | June 2021 |
| Targeting Oncogenic Drivers | |||||||
| NCT04132505 | I | Previously treated PDAC | 39 | Binimetinib + hydroxychloroquine | N/A | MTD | May 2020 |
| NCT04006301 | I | Solid tumors with KRAS G12C mutations | 10 | JNJ-74699157 | N/A | Safety; ORR | July 2020 |
| NCT03785249 (KRYSTAL-1) | I/II | Solid tumors with KRAS G12C mutations | 391 | MRTX849 monotherapy; MRTX849 + pembrolizumab; MRTX849 + afatinib; MRTX849 + cetuximab | N/A | Safety; PK; ORR | September 2021 |
| NCT03215511 | I/II | Solid tumors with NTRK fusions | 170 | Selitrectinib | N/A | MTD; ORR | February 2022 |
| NCT02912949 | I/II | Solid tumors with NRG1 fusions | 250 | Zenocutuzumab | N/A | Safety; ORR; DOR | September 2022 |
| NCT04214418 (MEKiAUTO) | I/II | Solid tumors with KRAS mutations | 175 | Cobimetinib + Hydroxychloroquine; Cobimetinib + Hydroxychlorquine + Atezolizumab | N/A | MTD | September 2023 |
| NCT03600883 (CodeBreak 100) | I/II | Solid tumors with KRAS G12C mutations | 533 | AMG 510 | N/A | Safety; ORR | March 2024 |
| NCT02576431 | II | Solid tumors with NTRK fusions | 203 | Larotrectinib | N/A | ORR | September 2025 |
| Target Cell Surface Markers | |||||||
| NCT02672917 | I | Solid tumors expressing Ca 19-9 | 108 | MVT-5873 | N/A | Safety; MTD | December 2020 |
| Immunotherapy | |||||||
| NCT02620865 | Ib/II | Maintenance / PDAC responding to chemotherapy | 39 | Anti-CD3 x anti-EGFR bispecific antibody armed T cells (BATs); aldesleukin and sargramostim | N/A | OS | December 2020 |
| NCT03214250 (PRINCE) | Ib/II | First line / untreated PDAC | 129 | Gemcitabine + nab-paclitaxel + either nivolumab, APX005M, or nivolumab + APX005M | N/A | Safety; OS | September 2022 |
| NCT04329949 (RELIANT) | III | Third line / previously treated PDAC | 80 | Relacorilant + nab-paclitaxel | N/A | ORR | January 2022 |
| NCT03193190 (Morpheus - Pancreatic Cancer) | Ib/II |
Cohort 1: First line / Untreated PDAC Cohort 2: Second line / Previously treated PDAC |
290 |
Cohort 1: Atezolizumab + gemcitabine + nab-paclitaxel + either selicrelumab, bevacizumab, AB928, tiragolumab, or tocilizumab Cohort 2: Atezolizumab + either Cobimetinib, PEGPH20, BL-8040, or RO687428 (Q2W or Q3W) |
Cohort 1: Gemcitabine + nab-paclitaxel Cohort 2: Either mFOLFIRINOX or gemcitabine + nab-paclitaxel |
ORR; Safety | November 2021 |
| NCT04104672 | I | First line / untreated PDAC | 150 | AB680; AB680 + zimberelimab; gemcitabine + nab-paclitaxel + either AB680 or AB680 + zimberelimab | N/A | Safety | January 2024 |
| NCT03745326 | I/II | Previously treated solid tumor expressing KRAS G12D and HLA-A*11:01 | 70 | Cyclophosphamide and fludarabine + anti-KRAS G12D mTCR PBL + aldesleukin | N/A | Safety; ORR | December 2028 |
| NCT03190941 | I/II | Previously treated solid tumor expressing KRAS G12V and HLA-A*11:01 | 110 | Cyclophosphamide and fludarabine + anti-KRAS G12V mTCR PBL + aldesleukin | N/A | Safety; ORR | December 2028 |
| Modulating the Tumor Microenvironment | |||||||
| NCT03941093 (LAPIS) | III | Locally advanced, untreated PDAC | 256 | Pamrevlumab (FG-3019) + gemcitabine + nab-paclitaxel | Gemcitabine + nab-paclitaxel | OS; proportion of R0/ R1 resection | December 2023 |
| Metabolic Therapies | |||||||
| NCT03504423 (AVENGER 500) | III | First line / untreated PDAC | 528 | Devimistat (CPI-613) + mFOLIFIRINOX | FOLFIRINOX | PFS | December 2021 |
| NCT03665441 (TRYBECA-1) | III | Second line / previously treated PDAC | 500 | Eryaspase + either FOLFIRI, gemcitabine + nab-paclitaxel, or irinotecan freebase + 5-FU/LV | FOLFIRI; gemcitabine + nab-paclitaxel; irinotecan freebase + 5-FU/LV | OS | October 2021 |
| NCT04229004 | II/III | Second line / Previously treated PDAC | 8251 | SM-88 + methoxsalen + phenytoin + sirolimus | mFOLFIRINOX; gemcitabine + nab-paclitaxel2 | OS | February 2024 |
825 patients is the estimated total enrollment across all arms in this multi-arm platform trial.
Patients will receive whichever regimen they did not receive in the first-line setting
Abbreviations: LV = leucovorin; OS = overall survival; PFS = progression-free survival; Q2W = every two weeks; HRD = homologous repair deficiency ORR = overall response rate; DOR = duration of response; PK = pharmacokinetics; MTD = maximum tolerated dose; mTCR = murine T cell receptor; PBL = peripheral blood lymphocytes;
Given the cumulative toxicity of both first-line chemotherapy regimens, investigators have evaluated the role of de-escalating to lower-intensity ‘maintenance’ therapy regimens in patients with response or stable disease. In a randomized phase II study (PRODIGE 35 PANOPTIMOX), four months of FOLFIRINOX followed by maintenance 5-FU and leucovorin was associated with similar PFS (6.3 v. 5.7 months, HR not reported) compared to six months of continuous FOLFIRINOX, though rates of grade 3 or 4 neurotoxicity in the maintenance arm were higher (19% v. 10%, OR not reported) due to higher cumulative doses of oxaliplatin. [7]
Following disease progression on first line therapy, one FDA-approved chemotherapy regimen in the second line setting is the combination of 5-fluorouracil, leucovorin, and nano-liposomal irinotecan. In the randomized Phase III NAPOLI-1 study, this regimen was associated with an increased median OS (6.1 months) compared to 5-FU with leucovorin (4.2 months; HR 0.67, 95% CI 0.49-0.29, p = 0.012). There was no significant difference in median OS between nano-liposomal monotherapy and 5-FU with leucovorin (4.9 v. 4.2 months; HR 0.99, 95% CI 0.77-1.28, p = 0.94). [8] Alternatively, for fit patients whose disease progressed on first line FOLFIRINOX, current National Comprehensive Cancer Network (NCCN) and American Society of Clinical Oncology (ASCO) guidelines also support the use of combination gemcitabine and nab-paclitaxel.[9,10] Supporting this approach, a prospective French multicenter cohort study of 57 patients whose tumors progressed on FOLFIRINOX demonstrated a ORR of 17% and a DCR of 58% with gemcitabine nab-paclitaxel, with a median PFS of 5.1 months (95% CI 3.2-6.2 months).[11] In patients with ECOG performance status of greater than 2, single agent gemcitabine or 5-FU can be considered in the second line setting.
