Next Article in Journal
Ring Finger Protein 125 Is an Anti-Proliferative Tumor Suppressor in Hepatocellular Carcinoma
Next Article in Special Issue
The Next Frontier in Pancreatic Cancer: Targeting the Tumor Immune Milieu and Molecular Pathways
Previous Article in Journal
Inhibition of HDAC and Signal Transduction Pathways Induces Tight Junctions and Promotes Differentiation in p63-Positive Salivary Duct Adenocarcinoma
Previous Article in Special Issue
Hemodynamic, Surgical and Oncological Outcomes of 40 Distal Pancreatectomies with Celiac and Left Gastric Arteries Resection (DP CAR) without Arterial Reconstructions and Preoperative Embolization
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Systemic Therapy of Metastatic Pancreatic Adenocarcinoma: Current Status, Challenges, and Opportunities

1
Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
2
Division of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(11), 2588; https://doi.org/10.3390/cancers14112588
Submission received: 23 April 2022 / Revised: 19 May 2022 / Accepted: 23 May 2022 / Published: 24 May 2022
(This article belongs to the Special Issue Frontiers in Pancreatic Cancer)

Abstract

:

Simple Summary

Pancreatic ductal adenocarcinoma (PDAC) is a common type of cancer originating from the pancreatic glands and is characterized by a rapidly progressive course and a dismal prognosis. Currently, chemotherapy is the cornerstone of treatment that has modest efficacy. Targeted treatment options are rapidly evolving with the growing understanding of pathobiological underpinnings of tumorigenesis, progression, and treatment resistance mechanisms. Combination strategies targeting multiple signaling pathways supporting tumor growth and propagation are active areas of contemporary research that will likely transform the treatment paradigm of pancreatic cancer.

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy characterized by nonspecific presenting symptoms, lack of a screening test, rapidly progressive clinical course, and presentation with an advanced-stage disease in the majority of patients. PDAC is essentially a systemic disease irrespective of the initial stage, as most patients with non-metastatic PDAC undergoing curative-intent treatment eventually experience metastatic relapse. Currently, cytotoxic chemotherapy remains the cornerstone of treatment in patients with advanced disease. However, the current standard treatment with multiagent chemotherapy has modest efficacy and results in median overall survival (OS) of less than a year and a 5-year OS of about 10%. The pathobiology of PDAC poses many challenges, including a unique tumor microenvironment interfering with drug delivery, intratumoral heterogeneity, and a strongly immunosuppressive microenvironment that supports cancer growth. Recent research is exploring a wide range of novel therapeutic targets, including genomic alterations, tumor microenvironment, and tumor metabolism. The rapid evolution of tumor genome sequencing technologies paves the way for personalized, targeted therapies. The present review summarizes the current chemotherapeutic treatment paradigm of advanced PDAC and discusses the evolving novel targets that are being investigated in a myriad of clinical trials.

1. Introduction

Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy that originates in the exocrine pancreatic glands. PDAC is the second-most frequent malignancy arising from the digestive tract, with an estimated annual incidence of 60,430 in the United States of America (USA) [1] and 340,000 worldwide [2]. PDAC has several unique features, including the rapidly progressive nature of the tumor, the proximity of the pancreas to important vascular structures, often precluding curative resection, absence of typical presenting signs or symptoms that make the diagnosis and treatment difficult [3]. PDAC is the fourth leading cause of cancer-related death in the USA [1], and the aggressive biology of PDAC is reflected in its poor 5-year relative survival rate of 10.8% [4]. It is important to note that the incidence of PDAC is increasing at such a pace that PDAC is projected to be the second leading cause of cancer mortality by the year 2030 [5]. Furthermore, approximately 80% of patients with localized PDAC harbor micrometastatic disease, evidenced by a high rate of cancer relapse in patients with localized PDAC receiving curative-intent treatment [6]. These data underscore the urgent need for effective systemic therapies for PDAC.
Chemotherapy is currently the cornerstone of systemic therapy for patients with metastatic PDAC (mPDAC), which results in median overall survival (mOS) of less than a year [7,8]. The evolution of systemic therapy for pancreatic cancer has been rather gradual, from gemcitabine monotherapy [9,10,11,12] to the current standard of combination chemotherapy with triplet [7] or doublet agents [8]. However, multidrug chemotherapy regimens have been associated with modest survival gain and significant toxicities limiting their utility. Advancements in the understanding of tumor biology, accelerated by the rapid evolution of genomic sequencing technologies, have led to newer therapies, including immunotherapy and targeted therapies, that hold significant promise. The present review provides a detailed account of the current treatment strategies and developing therapeutic paradigms that have the potential to transform the treatment of advanced pancreatic cancer dramatically.