New combination chemotherapy regimens are currently being investigated in the first and second line setting. A single-arm phase1/2 study of nano-liposomal irinotecan, 5-FU, leucovorin, and oxaliplatin (NALIRIFOX) in previously untreated patients demonstrated promising median PFS of 9.2 months and median OS of 12.6 months.[12] These findings led to the fast-track designation by the FDA in June 2020, and a randomized phase III trial (NAPOLI-3) comparing NALIRIFOX to gemcitabine nab-paclitaxel is currently underway (NCT04083235).[13] Another emerging first-line chemotherapy regimen is one of the addition of cisplatin to gemcitabine and nab-paclitaxel. In a single arm phase Ib/II study of 25 patients, this combination was associated with an ORR of 71% and a median OS of 16.4 months, albeit also associated with significant myelosuppression.[14]
Exploiting Homologous Repair Deficiency
Recently published NCCN[9] and ASCO[15] guidelines recommend up-front germline and somatic genetic testing prior to initiating therapy to 1) identify potential second-line or maintenance therapy options and 2) identify screening needs and counseling regarding other family members of patients in whom an inherited cancer syndrome is identified.
A major impetus for genetic testing is the identification of patients who harbor either somatic or germline mutations in genes that govern homologous recombination, including BRCA1, BRCA2, PALB2, and ATM. The DNA-damage repair (DDR) pathway plays a major role in maintaining genomic integrity.[16] Mutations in core homologous repair deficiency (HRD) genes were identified in 19% of a series of 262 PDAC specimens; among this population germline HRD mutations were approximately three times more frequently compared to somatic HRD mutations.[17] Though results of genetic testing are often not available prior to initiating first-line therapy, there is emerging retrospective evidence that patients with HRD mutations treated with platinum-based chemotherapy have improved outcomes compared to patients without these mutations.[17,18] In the aforementioned series of 262 patients with PDAC, among patients treated with first-line platinum based chemotherapy, those with HRD mutations had improved PFS compared to those that did not (HR 0.44; 95% CI 0.29-0.67, p < 0.01). As such, first-line therapy with FOLFIRINOX may be preferred first-line regimen in patients whose personal or family history is consistent with a known or suggestive of a potential germline HRD-related cancer syndrome.
Following response or stability to first-line platinum-based chemotherapy, maintenance therapy with the poly ADP-ribose polymerase (PARP) olaparib was recently FDA-approved for patients with germline BRCA1/2 mutations in late 2019. This approval was based on the Phase III POLO (Pancreas Olaparib Ongoing) trial, which demonstrated an improved median PFS in patients randomized to olaparib (7.4 months) compared to those randomized to placebo (3.8 months) following platinum-based chemotherapy (HR 0.53; 95% CI 0.35 to 0.82, p = 0.004). Two limitations of this study were that olaparib was compared to placebo as opposed to maintenance chemotherapy, and that overall survival data had not matured at the time of original publication—these data are expected in early 2021.[19] In the first line setting, the combination of the PARP inhibitor veliparib with gemcitabine and cisplatin did not lead to any significant increase in ORR (74 v. 65%, p = 0.55) or PFS (10.1 v. 9.7 months, p = 0.6) compared to gemcitabine and cisplatin alone for patients with germline BRCA1/2 or PALB2 mutations; however the survival data for cisplatin/gemcitabine were encouraging with approximately 30% of patients alive at 2 years and 18% at three years. The addition of veliparib was associated with increased hematologic toxicity including neutropenia, thrombocytopenia, and anemia.[20] The NCCN guidelines endorse either FOLFIRINOX or cisplatin and gemcitabine as therapy in the HRD disease setting.
Ongoing studies in this population include a Phase II trial of niraparib in pretreated metastatic PDAC with germline or somatic HRD mutations (NCT03553004), as well as combinations of PARP inhibitors with anti-angiogenic agents, ATR inhibitors, and checkpoint blockade. This work builds on prior small trials of single-agent olaparib,[21] rucaparib [22]and veliparib[23] in previously treated, BRCA1/2-mutated PDAC, which respectively were associated with ORRs of 21%, 16% and 0%. Bolstering the rationale for combining PARP inhibition with immunotherapeutic approaches, a small retrospective series of seven patients with HRD-mutated PDAC treated with combination ipilimumab/nivolumab demonstrated one complete response and one partial response among three evaluable patients.[24] Additionally, an upcoming study will evaluate autologous stem cell transplant with myeloablative conditioning germline BRCA1/2 with low-volume disease following first-line chemotherapy (NCT pending).
Targeting Oncogenic Drivers
In addition to identifying patients with germline and somatic HRD mutations, genetic testing of PDAC affords clinicians opportunity to identify other potentially actionable driver mutations. Of 189 patients who were identified to have an actionable driver mutation through a retrospective analysis of the Know Your Tumor registry, those that received molecularly targeted therapy matched to an actionable finding had significantly longer median OS compared to those that did not receive matched therapy (2.58 v. 1.51 years; HR 0.42; 95% CI 0.26-0.68; p = 0.0004) and compared to those who did not have an actionable finding (2.58 v. 1.32 years; HR 0.34; 95% CI 0.22-0.53; p < 0.0001).[25] Of significant note, the population with actionable drivers represented a minority of the 677 patients who were screened and had adequate follow up information for analysis.
The most common oncogenic driver in PDAC is the GTPase KRAS, which is mutated in 90% of pancreatic cancers.[26] The most frequently represented KRAS mutation is point mutation, leading to the substitution of an aspartic acid for a glycine at position 12 (KRAS G12D), which leads to structural changes that impair its ability to hydrolyze GTP.[27] As mutated KRAS has long been a challenging pharmacologic target, initial strategies sought to inhibit downstream and complementary signaling pathways. In the SWOG S1155 phase II randomized trial, patients with PDAC whose cancer had progressed on first-line gemcitabine-based therapy were randomized to either a combination of the oral MEK inhibitor selumetinib with MK-2206, an oral AKT inhibitor, or modified FOLFOX (mFOLFOX). Median PFS was shorter with selumetinib and MK-2206 (1.9 v. 2.0 months; HR 1.61, 95% CI, 1.07-2.43; P = .02), and median OS was favored mFOLFOX (3.9 v. 6.7 months; HR, 1.37; 95% CI, 0.90-2.08; P = .15).[28] More recently, targeted inhibition of the guanine exchange factor SOS1 has been shown to limit the formation of GTP-loaded K-Ras and lead to the regression of KRAS-driven cancers in combination with MEK inhibitors in preclinical models across multiple KRAS mutations.[29] While targeted therapies for patients with KRAS G12C are the furthest along in clinical development, this alternate strategy of combined SOS and MEK inhibition could provide an approach for patients whose tumors express other KRAS mutations (e.g., KRAS G12D, KRAS G12V) and will shortly be tested in the clinic.
For patients with KRAS G12C mutations, allele-specific targeted inhibitors such as sotorasib[30] (NCT03600883), MRTX849[31] (NCT03785249), and JNJ74699157 (NCT04006301) have been recently evaluated in a variety of solid tumors. KRAS G12C has been reported in 1-2% of Caucasian populations with PDAC,[32] though it is potentially more common in Asian populations.[33] It remains unclear whether targeting KRAS G12C will have comparable efficacy in PDAC compared to others solid tumors. In the Phase I trial of sotorasib there was an ORR of 9% (i.e., one partial response) among the 11 evaluable treated patients with PDAC—this was considerably lower than the ORR of 32% observed in the 59 treated patients with non-small cell lung cancer.[34] While these data provide some early proof-of-concept evidence that KRAS G12C inhibition may have some clinical activity in PDAC, monotherapy with a single agent may be insufficient for the majority of patients whose tumor harbor this mutation.
One possible resistance mechanism to K-Ras inhibition in PDAC is autophagy, a lysosome-mediated repurposing of cellular contents as energy sources (i.e., “self eating”). In preclinical models, inhibition of KRAS or ERK-mediated signaling increased metabolic flux through autophagy pathways.[35] Combined inhibition of ERK and autophagy mediators has been identified as a potential therapeutic strategy to overcome this resistance. In a single patient with KRAS mutated PDAC, off-label use of the MEK inhibitor trametinib and the autophagy inhibitor hydroxychloroquine was associated with in a 50% reduction in tumor burden.[36] An ongoing phase I trial with the MEK inhibitor binimetinib and hydroxychloroquine (NCT04132505) will provide additional data on this approach. Additionally, the ongoing phase 1/2 MEKiAUTO study of cobimetinib, hydroxychloroquine, and atezolizumab in advanced KRAS-mutated malignancies (NCT NCT04214418) will provide data on the combination of MEK and autophagy inhibition with immune checkpoint blockade.
Upstream of small molecule inhibition of KRAS and its signaling partners, an alternative approach is to target cognate KRAS mRNA for degradation. However, successful drug delivery of small interfering RNAs (siRNAs) has historically remained a major therapeutic challenge due to enzymatic systemic degradation. Recently, there has been preclinical evidence of sustained release of KRAS G12D siRNA from a biodegradable polymeric matrix and inhibition of tumor growth in orthotopic and xenograft mouse models of PDAC.[37] This technology, termed, KRAS G12D siRNA Local Drug EluteR, (LODER), is currently being tested in combination with chemotherapy in the PROTACT trial of locally advanced PDAC (NCT01676259).[38] In this trial, siRNA-loaded particles are inserted into the tumor endoscopically. A phase III trial of this agent is planned with standard of care chemotherapy in locally advanced PDAC.
KRAS wild-type tumors are identified in a minority of patients, and are more common in patients diagnosed under 50 years old.[39,40] These tumors are more likely to harbor other potentially targetable driver mutations, including mutations or fusions involving BRAF, ALK, NTRK, and NRG-1. There have been early signs of clinical efficacy of targeted agents in this albeit uncommon setting. For instance, BRAF is mutated in 3-4% of pancreatic cancers exclusive to KRAS[41]; partial responses have been reported in two cases of BRAF-mutated PDAC with trametinib[42] and dabrafenib[43]. In a study of 3,170 patients with PDAC, ALK rearrangements exclusive of KRAS were identified in less than 0.2% of tumors. Treatment with ALK inhibitors led to SD as best outcome in three of four patients with evaluable responses[44].
NTRK fusions are associated with constitutive activation of Ras and Akt pathways and have recently gained attention in non-small cell lung cancer. In PDAC, the NTRK inhibitors entrectinib[45] and larotrectinib[46] have both been associated with partial responses and both drugs have disease-agnostic Food and Drug Administration (FDA) approval. Ongoing studies of larotrectinib (NCT02576431) and selitrectinib, a next-generation TRK inhibitor, (NCT03215511) are currently recruiting patients. Similarly rare NRG-1 fusions are found in 0.5% of PDAC, and lead to increased HER2/HER3 heterodimerization and activation of downstream PI3K pathways.[47] In a proof of concept study, treatment of three patients with NRG-1 fused PDAC with the bispecific HER2/HER3 antibody zenocutuzumab led to durable partial responses.[48] A phase II basket trial of zenocutuzumab in multiple tumor types is currently ongoing (NCT02912949)[49].
Targeted Therapies against Cell Surface Markers
Another class of targeted therapies under development leverage the unique surface markers expressed on the PDAC cell membrane. In clinical practice, carbohydrate antigen 19-9 (CA 19-9) is the most commonly used serologic marker used to track PDAC disease activity. CA 19-9 contains the glycan known as sialyl Lewis A (sLea), which is expressed on PDAC epithelial surfaces and aberrantly secreted into the bloodstream.[50] BNT321 (MVT-5873) is a fully human IgG1 monoclonal antibody targeting sialyl Lewis A, and has been shown in preclinical models to lead to complement mediated and antibody dependent cytotoxicity[51]. This antibody is currently being studied in single-arm Phase I study of PDAC and other Ca 19-9 expressing (NCT02672917).[52] Diagnostically, immuno-PET imaging using a zirconium-tagged radiolabeled antibody against Ca 19-9 was reported to be safe and to demonstrate an encouraging imaging signal in a cohort of twelve patients.[53] While clinical data using this approach are preliminary, it is possible that Ca 19-9 targeted PET may provide an highly sensitive method to detect primary pancreatic tumors and metastases and a neoadjuvant imaging trial is shortly to commence (NCT pending).[53]
Tight junction protein claudin 18.2 is aberrantly expressed in a subset of PDAC.[54] In mouse xenograft models of PDAC, the anti-claudin 18.2 monoclonal antibody, Zolbetuximab, slowed tumor growth and attenuated the formation of lung metastases.[54] Zolbetuximab is currently being studied in a randomized Phase II trial in combination with gemcitabine and nab-paclitaxel (NCT03816163) in patients whose tumors express claudin 18.2 by immunohistochemistry.
Immunotherapeutic Approaches
While checkpoint blockade has transformed the treatment of other solid tumors, limited responses have been observed to date in patients with PDAC with single agent inhibitors. Low neoantigen load paired with an immunosuppressive tumor microenvironment have been postulated as potential hurdles limiting the efficacy of immunotherapy in this disease.[55]
Currently, there are two FDA-approved indications for the anti-PD-1 monoclonal antibody pembrolizumab relevant to a subset of patients with metastatic PDAC. In 2018, pembrolizumab was approved for use in mismatch-repair deficient (MMR-D) tumors agnostic of tissue type. The mismatch repair pathway plays a critical role in maintaining genome integrity,[56] and is controlled by the proteins expressed by the genes MLH1, MSH2, MSH6, and PMS2. It is thought that the increased number of neoantigens expressed in MMR-D tumors provides additional potential targets for the immune system to recognize.[57] Clinically, MMR-D status can be identified by the identification of di- or tri-nucleotide repeats,[58] targeted sequencing of the MMR genes,[59] or immunohistochemical staining for the proteins they encode.[60]
Significant clinical responses were initially observed in a Phase II trial of 41 patients with a variety of MMR-D solid tumors treated with pembrolizumab.[61] MMR-D status in PDAC is rare, occurring in less than 1% of cancers,[62] and up to 83% of patients with MMR-D PDAC are found to have germline mutations genes associated with Lynch syndrome.[63] In the Phase II KEYNOTE-158 trial of 233 patients with MMR-D non-colorectal cancer,[64] the overall response rate (ORR) for 22 patients with MMR-D PDAC was 18%. This was considerably lower than the 34% ORR observed the entire trial population, and lower than the 33% ORR observed in MMR-D colorectal cancer in the KEYNOTE-164 trial.[65] Further investigation is needed to understand why responses are less frequent and less durable in PDAC compared to other MMR-D tumors.
Following the approval of pembrolizumab in MMR-D tumors, it also was approved in 2020 for patients with a tumor mutational burden (TMB) of greater than 10 mutations per megabase (mut/Mb). In the KEYNOTE-158 trial, an ORR of 29% was observed in a population of 102 patients with TMB ≥ 10 mut/Mb.[66] Notably, there were no patients with PDAC included in this study and TMB is relatively low in PDAC, typically 1-3 Mut/Mb,[62,67] potentially limiting the extension of these findings to most patients with PDAC.
In unselected populations of patients with PDAC, checkpoint blockade with and without concomitant chemotherapy has not been associated with improved outcomes. In a Phase II trial of 27 patients with advanced PDAC, monotherapy with CTLA-4 antibody ipilimumab was associated with no objective responses by RECIST criteria, though one patient who was treated beyond progression experienced delayed regression of the primary tumor as well as multiple hepatic metastases.[68] Similarly, a single-arm Phase Ib dose-escalation trial of combination gemcitabine and ipilimumab in 21 patients with advanced PDAC demonstrated an ORR of 14%, which was considered similar to historical response rates to gemcitabine monotherapy.[69] It was further noted that the three responders in this study had a median response duration of 11 months, and correlative studies characterizing these patients were not reported.