2. Advanced PDAC: Current Standard of Care

Advanced PDAC is incurable, and hence the treatment goals include controlling the tumor growth to prolong life, controlling symptoms, and maintaining the quality of life. The survival benefit of systemic chemotherapy over best supportive care has long been established in patients with mPDAC [13,14]. The current standard treatment consists of multiagent chemotherapy regimens in patients with good Eastern Cooperative Oncology Group (ECOG) performance status (PS), although the toxicity limits the utility of such regimens. Consequently, maintenance therapy following an initial period of induction therapy is being investigated. Immunotherapy has been accepted as a standard treatment in a minority of patients with deficient mismatch repair (dMMR) tumors.

2.1. First-Line Chemotherapy

Gemcitabine monotherapy was established as the standard of care for patients with mPDAC in the 1990s based on a randomized trial. In this trial, treatment-naïve patients with mPDAC received single-agent gemcitabine vs. 5-FU. The mOS improved modestly with gemcitabine (5.65 months vs. 4.41 months with 5-FU, p = 0.0025) [10]. Although a subsequent study with a combination of gemcitabine and erlotinib, an epidermal growth factor receptor (EGFR) inhibitor, showed a slightly better mOS compared to gemcitabine monotherapy (6.2 months vs. 5.9 months) [15], single-agent gemcitabine remained the standard first-line chemotherapy until 2011 owing to significant toxicities associated with the gemcitabine-erlotinib combination for a relatively small clinical benefit. Gemcitabine monotherapy is currently reserved for patients with an ECOG PS of 2 or higher or comorbidities precluding multiagent chemotherapy regimens. Table 1 summarizes the first-line studies.
In treatment-naïve patients of mPDAC with good ECOG PS, two multiagent regimens, FOLFIRONOX (5-FU, irinotecan, and oxaliplatin) and gemcitabine/nab-paclitaxel (GnP), are currently the preferred regimens. PRODIGE 4, a phase III randomized trial, compared FOLFIRINOX with gemcitabine monotherapy in treatment-naïve patients with mPDAC and reported a significant survival benefit with FOLFIRINOX among 342 treatment-naïve patients (mOS: 11.1 vs. 6.8 months, 95% CI, 0.45–0.73; p < 0.001) [7]. The overall response rate (ORR) and median progression-free survival (mPFS) also favored FOLFIRINOX (ORR: 31.6% vs. 9.4%, mPFS: 6.4 vs. 3.3 months). FOLFIRINOX, however, was associated with significantly higher rate of grade 3 or 4 neutropenia (45.7% vs. 21%), febrile neutropenia (5.4% vs. 1.2%), diarrhea (12.7% vs. 1.8%), and sensory neuropathy (9% vs. 0%) compared to gemcitabine. The role of GnP in the first-line setting was established by a phase III trial that randomly assigned 861 previously untreated patients to either GnP or gemcitabine [8]. The GnP regimen was associated with significantly improved mOS (8.5 vs. 6.7 months, HR 0.72, p < 0.001), mPFS (5.5 vs. 3.7 months, HR 0.69, p < 0.001), and ORR (23% vs. 7%, HR 3.19, p < 0.001). However, grade 3 or higher toxicities were more frequent with GnP (neutropenia 38% vs. 27%, peripheral neuropathy 17% vs. 1%, and diarrhea 6% vs. 1%). It should be noted that, unlike the PRODIGE4 study, this trial enrolled over 60% of patients from the USA with relatively less robust performance scores (Karnofsky performance score of 70 or more). In addition, there was no age limit for participation in this trial.
Concerns related to the toxicities with FOLFIRINOX triggered several subsequent studies with modified FOLFIRINOX regimens. A prospective, single-arm, phase II study evaluated a modified FOLFIRINOX regimen (oxaliplatin 85 mg/m2, irinotecan 135 mg/m2, 5-FU 300 mg/m2 bolus, followed by 2400 mg/m2 continuous infusion for 46 h every 14 days) in patients with metastatic and locally advanced PDAC (n = 31) [19]. The study reported a much lower incidence of Grade 3 or 4 neutropenia and diarrhea, 12.2% and 16.2%, respectively. Furthermore, the efficacy appeared to be comparable to the PRODIGE 4 study results with an mOS of 10.2 months (95% CI, 7.6 to 14.3), an mPFS of 6.1 months (95% CI, 5.1 to 8.3), and an ORR of 35.1%. Another retrospective study with metastatic and nonmetastatic PDAC (n = 60) patients evaluated a different version of modified FOLFIRINOX regimen that consisted of oxaliplatin 85 mg/m2, irinotecan 180 mg/m2, and 5-FU 2400 mg/m2 infusion over 46 h without a 5-FU bolus (n = 60) [20]. The mOS and mPFS were 9 months (95% CI, 7.1 to not estimable) and 8.5 months (95% CI, 3.7 to 11), respectively. The ORR was 30%, with the rate of grade 4 neutropenia, grade 3/4 diarrhea, and fatigue of 3%, 13%, and 13%, respectively. Subsequently, a large meta-analysis that included 1461 patients in 32 studies showed no difference in mOS, mPFS, and ORR between the standard FOLFIRINOX regimen utilized in the PRODIGE4 study and the modified FOLFIRINOX regimens [21]. Based on these studies, modified FOLFIRINOX regimens have been adopted widely in routine clinical practice.
A prospective randomized trial has not been completed to date comparing FOLFIRINOX with GnP in untreated patients with mPDAC. However, several retrospective studies have suggested similar efficacy and safety with both regimens [22,23]. A meta-analysis of 16 retrospective studies that included 3813 patients (1690 patients treated with FOLFIRINOX and 2123 patients treated with GnP) reported similar mOS (HR 0.99; 95% CI, 0.84–1.16; p = 0.9), mPFS (HR 0.88; 95% CI, 0.71–1.1; p = 0.26), and ORR (25% with GnP vs. 24% with FOLFIRINOX) with both regimens [24]. However, a median weighted OS difference favored FOLFIRINOX (mean difference: 1.15, 95% CI, 0.08–2.22, p = 0.03), and the toxicity profile was different, grade 3/4 neutropenia, nausea, and febrile neutropenia were higher with FOLFIRINOX, while neurotoxicity and anemia were higher with GnP. Two-phase III randomized trials are currently underway comparing modified FOLFIRINOX and GnP in treatment-naïve patients with mPDAC (NCT04229004 and NCT04469556).
A single-arm, open-label, phase 1b/2 study investigated the role of a triplet regimen (cisplatin, gemcitabine, and nab-paclitaxel) in 25 patients with previously untreated mPDAC and reported an ORR of 71%, a disease control rate (DCR) of 88%, mOS of 16.4 months (95% CI, 10.2–25.3), and mPFS of 10.1 (95% CI, 6.0–12.5) months [16]. Based on this study, a single-arm phase II open-label study (NCT03915444) with the triplet regimen in untreated mPDAC patients has fully accrued recently, the results of which are pending at this time.
An open-label, phase I/II study has recently evaluated the safety and efficacy of NALIRIFOX (liposomal irinotecan + 5-FU + leucovorin + oxaliplatin) in patients with mPDAC in the first-line setting [18]. Among the 32 patients who received the maximum tolerated dose (MTD) that consisted of liposomal irinotecan 50 mg/m2, oxaliplatin 60 mg/m2, 5-fluorouracil 2400 mg/m2, and leucovorin 400 mg/m2 every 2 weeks, the ORR, mPFS, and mOS were 34.4%, 9.2 months (95% CI, 7.69–11.96), and 12.6 months (95% CI, 8.74–18.69), respectively. In this study population, 22 of 32 had grade ≥3 treatment-related adverse events (TRAEs), including neutropenia (31.3%), febrile neutropenia (12.5%), and hypokalemia (12.5%). A phase III randomized study (NAPOLI 3) is currently underway, comparing NALIRIFOX to GnP in the first-line setting (NCT 04083235).
Germline mutations in the genes that are critical for the homologous recombination repair of DNA, including BRCA1/2 and PALB2 genes, are well-known predisposing factors for developing pancreatic cancer [25]. Approximately 5% to 9% of patients with PDAC harbor mutations in BRCA1/2 or PALB2 genes [25]. Previous studies have reported that mutations in BRCA1/2 and PALB2 genes are associated with increased sensitivity to platinum agents and synthetically lethal interaction with polyadenosine diphosphate-ribose polymerase (PARP) inhibitor (PARPi) [26]. Studies demonstrating a high response rate with cisplatin, gemcitabine, and a PARPi veliparib in patients with mPDAC [27,28] led to a randomized phase II trial to evaluate the efficacy of gemcitabine, cisplatin with or without veliparib in mPDAC patients with germline BRCA/PALB2 mutations [17]. Although this study did not show improved efficacy with the addition of veliparib, the ORR, mPFS, and mOS were impressive with the cisplatin and gemcitabine combination, with an ORR of 65.2%, an mPFS of 9.7 months, and an mOS of 16.4 months. This study established cisplatin and gemcitabine combination as one of the first-line treatment options in PDAC patients harboring BRCA/PALB2 mutations.
GABRINOX, a prospective, single-arm phase II study, evaluated a sequential strategy in which 58 treatment-naïve patients with mPDAC received GnP (day 1, 8, and 15) followed by FOLFIRINOX (day 29 and 43) [29]. The regimen was well-tolerated, and the patients received a median of 4 (1–9) cycles in 8.5 months (0.5–19.8 months). The study reported an impressive ORR of 64.9% (95% CI, 51.1 to 77.1%), including 3.5% of CR, an mPFS of 10.5 months, and an mOS of 15.1 months. Of note, the incidence of grade 3 or higher peripheral neuropathy was remarkably low (5.2%).