Additionally, small trials have investigated the combination of PD-1 blockade with combination chemotherapy. A phase Ib/II study of 19 patients with PDAC treated with pembrolizumab, gemcitabine, and nab-paclitaxel did not meet its pre-specified primary endpoint of complete response > 15% in 15 patients with evaluable responses.[70] A phase I trial of 50 patients with PDAC treated with nivolumab, gemcitabine, and nab-paclitaxel demonstrated an ORR of 18% (95% CI 8.6-31.4%) with a median PFS of 5.5 months (95% CI 3.3 – 7.2 months) in the first line setting, which did not lead to further investigation.[71] Combination of CTLA-4 and PD-1 blockade with durvalumab and tremelimumab were associated with a low ORR (3.1%, 95% CI 0.1 −16.2%) in the second line setting,[72] and there were no objective responses (ORR 0%, 95% CI 0 – 10.6%) in patients randomized to durvalumab monotherapy in this trial. In a Canadian Clinical Trials Group (CCTG PA.7) randomized phase II trial of previously untreated PDAC, durvalumab and tremelimumab in combination with gemcitabine and nab-paclitaxel was not associated with improved median OS compared to gemcitabine and nab-paclitaxel alone (9.8 v. 8.8 months, HR 0.94, 90% CI 0.71-1.25, p = 0.72).[73] The currently enrolling MORPHEUS-Pancreatic Cancer platform study is evaluating multiple atezolizumab and gemcitabine nab-paclitaxel combinations with several novel immune targets, inhibitors of the adenosine pathway and anti-vascular therapies, randomized against a control chemotherapy alone arm (NCT03193190).
Novel immune modulators are currently being studied in combination with established checkpoint blockade agents in an effort to overcome the immune suppressive microenvironment of pancreatic cancer. In preclinical models, interactions between CD40 expressed on antigen presenting cells and its ligand CD154 has been shown to play a key role in T cell priming to alloantigens.[74] The CD40 agonist APX005M demonstrated an acceptable safety profile and an encouraging efficacy signal in a select patient population in a phase Ib trial in combination with gemcitabine and nab-paclitaxel with APX005M and with/without nivolumab. Among 23 treated patients with evaluable responses, there were 14 (58%) patients with partial responses, eight with stable disease, and one with progressive disease. Additionally, on-treatment samples of peripheral blood mononuclear cells demonstrated remodeling of the myeloid compartment .[75] A randomized phase II study (NCT03214250) is further investigating these combinations and results are anticipated in 2021. The CXCR-4 antagonist motixafortide (BL-8040) has been demonstrated to increase T cell infiltration into the tumor microenvironment.[76] In the phase IIa COMBAT trial of previously treated patients with PDAC, combination motixafortide and pembrolizumab were associated with a disease control rate (DCR) of 34.5% and a DCR of 77% when the two agents were combined with nano-liposomal irinotecan and 5-fluoruracil. Additionally, an increased number of tumor infiltrating lymphocytes and activated CD8+ granzyme B+ cytotoxic T cells were observed in a limited number of patients with pre- and post-treatment biopsies.[77]
Other approaches directly target CD11b+ myeloid-derived suppressor cells (MDSCs), which are thought to abrogate T cell mediated tumor killing. In preclinical models, the CD11b modulator GB1275 was shown to reduce MDSCs in the tumor microenvironment, repolarize macrophages from the M2 to the M1 phenotype, and increase tumor infiltrating lymphocytes.[78] The ongoing KEYNOTE-A36 will investigate the efficacy of this agent in combination with gemcitabine and nab-paclitaxel in untreated stage IV PDAC.[79] Another strategy to downregulate the activity of MDSCs and immunosuppressive M2 macrophages is to decrease intratumoral levels of adenosine through the inhibition of CD73.[80] An ongoing trial with the CD73 inhibitor AB680 in combination with the anti-PD-1 antibody zimberelimab, gemcitabine, and nab-paclitaxel will evaluate the efficacy of this approach in the first line setting (NCT04104672).
Cellular therapies are also being actively investigated in PDAC. A phase Ib study of autologous T cells treated with a bispecific antibody targeting anti-CD3 and anti-EGFR (“BATs”) is currently enrolling patients[81] (NCT02620865); this strategy exploits the overexpression of EGFR observed in up to 50% of PDAC tumors.[82] Another cell therapy approach under investigation is the transduction of peripheral blood lymphocytes with murine T cell receptors targeting G12D and G12V mutated KRAS (NCT03745326; NCT03190941).
Modulating the Tumor Microenvironment
The extensive desmoplastic and hypovascular stroma associated with PDAC has long been postulated as a major barrier to effective immunotherapeutic and cytotoxic therapies. Agents that specifically target the stroma have been studied in combination with other therapies, though this strategy has yet to yield significant improvements in clinical outcomes.
Disappointingly, the randomized phase III HALO109-301 trial of the hyaluronidase pegvorhyaluronidase alfa (PEGPH20) in combination with gemcitabine and nab paclitaxel did not demonstrate any significant improvement in median OS compared to placebo (11.2 v. 11.5 months; HR 1.00, 95% CI 0.80-1.27, p = 0.97).[83] More than a decade ago, the addition of anti-vascular endothelial growth factor (VEGF) antibody bevacizumab to gemcitabine in the Phase III Cancer and Leukemia Group B (CALGB) 80303 trial did not lead to any improvement in median OS compared to gemcitabine monotherapy (5.8 v. 5.9 months, p = 0.95 months)[84]. In an ongoing phase III trial, pamrevlumab, a monoclonal antibody against connective tissue growth factor (CTGF), is being administered in combination with gemcitabine and nab-paclitaxel in locally advanced PDAC (NCT03941093); pending these results, further development in the metastatic setting may be forthcoming. Promisingly, pamrevlumab has previously been shown to reduce the rate of decline in forced vital capacity compared to placebo in a randomized trial of 103 patients with idiopathic pulmonary fibrosis (−2.9% v. −7.2% at 48 weeks; p =0.033),[85] though the relevance of this finding to the fibrotic PDAC tumor microenvironment has yet to be determined.
An alternative approach is modulating the immune microenvironment as maintenance therapy, after response to initial chemotherapy. Sunitinib, a multi-targeted receptor tyrosine kinase inhibitor that inhibits VEGF receptors and platelet-derived growth factor receptors(PDGF-Rs), has been tested in this setting. In a small phase II PACT-12 trial, 56 patients with metastatic PDAC and no evidence of progression after six months of chemotherapy were randomized to either maintenance sunitinib or observation. The trial met its primary endpoint of six-month PFS, which was 22.2% (95% CI 6.2-38.2%) in the sunitinib arm and 3.6% (95% CI 0-10.6%; p < 0.01) in the arm undergoing observation.[86] For patients with HRD-mutated PDAC, combination of VEGF inhibition in combination with PARP inhibitors may be an attractive maintenance strategy given recent demonstration of a considerable PFS benefit with this approach in patients with HRD-mutated ovarian cancer. In the PAOLA-1 trial of 806 women with newly diagnosed ovarian cancer randomized 2:1 to olaparib and bevacizumab or bevacizumab alone following response to first-line chemotherapy with bevacizumab, PFS for all women in the combination olaparib and bevacizumab arm was 22.1 months while median PFS was 16.6 months in the bevacizumab alone group (HR 0.59; 95% CI 0.49-0.72, p < 0.001). The difference in PFS with combination therapy was more pronounced in women with HRD positive tumors (37.2 v. 17.7 months; HR 0.33; 95% CI 0.25-0.45). [87]
Targeting Metabolism
Recent advances in the understanding of cancer metabolism has led to the development of investigational therapeutics that seek to exploit unique metabolic vulnerabilities of cancer cells. One example is SM-88, a tyrosine analogue that interferes with protein synthesis and increases oxidative stress in PDAC cells. In a dose-finding trial of SM-88 monotherapy in a heavily pretreated group of 36 patients with PDAC, SM-88 was well tolerated and led to radiologic responses by RECIST or SUV uptake in 3 of 4 evaluable patients.[88] The expansion phase of this trial, which is actively recruiting, will compare SM-88 in combination with methoxsalen, phenytoin and sirolimus (MPS) against physician’s choice chemotherapy in the third-line setting. Additionally, in the ongoing Precision Promise multicenter platform trial, SM-88 and MPS is being randomized against either FOLFIRINOX or gemcitabine nab-paclitaxel in the second line setting (NCT04229004).