2.2. Maintenance Therapy

Toxicities, especially neurotoxicities and cytopenias, are major barriers to the long-standing use of FOLFIRINOX or GnP in advanced pancreatic cancer patients. Consequently, several clinical trials have evaluated de-escalation after a period of induction therapy termed maintenance therapy. The FDA approval of maintenance treatment with a PARP inhibitor, olaparib, for adult mPDAC patients with deleterious germline BRCA mutations who have not progressed on the first-line platinum-based chemotherapy has brought the discussion around maintenance therapy in advanced pancreatic cancer to the forefront. The FDA approval of olaparib was based on the POLO trial [30], a phase III study in which 154 patients with mPDAC harboring germline BRCA 1 or 2 mutations remaining progression-free on the first-line platinum-containing regimen for at least 16 weeks were randomized to olaparib or placebo (92 patients to olaparib and 62 patients to placebo) [30]. The mPFS in the olaparib group was significantly longer than in the placebo group (7.4 months vs. 3.8 months; HR: 0.53; 95% CI, 0.35 to 0.82; p = 0.004). However, the final mOS was similar between the two arms- 19 months in the olaparib arm vs. 19.2 months in the placebo arm, p = 0.3487 [31]. Of note, the 36-month OS rate was higher in the olaparib arm (33.9% vs. 17.8%). The FDA approval of olaparib maintenance therapy based on the POLO trial data faced several critiques, including a comparison of olaparib maintenance with the placebo rather than more commonly practiced maintenance chemotherapy (for example, FOLFIRI [5-FU plus irinotecan] or single-agent 5-FU), and the 16 weeks of induction chemotherapy has been viewed as a rather short duration of induction therapy, and the lack of OS benefit. It is important to note that the study was not adequately powered to detect a difference in OS and 26% of patients in the placebo arm subsequently received a PARP inhibitor, potentially extending the OS in the placebo arm, although cross-over was not allowed in the trial. The other important consideration is that 40% of patients in the olaparib group experienced grade 3 or higher toxicities (most commonly anemia, fatigue, and decreased appetite), and toxicities led to discontinuation of olaparib in 5% of patients [30]. A similar study reported significant activity of another PARP inhibitor, rucaparib, as maintenance therapy in advanced PDAC patients with germline or somatic pathogenic variant mutation in BRCA1, BRCA2, or PALB2 [32]. Eligible patients received a minimum of 16 weeks of platinum-based chemotherapy without any evidence of platinum resistance, followed by rucaparib 600 mg orally twice a day until progression. The PFS at 6 months (primary endpoint) was 59.5% (95% CI, 44.6 to 74.4), median PFS was 13.1 months (95% CI, 4.4 to 21.8), and median OS was 23.5 months (95% CI, 20 to 27).
The other maintenance therapy strategies include induction FOLFIRINOX followed by 5-FU [33,34] or capecitabine or FOLFIRI [33] or FOLFOX [33], induction GnP followed by gemcitabine maintenance [35], and maintenance sunitinib after first-line induction chemotherapy [36]. The optimal maintenance strategy, however, is not defined. The studies evaluating various maintenance treatment strategies are summarized in Table 2.

2.3. 2nd Line Chemotherapy

Approximately one-third to half of the patients with mPDAC progressing on first-line chemotherapy received second-line chemotherapy [37]. The prospective data supporting the benefit of second-line chemotherapy over best supportive care (BSC) are sparse. A phase III study by the German CONKO study group randomized patients with mPDAC progressing on gemcitabine to OFF (5-FU, folinic acid, and oxaliplatin) plus BSC or BSC alone. The patient group receiving OFF plus BSC had a significantly longer median survival from the initiation of second-line treatment (4.8 vs. 2.3 months) and also from the initiation of first-line gemcitabine (9 vs. 7.9 months, p = 0.031). Of note, a survival benefit with second-line treatment after progression on currently available multiagent first-line regimens (FOLFIRINOX or GA) is not supported by any prospective data.
The decision to pursue second-line therapy and the choice of therapy depend on a variety of factors, including performance status, residual toxicity from previous treatment, first-line therapy administered, comorbidities, presence of targetable genomic alterations, and patient preference.
The second-line chemotherapy regimens can be divided into two broad groups—(1) regimens for patients who progressed on FOLFIRINOX and (2) regimens for patients who progressed on first-line gemcitabine-based regimens (Table 3). After progression on FOLFIRINOX, treatment with GnP is feasible, as evaluated in two trials [38,39], although neuropathy could be an important barrier. Single-agent gemcitabine is a reasonable option in patients with significant residual neuropathy or inadequate performance status [40].
After progression on first-line gemcitabine, a wide variety of regimens are available, including liposomal irinotecan plus 5-FU/leucovorin combination [41], OFF (oxaliplatin, 5-FU/LV) [42], modified FOLFOX [43], modified FOLFIRINOX [44,45], FOLFIRI [46], XELOX [47], and docetaxel plus oxaliplatin [48] combination. Although none of these regimens is clearly superior to others, a meta-analysis that compared the efficacy of adding oxaliplatin vs. irinotecan formulations to a fluoropyrimidine after progression on first-line gemcitabine reported that the combination of a fluoropyrimidine plus irinotecan significantly improved both mPFS and mOS (HR: 0.7; 95% CI, 0.55–0.89), while oxaliplatin-based combinations modestly improved PFS but not the OS [49]. Table 3 summarizes important second-line studies in mPDAC.

3. Oligometastatic Disease

A subset of patients with mPDAC has limited metastatic disease, termed the oligometastatic disease, and may derive benefit from aggressive systemic therapy combined with resection of metastatic tumors. Although a well-agreed definition of oligometastatic pancreatic cancer does not exist, a proposed definition [50] includes the following criteria: (1) four or fewer hepatic or pulmonary metastatic lesions, (2) CA 19–9 of less than 1000 U/mol, (3) patients who responded to systemic chemotherapy. Approximately 8% of patients with mPDAC satisfy the criteria for oligometastatic disease [51]. The role of metastasectomy has not been well defined in mPDAC and should be restricted to select patients. The best candidates are patients who achieve prolonged radiologic and biochemical responses and have low-volume, lung-only metastases.
The optimum treatment protocol of oligometastatic mPDAC is not established. A systematic review and meta-analysis of six observational retrospective studies that included 2087 patients reported higher survival rates in patients who received chemotherapy followed by surgery vs. patients who received chemotherapy alone [52], with an mOS of 23 to 56 months after surgery compared to 11 to 16.4 months after chemotherapy alone. In this systematic review, FOLFIRINOX was found to be the most common chemotherapy regimen utilized. A single-center retrospective study (n = 85) reported an mOS of 12.3 months and a 5-year survival of 8.1% [53]. Another single-center retrospective study (n = 78) suggested that oligometastatic mPDAC with lung only metastasis has an indolent course, and the mOS was prolonged in patients who underwent surgery or stereotactic radiotherapy (67.5 months) vs. patients who received chemotherapy only (33.8 months) or observation (29.9 months) [54].

4. Therapies Targeting Genomic Alterations

The most common genomic alteration in PDAC is Kirsten rat sarcoma viral oncogene homolog (KRAS) mutation which is present in ~90% of tumors [55]. Point mutations at G12 (98%), G13 (1%), or Q61 (1%) residues of KRAS lead to the constitutive activation of RAS and its downstream signaling pathways leading to tumor development and maintenance [55]. The direct targeting of KRAS or its downstream pathway is a major active area of research in PDAC.

4.1. Targeting KRAS

KRAS G12C, accounting for <2% of KRAS mutations in PDAC [56], can be targeted with covalent inhibitors that lock KRAS G12C in its inactive GDP bound form [56]. Adagrasib (MRTX849) is one such inhibitor with promising preliminary results among PDAC patients in the ongoing KRYSTAL-1 trial [57]. Among the 10 evaluable PDAC patients enrolled in the KRYSTAL-1 study, adagrasib 600 mg twice daily was associated with an ORR of 50%, disease control rate (DCR) of 100%, an mPFS of 6.6 months (95% CI, 1.0–9.7), and 50% of these patients have ongoing responses at the time of data analysis (median follow up 8.1 months) [57]. Sotorasib (AMG510), another drug with the same mechanism of action which is FDA approved for KRAS G12C mutated non-small cell lung cancer, showed modest, short-lived responses in PDAC patients treated as part of phase I/II CodeBreaK100 trial (NCT03600883). Among the 38 PDAC patients at a median follow-up of 16.8 months, ORR was 21%, DCR 84.2%, and mPFS of 3.98 months [58]. Preclinical synergy studies and understanding of the resistance mechanisms led the field of KRAS G12C inhibition to combination therapies such as EGFR inhibitors and/or immune checkpoint inhibitors (ICIs) [59] (Table 4).
While the drugs targeting KRAS G12C are more advanced in the field of drug development, KRAS G12D (~50%), G12V (~30%), and G12R (~10%) are the more common KRAS mutations in PDAC. Many studies support the notion that not all KRAS mutations are the same [60,61]. Understanding the similarities and differences can help design KRAS mutation-selective and pan-mutant inhibitors. Table 4 presents examples of mutant-specific and mutant nonspecific studies targeting KRAS mutations.