Two active trials are evaluating the addition of metabolic agents in combination with chemotherapy. The Phase III AVENGER 500 trial (NCT03504423)[89] has randomized 500 patients with untreated PDAC to first-line FOLFIRINOX or modified FOLFIRINOX with devimistat (CPI-613), a citric acid inhibitor that significantly disrupted mitochondrial metabolism in preclinical models of PDAC and non-small cell lung cancer.[90] Accrual has completed and results are expected in 2021. Additionally, the TRYBECA-01 (NCT03665441) trial is evaluating erythrocyte-encapsulated asparaginase (eryaspase) in combination with either gemcitabine nab-paclitaxel or FOLFIRI against gemcitabine nab paclitaxel in the second line setting. In a Phase IIb study of 141 patients, the combination of second-line eryaspase with either mFOLFOX6 or gemcitabine was associate with a significantly longer median OS (6.0 v. 4.4 months; HR 0.60; 95% CI 0.40-1.12; p = 0.008) and PFS (2.0 v. 1.6 months; HR 0.56; 95% CI 0.37-0.84; p = 0.005) compared to mFOLFOX6 or gemcitabine alone. [91]
Identifying PDAC Subtypes and Circulating Biomarkers
Going forward, an improved understanding of underlying PDAC biology may guide the development of therapies for more targeted patient populations. To this end, molecularly distinct PDAC subtypes have been identified and associated with clinical outcomes. An early analysis used pooled transcriptional data from multiple clinically annotated patient cohorts to define three distinct phenotypes: classical, quasi-mesenchymal, and exocrine-like. Of these subgroups, the classical phenotype was an independent predictor of favorable OS in a multivariate Cox regression (p = 0.024).[92] More recently, Moffitt et al. used transcriptional profiling of micro-dissected primary and metastatic PDAC specimens to identify two tumor-specific and two stroma-specific subtypes. Between the tumor-specific subtypes, patients with ‘basal-like’ tumors had worse median OS compared to patients with classical subtype tumors (11 v. 19 months, p = 0.007). The mRNA expression pattern of ‘basal-like’ tumors resembled those of basal-like breast and bladder tumors. Between the two stroma-specific subtypes, patients with an ‘activated’ stromal signature had shorter median OS compared to patients with a ‘normal’ stromal signature (15 v. 24 months, p = 0.019).[93]
Clinically, transcriptional profiling was prospectively incorporated into the COMPASS trial, which evaluated the feasibility of real-time whole-genome sequencing (WGS) and RNA sequencing (RNAseq) in parallel with first-line treatment for metastatic PDAC. Of 63 patients enrolled in the study, WGS and RNAseq were successfully performed prior to the first radiographic evaluation of treatment response in 98% and 95% of patients, respectively. As secondary endpoints, improved response to chemotherapy was identified in patients with classical subtype tumors (p = 0.004) and tumor expression of GATA6 was identified as a potential surrogate biomarker for distinguishing classical from basal subtype PDAC.[94] The ongoing PASS-01 trial (NCT04469556) is prospectively correlating transcriptional tumor profiling and GATA-6 expression to responses to chemotherapy in patients randomized to FOLFIRINOX or gemcitabine nab-paclitaxel in the first-line setting.
In addition to leveraging a refined understanding of PDAC subtypes, interrogation of blood-based biomarkers may afford new opportunities to define, stratify, and track clinical trial cohorts. Circulating tumor DNA (ctDNA) is an emerging technology used across a variety of tumor types to identify radiologically occult disease and identify mechanisms of resistance to targeted therapy (e.g., T790M in EGFR-mutated non-small cell lung cancer). In PDAC, a recent meta-analysis identified a sensitivity of 28% and a specificity of 95% for ctDNA as a diagnostic tool to distinguish 557 patients with biopsy-proven PDAC from 212 patients with benign pancreatic lesions. The same meta-analysis identifed an association between shorter OS and circulating KRAS ctDNA across all stages of pancreatic cancer (HR 1.92, 95% CI 1.15–3.22).[95] As molecularly targeted therapies are further developed in PDAC (e.g., against KRAS G12D), ctDNA may provide a more easily accessible means for serial sampling in a disease where repeat tissue sampling can be challenging.
In addition to cell-free ctDNA, circulating tumor cells (CTCs) may also provide accessible information about tumor biology, specifically with regard to predicting treatment response. In a prospective study of 50 patients with advanced or locally advanced PDAC, circulating tumor and invasive cells (CTICs) were isolated from peripheral blood through a collagen invasion assay. Pharmacogenomic profiling of CTIC DNA accurately predicted patient responses to chemotherapy and median PFS (10.4 v 3.6 months; HR = 0.14, p < 0.0001) and OS (17.2v 8.3 months; HR 0.29, p < 0.0249) were significantly higher in patients whom were treated with agents for which CTC analysis predicted response.[96] These findings were recapitulated in a separate prospective cohort of 37 patients with PDAC treated with gemcitabine and nab-paclitaxel and here median PFS was higher in patients whose CTICs predicted response to gemcitabine nab-paclitaxel compared to patients whose CTICs predicted response to FOLFIRINOX (8.7 v 5.2 months; HR 0.34, p = 0.0031). [97] An ongoing study will prospectively evaluate the utility of pharmacogenomic profiling of CTCs in 80 patients treated with either FOLFIRINOX or gemcitabine nab-paclitaxel (NCT03033927).
Lastly, a third potential class of circulating biomarkers are endosomes, or extracellular vesicles and particles (EVPs). EVPs are endosome-derived membrane-bound nanoparticles that carry contents specific to the tumor microenvironment throughout the bloodstream. Recently, investigators have identified EVP protein signatures specific to PDAC that could potentially be used as a tool for early detection of recurrence or for primary screening.[98]
Conclusions
Cytotoxic therapy with multidrug combinations represent a standard in all disease settings of PDAC. Recent insights into the molecular underpinnings of HRD-mutated and KRAS wild type PDAC have provided opportunity for targeting beyond cytotoxic therapy and several agents, e.g., olaparib and pembrolizumab are approved in genomically selected settings. Going forward, evolving insights into immune dysregulation, cancer metabolism, and the tumor microenvironment may yield additional novel therapeutic targets that will hopefully improve clinical outcomes. Additionally, more precise identification of patient populations through transcriptional subtyping and circulating biomarkers may lead to more personalized approaches for patients with this historically challenging disease entity.
Supplementary Material
Synopsis.
Pancreas ductal adenocarcinoma is a malignancy with a rising incidence and poor prognosis, nonetheless outcomes are slowly improving. Treatment strategies for advanced disease rely primarily on cytotoxic therapy. Important developments over the last decade include a deep understanding of the genetic context and pathobiology of pancreas cancer. These findings underpin the emerging era of molecular targeted therapies in this disease and this article describes the current standard practice for treating pancreas cancer and highlights emerging therapeutic directions including genomic and immune directed approaches.
Disclosures and funding sources
JWS reports no conflicts of interest. EOR Research Funding to institution (MSK): Genentech/Roche, Celgene/BMS, BioNTech, BioAtla, AstraZeneca, Arcus. Consulting Role: Cytomx Therapeutics (DSMB), Rafael Therapeutics (DSMB), Sobi, Molecular Templates, Boehringer Ingelheim, BioNTech, Ipsen, Merck, AstraZeneca, Bayer (family), Genentech-Roche (family), Celgene-BMS (family), Eisai (family)
Funding
Cancer Center Support Grant P30 CA 008748
David M. Rubenstein Center for Pancreas Cancer Research
Data Availability Statement
This article is a review article and there is no unpublished data presented.
Sources
- 1.Siegel RL, Miller KD, Jemal A: Cancer statistics, 2020. CA Cancer J Clin 2020;70:7–30. [DOI] [PubMed] [Google Scholar]
- 2.Mizrahi JD, Surana R, Valle JW, Shroff RT: Pancreatic cancer. Lancet 2020;395:2008–2020. [DOI] [PubMed] [Google Scholar]
- 3.Conroy T, Desseigne F, Ychou M, et al. : FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 2011;364:1817–1825. [DOI] [PubMed] [Google Scholar]
- 4.Von Hoff DD, Ervin T, Arena FP, et al. : Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 2013;369:1691–1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Pusceddu S, Ghidini M, Torchio M, et al. : Comparative Effectiveness of Gemcitabine plus Nab-Paclitaxel and FOLFIRINOX in the First-Line Setting of Metastatic Pancreatic Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel) 2019;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Sohal D, Duong MT, Ahmad SA, et al. : SWOG S1505: Results of perioperative chemotherapy (peri-op CTx) with mfolfirinox versus gemcitabine/nab-paclitaxel (Gem/nabP) for resectable pancreatic ductal adenocarcinoma (PDA). Journal of Clinical Oncology 2020;38:4504–4504. [Google Scholar]
- 7.Dahan L, Phelip J, Le Malicot K, et al. : FOLFIRINOX until progression, FOLFIRINOX with maintenance treatment, or sequential treatment with gemcitabine and FOLFIRI.3 for first-line treatment of metastatic pancreatic cancer: A randomized phase II trial (PRODIGE 35-PANOPTIMOX). J Clin Oncol 2018;36. [Google Scholar]
- 8.Wang-Gillam A, Li CP, Bodoky G, et al. : Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, open-label, phase 3 trial. Lancet 2016;387:545–557. [DOI] [PubMed] [Google Scholar]
- 9.Tempero MA: NCCN Guidelines Updates: Pancreatic Cancer. J Natl Compr Canc Netw 2019;17:603–605. [DOI] [PubMed] [Google Scholar]
- 10.National Comprehensive Cancer Network. Pancreatic Adenocarcinoma (Version 1.2021). 2020. [DOI] [PubMed] [Google Scholar]
- 11.Portal A, Pernot S, Tougeron D, et al. : Nab-paclitaxel plus gemcitabine for metastatic pancreatic adenocarcinoma after Folfirinox failure: an AGEO prospective multicentre cohort. Br J Cancer 2015;113:989–995. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Wainberg Z, Bekaii-Saab T, Boland PM, et al. : First-line liposomal irinotecan 5 fluorouracil/leucovorin oxaliplatin in patients with pancreatic ductal adenocarcinoma: Primary analysis from a phase 1/2 study. Annals of Oncology 2020;21:S241. [Google Scholar]
- 13.Wainberg Z, Bekaii-Saab T, Hubner R, et al. : NAPOLI-3: An open-label, randomized, phase III study of first-line liposomal irinotecan + 5-fluorouracil/leucovorin + oxaliplatin versus nab-paclitaxel + gemcitabine in patients with metastatic pancreatic ductal adenocarcinoma. Journal of Clinical Oncology 2020;38:TPS4661–TPS4661. [Google Scholar]
- 14.Jameson GS, Borazanci E, Babiker HM, et al. : Response Rate Following Albumin-Bound Paclitaxel Plus Gemcitabine Plus Cisplatin Treatment Among Patients With Advanced Pancreatic Cancer: A Phase 1b/2 Pilot Clinical Trial. JAMA Oncol 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Sohal DPS, Kennedy EB, Cinar P, et al. : Metastatic Pancreatic Cancer: ASCO Guideline Update. J Clin Oncol 2020:JCO2001364. [DOI] [PubMed] [Google Scholar]
- 16.Perkhofer L, Gout J, Roger E, et al. : DNA damage repair as a target in pancreatic cancer: state-of-the-art and future perspectives. Gut 2020:gutjnl-2019-319984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Park W, Chen J, Chou JF, et al. : Genomic Methods Identify Homologous Recombination Deficiency in Pancreas Adenocarcinoma and Optimize Treatment Selection. Clin Cancer Res 2020;26:3239–3247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Pishvaian MJ, Blais EM, Brody JR, 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. Journal of Clinical Oncology 2019;37. [DOI] [PubMed] [Google Scholar]
- 19.Golan T, Hammel P, Reni M, et al. : Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. N Engl J Med 2019;381:317–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.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;38:1378–1388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Kaufman B, Shapira-Frommer R, Schmutzler RK, et al. : Olaparib monotherapy in patients with advanced cancer and a germline BRCA1/2 mutation. J Clin Oncol 2015;33:244–250. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Shroff RT, Hendifar A, McWilliams RR, et al. : Rucaparib Monotherapy in Patients With Pancreatic Cancer and a Known Deleterious BRCA Mutation. JCO Precis Oncol 2018;2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lowery MA, Kelsen DP, Capanu M, et al. : Phase II trial of veliparib in patients with previously treated BRCA-mutated pancreas ductal adenocarcinoma. Eur J Cancer 2018;89:19–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Terrero G, Pollack T, Sussman D, et al. : Exceptional responses to ipilimumab/nivolumab (ipi/nivo) in patients (pts) with refractory pancreatic ductal adenocarcinoma (PDAC) and germline BRCA or RAD51 mutations. J Clin Oncol 2020;38:754. [Google Scholar]
- 25.Pishvaian MJ, Blais EM, Brody JR, et al. : Overall survival in patients with pancreatic cancer receiving matched therapies following molecular profiling: a retrospective analysis of the Know Your Tumor registry trial. Lancet Oncol 2020;21:508–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Biankin AV, Waddell N, Kassahn KS, et al. : Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 2012;491:399–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Vatansever S, Erman B, Gümüş ZH: Oncogenic G12D mutation alters local conformations and dynamics of K-Ras. Scientific Reports 2019;9:11730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Chung V, McDonough S, Philip PA, et al. : Effect of Selumetinib and MK-2206 vs Oxaliplatin and Fluorouracil in Patients With Metastatic Pancreatic Cancer After Prior Therapy: SWOG S1115 Study Randomized Clinical Trial. JAMA Oncol 2017;3:516–522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Hofmann MH, Gmachl M, Ramharter J, et al. : BI-3406, a potent and selective SOS1::KRAS interaction inhibitor, is effective in KRAS-driven cancers through combined MEK inhibition. Cancer Discov 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Canon J, Rex K, Saiki AY, et al. : The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature 2019;575:217–223. [DOI] [PubMed] [Google Scholar]
- 31.Hallin J, Engstrom LD, Hargis L, et al. : The KRAS(G12C) Inhibitor MRTX849 Provides Insight toward Therapeutic Susceptibility of KRAS-Mutant Cancers in Mouse Models and Patients. Cancer Discov 2020;10:54–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Forbes SA, Beare D, Gunasekaran P, et al. : COSMIC: exploring the world's knowledge of somatic mutations in human cancer. Nucleic Acids Res 2015;43:D805–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zhou L, Baba Y, Kitano Y, et al. : KRAS, BRAF, and PIK3CA mutations, and patient prognosis in 126 pancreatic cancers: pyrosequencing technology and literature review. Med Oncol 2016;33:32. [DOI] [PubMed] [Google Scholar]