4.2. Targeting the Downstream Pathway of KRAS

The constitutive activation of the MAPK pathway is a hallmark of PDAC. Targeting MEK alone has not led to meaningful and/or durable responses in PDAC [62]. Preclinical studies suggested increased dependency of PDAC on autophagy in the context of MEK or ERK inhibition [63,64]. Furthermore, both MEK/ERK and autophagy inhibition are linked to the enhanced efficacy of ICIs in PDAC [65,66]. Multiple trials are evaluating the role of the combination of MEK or ERK inhibition with autophagy inhibition and/or ICIs (Table 4).

4.3. KRAS-Wild Type Tumors

KRAS-wild type tumors represent ~10% of PDAC tumors [67]. These tumors are rich in potentially actionable alterations. BRAF alterations are common in this population and can be targeted with BRAF, MEK, or BRAF plus MEK inhibitors [68]. NRG1 fusion, an alteration with a predilection for younger KRAS-Wild type patients [69], can be targeted by pan-ERBB inhibitors such as Afatinib. Fusions in genes such as RET, ALK, ROS1, NTRK 1/2/3, MET, FGFR 1–3, and mutations in EGFR are among the other alterations with available targeted therapies [67].

4.4. Somatic Mutations in the Homologous Recombination Repair (HRR) Pathway

Patients with somatic mutations in the HRR pathway genes leading to homologous recombination deficiency (HRD), specifically if biallelic (defined as pathogenic mutations in both alleles of the HRR gene or presence of a pathogenic mutation in one allele and loss of the wild-type allele), can also benefit from treatment with platinum-based chemotherapy [70]. Among the patients with PDAC, the prevalence of HRD varies depending on the testing method—14% to 16% with targeted next-generation sequencing and up to 24% to 44% with whole exome and genome sequencing [71]. Efforts are ongoing to standardize the definition of HRD in PDAC, expand access to tests that detect HRD [68], and find the best biomarker that can predict response to platinum, PARP inhibitors, and other DNA damaging agents (Table 4).

4.5. Cyclin-Dependent Kinase Inhibitor 2A (CDKN2A) Alterations

CDKN2A is a tumor suppressor gene located on chromosome 9p. CDKN2A alteration is frequent in PDAC. Nearly 29% of PDAC patients harbor CDKN2A deletions, and 21% harbor CDKN2A mutations [72]. Alterations in CDKN2A/B are associated with CDK4/6 upregulation. Monotherapy with CDK4/6 inhibitor palbociclib did not result in meaningful responses in the TAPUR study [73]. However, the combination of CDK4/6 inhibitor plus MEK inhibitor has demonstrated preliminary activity in patients with concomitant KRAS and CDKN2A/B alterations [74].
The methylthioadenosine phosphorylase (MTAP) gene, encoding an enzyme crucial for methionine and adenine salvage, is in close proximity to the CDKN2A gene. Deletions of CDKN2A are therefore frequently associated with co-deletions of MTAP. MTAP loss results in the accumulation of methyladenosine, which in turn results in partial inhibition of the activity of the protein arginine methyltransferase 5 (PRMT5) [75]. MTAP deficient cells are susceptible to PRMT5 inhibitors or compounds that reduce the activity of PRMT5 protein, such as methionine adenosyltransferase 2a (MAT2A) inhibitors [76]. AG-270 is a MAT2A inhibitor in clinical trials in PDAC (Table 4).