- 34.Hong DS, Fakih MG, Strickler JH, et al. : KRAS(G12C) Inhibition with Sotorasib in Advanced Solid Tumors. N Engl J Med 2020;383:1207–1217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bryant KL, Stalnecker CA, Zeitouni D, et al. : Combination of ERK and autophagy inhibition as a treatment approach for pancreatic cancer. Nat Med 2019;25:628–640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kinsey CG, Camolotto SA, Boespflug AM, et al. : Protective autophagy elicited by RAF→MEK→ERK inhibition suggests a treatment strategy for RAS-driven cancers. Nat Med 2019;25:620–627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zorde Khvalevsky E, Gabai R, Rachmut IH, et al. : Mutant KRAS is a druggable target for pancreatic cancer. Proc Natl Acad Sci U S A 2013;110:20723–20728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Varghese A, Ang C, Dimaio C, et al. : A phase II study of siG12D-LODER in combination with chemotherapy in patients with locally advanced pancreatic cancer (PROTACT). Journal of Clinical Oncology 2020;38:TPS4672–TPS4672. [Google Scholar]
- 39.Varghese A, Singh I, Singh R, et al. : Young-onset pancreas cancer (PC) in patients less than or equal to 50 years old at Memorial Sloan Kettering (MSK): Descriptors, genomics, and outcomes. Journal of Clinical Oncology 2020;38:774–774. [Google Scholar]
- 40.Ben-Aharon I, Elkabets M, Pelossof R, et al. : Genomic Landscape of Pancreatic Adenocarcinoma in Younger versus Older Patients: Does Age Matter? Clin Cancer Res 2019;25:2185–2193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lowery MA, Jordan EJ, Basturk O, et al. : Real-Time Genomic Profiling of Pancreatic Ductal Adenocarcinoma: Potential Actionability and Correlation with Clinical Phenotype. Clin Cancer Res 2017;23:6094–6100. [DOI] [PubMed] [Google Scholar]
- 42.Aguirre AJ, Nowak JA, Camarda ND, et al. : Real-time Genomic Characterization of Advanced Pancreatic Cancer to Enable Precision Medicine. Cancer Discov 2018;8:1096–1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wrzeszczynski KO, Rahman S, Frank MO, et al. : Identification of targetable BRAF ΔN486_P490 variant by whole-genome sequencing leading to dabrafenib-induced remission of a BRAF-mutant pancreatic adenocarcinoma. Cold Spring Harb Mol Case Stud 2019;5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Singhi AD, Ali SM, Lacy J, et al. : Identification of Targetable ALK Rearrangements in Pancreatic Ductal Adenocarcinoma. J Natl Compr Canc Netw 2017;15:555–562. [DOI] [PubMed] [Google Scholar]
- 45.Pishvaian M, Diego Rolfo C, Liu S, et al. : Clinical benefit of entrectinib for patients with metastatic pancreatic cancer who harbor NTRK and ROS1 fusions. Journal of Clinical Oncology 2018;36:521–521. [Google Scholar]
- 46.O'Reilly EM, Hechtman JF: Tumour response to TRK inhibition in a patient with pancreatic adenocarcinoma harbouring an NTRK gene fusion. Ann Oncol 2019;30 Suppl 8:viii36–viii40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Fernandez-Cuesta L, Plenker D, Osada H, et al. : CD74–NRG1Fusions in Lung Adenocarcinoma. Cancer Discovery 2014;4:415–422. [DOI] [PubMed] [Google Scholar]
- 48.Schram A, O’Reilly E, Somwar R, et al. : Clinical proof-of-concept for MCLA-128, a bispecific HER2/3 antibody therapy, in NRG1 fusion-positive cancers. 2019. [Google Scholar]
- 49.Schram A, Drilon A, Macarulla T, et al. : A phase II basket study of MCLA-128, a bispecific antibody targeting the HER3 pathway, in NRG1 fusion-positive advanced solid tumors. Journal of Clinical Oncology 2020;38:TPS3654–TPS3654. [Google Scholar]
- 50.Munkley J: The glycosylation landscape of pancreatic cancer. Oncol Lett 2019;17:2569–2575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Sawada R, Sun SM, Wu X, et al. : Human monoclonal antibodies to sialyl-Lewis (CA19.9) with potent CDC, ADCC, and antitumor activity. Clin Cancer Res 2011;17:1024–1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.O'Reilly E, Borazanci E, Yu K, et al. : HuMab-5B1 (MVT-5873), a mAb targeting sLea, in combination with first-line gemcitabine plus nab-paclitaxel (gem/nab-P) for patients with pancreatic cancer (PDAC) and other CA19-9 positive malignancies. Journal of Clinical Oncology 2018;36:e16235–e16235. [Google Scholar]
- 53.Lohrmann C, O'Reilly EM, O'Donoghue JA, et al. : Retooling a Blood-Based Biomarker: Phase I Assessment of the High-Affinity CA19-9 Antibody HuMab-5B1 for Immuno-PET Imaging of Pancreatic Cancer. Clin Cancer Res 2019;25:7014–7023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Tureci, Mitnacht-Kraus R, Woll S, et al. : Characterization of zolbetuximab in pancreatic cancer models. Oncoimmunology 2019;8:e1523096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wu AA, Jaffee E, Lee V: Current Status of Immunotherapies for Treating Pancreatic Cancer. Curr Oncol Rep 2019;21:60. [DOI] [PubMed] [Google Scholar]
- 56.Pecina-Slaus N, Kafka A, Salamon I, Bukovac A: Mismatch Repair Pathway, Genome Stability and Cancer. Front Mol Biosci 2020;7:122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Le DT, Durham JN, Smith KN, et al. : Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017;357:409–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Zhang L: Immunohistochemistry versus microsatellite instability testing for screening colorectal cancer patients at risk for hereditary nonpolyposis colorectal cancer syndrome. Part II. The utility of microsatellite instability testing. J Mol Diagn 2008;10:301–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Schrader KA, Cheng DT, Joseph V, et al. : Germline Variants in Targeted Tumor Sequencing Using Matched Normal DNA. JAMA Oncol 2016;2:104–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Shia J: Immunohistochemistry versus microsatellite instability testing for screening colorectal cancer patients at risk for hereditary nonpolyposis colorectal cancer syndrome. Part I. The utility of immunohistochemistry. J Mol Diagn 2008;10:293–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Le DT, Uram JN, Wang H, et al. : PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med 2015;372:2509–2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Hu ZI, Shia J, Stadler ZK, et al. : Evaluating Mismatch Repair Deficiency in Pancreatic Adenocarcinoma: Challenges and Recommendations. Clin Cancer Res 2018;24:1326–1336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Latham A, Srinivasan P, Kemel Y, et al. : Microsatellite Instability Is Associated With the Presence of Lynch Syndrome Pan-Cancer. J Clin Oncol 2019;37:286–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Marabelle A, Le DT, Ascierto PA, et al. : Efficacy of Pembrolizumab in Patients With Noncolorectal High Microsatellite Instability/Mismatch Repair-Deficient Cancer: Results From the Phase II KEYNOTE-158 Study. J Clin Oncol 2020;38:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Le DT, Kim TW, Van Cutsem E, et al. : Phase II Open-Label Study of Pembrolizumab in Treatment-Refractory, Microsatellite Instability-High/Mismatch Repair-Deficient Metastatic Colorectal Cancer: KEYNOTE-164. J Clin Oncol 2020;38:11–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Marabelle A, Fakih M, Lopez J, et al. : Association of tumour mutational burden with outcomes in patients with advanced solid tumours treated with pembrolizumab: prospective biomarker analysis of the multicohort, open-label, phase 2 KEYNOTE-158 study. Lancet Oncol 2020;21:1353–1365. [DOI] [PubMed] [Google Scholar]