5. Immunotherapy

The only form of mPDAC that responds well to immunotherapy with an immune checkpoint inhibitor (ICI) is the tumor harboring mismatch repair deficiency (dMMR), leading to high microsatellite instability (MSI-H). However, the MSI-H signature is rare in PDAC, occurring only in <1% of patients and most often associated with Lynch syndrome [77,78]. Robust responses to ICIs have been documented in MSI-H pancreatic cancer patients [79,80,81,82], with an ORR varying from 18.2% [82] to 77% [79]. Currently, pembrolizumab, a PD-1 blocker, is approved by the FDA for patients with chemotherapy-refractory advanced MSI-H/dMMR PDAC.
As most pancreatic tumors lack a MSI-H signature and the tumor microenvironment (TME) is heavily infiltrated by immunosuppressive cell populations, including tumor-associated macrophages(TAMs), regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and neutrophils that enable cancer cells to evade the immune system, currently available immunotherapies have been proven to be ineffective in pancreatic cancer [83]. Several trials have confirmed that the activity of ICI monotherapy in mPDAC is disappointing. A phase II trial with 27 patients of mPDAC showed no objective responses to ipilimumab, an anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA4) agent [84]. A randomized phase II study with durvalumab (anti-PD-L1 drug) with or without tremelimumab (anti-CTLA4 agent) in the second-line setting also reported a disappointing response-ORR of 3.1% with the combination therapy and 0% with the durvalumab monotherapy [85]. Conversely, treatment with a combination of ipilimumab and nivolumab in patients with Homologous Recombination Deficiency (HRD) Pathogenic Germline Variants has shown encouraging preliminary results. In a small retrospective series in mostly PDAC patients with HRD, treatment with ipilimumab/nivolumab combination resulted in an objective response rate of 42%, with a disease control rate of 58%. Four patients achieved CR without evidence of disease progression 11 to 41 months after the initiation of immunotherapy [86].
Several strategies are being investigated to recruit the immune system against pancreatic tumors. One of the promising strategies is targeting CD40 with agonist antibodies [87]. CD40, a cell-surface protein belonging to the tissue necrosis factor (TNF) superfamily and a critical component of both adaptive and innate immune systems, is expressed on a wide variety of immune cells, including macrophages, monocytes, dendritic cells, and B cells. CD40 agonist antibodies exploit the following mechanisms to activate anti-tumor immunity: (1) activation of antigen-presenting cells (APCs) resulting in the generation of tumor-specific cytotoxic T cells that do not require helper CD4 T cells, (2) direct stimulation of macrophages that depletes tumor stroma, converting ‘cold’ tumors into ‘hot’ tumors, and (3) APCs activated by CD40 stimulation produce interleukins (IL) IL-12 and IL-15 facilitating NK cell-mediated tumor cell killing.
The rationale for the clinical development of CD40 agonist antibodies as monotherapy or in combination with ICIs, vaccines, chemotherapy, and radiations has emerged from numerous preclinical studies in murine models [87]. A combination of CD40 agonist and gemcitabine in a small group of treatment-naïve mPDAC patients has reported promising activity- 88% response rate by FDG-PET, partial response (PR) in 4 out of 21 patients, and stable disease (SD) in 11 patients according to the RECIST criteria [88]. A study is currently undergoing to evaluate the safety and efficacy of CDX-1140 (CD40 agonist antibody) alone (Part 1) or in combination with CDX-301, a hematopoietic cytokine (Fms-related tyrosine kinase 3 ligand) (Part 2), pembrolizumab (Part 3), or chemotherapy (Part 4) in patients with a variety of solid tumors (NCT03329950).
The other emerging immunotherapy approach is vaccine therapy. GVAX pancreas, an irradiated allogeneic whole pancreatic tumor cell vaccine programmed to express granulocyte-macrophage colony-stimulating factor (GM-CSF), combined with cyclophosphamide and CRS-207, live-attenuated Listeria monocytogenes-expressing mesothelin, showed a median OS of 9.7 months in a cohort of 90 patients with previously treated mPDAC, a result seemingly better than historical OS achievable with chemotherapy [89]. However, a subsequent larger phase IIb study with this combination did not show improved OS compared to chemotherapy [90]. A randomized phase II trial in previously treated patients also failed to improve OS with the nivolumab and GVAX/CRS 207 combination [91]. A phase II randomized study is currently recruiting to evaluate the efficacy of GVAX/CRS 207 plus dual ICI (ipilimumab and nivolumab) combination (NCT03190265).
Chimeric antigen receptor (CAR)-T cell therapy that employs genetically-engineered T cells directed to specific cancer-associated antigens is also being evaluated in PDAC. Preliminary results of preclinical and phase I studies in PDAC are encouraging [92,93], and several phase I/II trials are currently underway with CAR-T cell therapy in a variety of solid tumors, including PDAC (NCT02744287, NCT03159819).

6. Therapies Targeting Tumor Microenvironment and Metabolomics

Deranged mitochondrial metabolism is a common characteristic in cancer cells [94]. Devimistat (CPI-613), a lipoate analog that can inactivate two crucial enzymes in the Krebs cycle, specifically in tumor cells, was evaluated in the phase 3 AVENGER 500 trial along with modified FOLFIRINOX, compared to FOLFIRINOX alone (NCT03504423). Based on the press release from the company, the mOS was 11.1 months in the devimistat + chemotherapy arm vs. 11.7 months in the chemotherapy arm (HR 0.95, p = 0.66).
Altered amino acid metabolism is another feature of cancer. Tumor cells rely on the external supply of non-essential amino acids such as asparagine to survive [95]. L-asparagine depletes the plasma arginine and leads to cancer cell death [96]. Eryaspase, RBC encapsulated L-asparaginase, was tested in phase 3 Trybeca-1 trial and did not improve OS in patients with mPDAC treated with eryaspase + chemotherapy compared to chemotherapy alone in the second-line setting [97]. ADI-PEG 20, a pegylated arginine deaminase, and GnP led to an ORR of 45% in a small study in treatment-naive PDAC patients [98].