- 67.Castle JC, Uduman M, Pabla S, et al. : Mutation-Derived Neoantigens for Cancer Immunotherapy. Front Immunol 2019;10:1856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Royal RE, Levy C, Turner K, et al. : Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother 2010;33:828–833. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kamath SD, Kalyan A, Kircher S, et al. : Ipilimumab and Gemcitabine for Advanced Pancreatic Cancer: A Phase Ib Study. Oncologist 2020;25:e808–e815. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Weiss GJ, Blaydorn L, Beck J, et al. : Phase Ib/II study of gemcitabine, nab-paclitaxel, and pembrolizumab in metastatic pancreatic adenocarcinoma. Invest New Drugs 2018;36:96–102. [DOI] [PubMed] [Google Scholar]
- 71.Wainberg ZA, Hochster HS, Kim EJ, et al. : Open-label, Phase I Study of Nivolumab Combined with nab-Paclitaxel Plus Gemcitabine in Advanced Pancreatic Cancer. Clin Cancer Res 2020;26:4814–4822. [DOI] [PubMed] [Google Scholar]
- 72.O'Reilly EM, Oh DY, Dhani N, et al. : Durvalumab With or Without Tremelimumab for Patients With Metastatic Pancreatic Ductal Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncol 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Renouf DJ, Knox J, Kavan P, et al. : LBA65 The Canadian Cancer Trials Group PA.7 trial: Results of a randomized phase II study of gemcitabine (GEM) and nab-paclitaxel (Nab-P) vs GEM, nab-P, durvalumab (D) and tremelimumab (T) as first line therapy in metastatic pancreatic ductal adenocarcinoma (mPDAC). Annals of Oncology 2020;31. [Google Scholar]
- 74.Grewal IS, Flavell RA: CD40 and CD154 in cell-mediated immunity. Annu Rev Immunol 1998;16:111–135. [DOI] [PubMed] [Google Scholar]
- 75.O'Hara M, O'Reilly E, Rosemarie M, et al. : Abstract CT004: A Phase Ib study of CD40 agonistic monoclonal antibody APX005M together with gemcitabine (Gem) and nab-paclitaxel (NP) with or without nivolumab (Nivo) in untreated metastatic ductal pancreatic adenocarcinoma (PDAC) patients. Cancer Research 2019;79. [Google Scholar]
- 76.Seo YD, Jiang X, Sullivan KM, et al. : Mobilization of CD8(+) T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer. Clin Cancer Res 2019;25:3934–3945. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Bockorny B, Semenisty V, Macarulla T, et al. : BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: the COMBAT trial. Nat Med 2020;26:878–885. [DOI] [PubMed] [Google Scholar]
- 78.Panni RZ, Herndon JM, Zuo C, et al. : Agonism of CD11b reprograms innate immunity to sensitize pancreatic cancer to immunotherapies. Sci Transl Med 2019;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Wang-Gillam A, Rasco D, Park W, et al. : Abstract CT247: A phase 1/2 study of GB1275, a first-in-class CD11b modulator, as monotherapy and with an anti-PD-1 antibody in specified advanced solid tumors or with chemotherapy in metastatic pancreatic cancer (KEYNOTE-A36). Cancer Research 2020;80. [Google Scholar]
- 80.Zhang B: CD73: a novel target for cancer immunotherapy. Cancer Res 2010;70:6407–6411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Lum LG, Thakur A, Choi M, et al. : Clinical and immune responses to anti-CD3 x anti-EGFR bispecific antibody armed activated T cells (EGFR BATs) in pancreatic cancer patients. Oncoimmunology 2020;9:1773201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ueda S, Ogata S, Tsuda H, et al. : The correlation between cytoplasmic overexpression of epidermal growth factor receptor and tumor aggressiveness: poor prognosis in patients with pancreatic ductal adenocarcinoma. Pancreas 2004;29:e1–8. [DOI] [PubMed] [Google Scholar]
- 83.Van Cutsem E, Tempero MA, Sigal D, et al. : Randomized Phase III Trial of Pegvorhyaluronidase Alfa With Nab-Paclitaxel Plus Gemcitabine for Patients With Hyaluronan-High Metastatic Pancreatic Adenocarcinoma. J Clin Oncol 2020;38:3185–3194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kindler HL, Niedzwiecki D, Hollis D, et al. : Gemcitabine plus bevacizumab compared with gemcitabine plus placebo in patients with advanced pancreatic cancer: phase III trial of the Cancer and Leukemia Group B (CALGB 80303). J Clin Oncol 2010;28:3617–3622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Richeldi L, Fernández Pérez ER, Costabel U, et al. : Pamrevlumab, an anti-connective tissue growth factor therapy, for idiopathic pulmonary fibrosis (PRAISE): a phase 2, randomised, double-blind, placebo-controlled trial. Lancet Respir Med 2020;8:25–33. [DOI] [PubMed] [Google Scholar]
- 86.Reni M, Cereda S, Milella M, et al. : Maintenance sunitinib or observation in metastatic pancreatic adenocarcinoma: a phase II randomised trial. Eur J Cancer 2013;49:3609–3615. [DOI] [PubMed] [Google Scholar]
- 87.Ray-Coquard I, Pautier P, Pignata S, et al. : Olaparib plus Bevacizumab as First-Line Maintenance in Ovarian Cancer. N Engl J Med 2019;381:2416–2428. [DOI] [PubMed] [Google Scholar]
- 88.Noel MS, Wang-Gillam A, Ocean AJ, et al. : Phase II trial of SM-88 in patients with metastatic pancreatic cancer: Preliminary results of the first stage. Journal of Clinical Oncology 2019;37:200–200. [Google Scholar]
- 89.Philip PA, Buyse ME, Alistar AT, et al. : Avenger 500, a phase III open-label randomized trial of the combination of CPI-613 with modified FOLFIRINOX (mFFX) versus FOLFIRINOX (FFX) in patients with metastatic adenocarcinoma of the pancreas. Journal of Clinical Oncology 2019;37:TPS479–TPS479. [Google Scholar]
- 90.Zachar Z, Marecek J, Maturo C, et al. : Non-redox-active lipoate derivates disrupt cancer cell mitochondrial metabolism and are potent anticancer agents in vivo. J Mol Med (Berl) 2011;89:1137–1148. [DOI] [PubMed] [Google Scholar]
- 91.Hammel P, Fabienne P, Mineur L, et al. : Erythrocyte-encapsulated asparaginase (eryaspase) combined with chemotherapy in second-line treatment of advanced pancreatic cancer: An open-label, randomized Phase IIb trial. Eur J Cancer 2020;124:91–101. [DOI] [PubMed] [Google Scholar]
- 92.Collisson EA, Sadanandam A, Olson P, et al. : Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med 2011;17:500–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Moffitt RA, Marayati R, Flate EL, et al. : Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet 2015;47:1168–1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Aung KL, Fischer SE, Denroche RE, et al. : Genomics-Driven Precision Medicine for Advanced Pancreatic Cancer: Early Results from the COMPASS Trial. Clin Cancer Res 2018;24:1344–1354. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Creemers A, Krausz S, Strijker M, et al. : Clinical value of ctDNA in upper-GI cancers: A systematic review and meta-analysis. Biochim Biophys Acta Rev Cancer 2017;1868:394–403. [DOI] [PubMed] [Google Scholar]
- 96.Yu KH, Ricigliano M, Hidalgo M, et al. : Pharmacogenomic modeling of circulating tumor and invasive cells for prediction of chemotherapy response and resistance in pancreatic cancer. Clin Cancer Res 2014;20:5281–5289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Yu KH, Ricigliano M, McCarthy B, et al. : Circulating Tumor and Invasive Cell Gene Expression Profile Predicts Treatment Response and Survival in Pancreatic Adenocarcinoma. Cancers (Basel) 2018;10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Hoshino A, Kim HS, Bojmar L, et al. : Extracellular Vesicle and Particle Biomarkers Define Multiple Human Cancers. Cell 2020;182:1044–1061 e1018. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
This article is a review article and there is no unpublished data presented.