7. Targeting Stroma

The dense desmoplastic stroma in PDAC is associated with decreased drug delivery to the cancer cells [99]. Approaches to deplete the stroma in PDAC have been unsuccessful so far. PEGPH20, a pegylated human hyaluronidase, in combination with FOLFIRINOX compared to FOLFIRINOX alone in a hyaluronic acid (HA)-high unselected population led to inferior outcomes [100]. Furthermore, In the phase 3 trial of HA-high PDAC patients, the addition of PEGPH20 to GnP did not lead to improved OS or PFS [101]. Similarly, pembrolizumab plus PEGPH20 did not improve PFS compared to historical data in the advanced treatment-refractory setting [102].
Current approaches mostly focus on stromal remodeling and normalization (Table 4). Pamrevlumab (FG-3019) is a monoclonal antibody targeting connective tissue growth factors. Preclinical studies showed decreased tumor growth, metastasis, and fibrosis with the use of pamrevlumab [103]. A phase 3 study is evaluating the role of this agent in mPDAC (Table 4). Vitamin D receptor (VDR) is highly expressed in PDAC stroma, including among cancer-associated fibroblasts (CAFs), which are promoters of PDAC growth and aggressiveness [104]. Vitamin D agonists can shift CAFs to a more quiescent and less aggressive form reducing tumor growth and improving chemotherapy delivery [105]. Paricalcitol, a VDR agonist, is being studied in multiple PDAC trials (Table 4). Vitamin A derivative all-trans retinoic acid, with an established role in promyelocytic leukemia management, is undergoing testing for its ability to reprogram the stroma in PDAC [106]. The efficacy of ATRA + GnP is being explored in a phase 2b study (Table 4).

8. Future Perspective and Conclusions

The current treatment of advanced pancreatic cancer is largely empiric. The genomic profiling technology unraveling the signaling pathways that support tumor growth and propagation has created precision treatment opportunities for many cancer types, including PDAC. Although a road to precision oncology has not been clearly mapped out yet, the recent discovery of several treatment-relevant predictive biomarkers has helped the precision therapy paradigm. However, tumor heterogeneity ‘in space and time’ in pancreatic cancer poses formidable challenges in finding predictive biomarkers that can reliably inform clinicians to choose effective treatment for a given patient at a given point in time. A wide variety of research tools have emerged attempting to solve these problems.
Research to personalize chemotherapy in patients with PDAC is rapidly evolving. Multiple approaches to personalize chemotherapy are currently ongoing. Based on tumor transcriptomics, distinct molecular subtypes of PDAC have been identified. Of these subtypes, the classical and basal-like patterns in a modified Moffitt classification have shown prognostic and predictive values [107,108]. The COMPASS trial that performed real-time RNA and whole-genome sequencing (WGS) in patients undergoing first-line chemotherapy identified improved chemotherapy response in the classical subtype, especially with FOLFIRINOX [107]. Furthermore, there was a suggestion of possible improved outcomes in the basal-like tumors treated with GnP [107]. More importantly, this study revealed that GATA6 RNA expression could potentially distinguish the two sub-types [107]. Based on these data, GATA 6 tissue immunohistochemistry (IHC) has been proposed as a simpler surrogate to differentiate between the two sub-types [109]. The PASS-01 trial is prospectively evaluating if the response to GnP and FOLFIRINOX correlates with GATA-6 expression in the first-line setting (NCT04469556). Alternatively, Purity Independent Subtyping of Tumors (purist) was developed as a single sample classifier that could successfully identify tumor subtypes with high performance even utilizing tissues with limited cellularities, such as those obtained by fine-needle aspiration at the time of diagnosis [110]. The basal-like tumor subtype based on purist classification system was associated with resistance to FOLFIRINOX [110]. The ongoing phase 2 PANCREAS trial is evaluating if the purist classification system can guide treatment in the neoadjuvant setting (NCT04683315).
Patient-derived organoids (PDOs) are another tool that can potentially provide a unique opportunity to develop timely models for treatment response prediction before such treatments are administered to the patient [111]. This approach, however, is currently limited by the pace of organoid development and the inability to replicate intratumor heterogeneity and tumor microenvironment. Circulating tumor cells (CTCs) may also be another tool to help in treatment response prediction [112]. The PASS-01 trial, a prospective randomized phase 2 trial evaluating GnP vs. modified FOLFIRINOX in the first-line setting, aims to systematically synthesize data from PDOs, WGS, RNA sequencing, and serial circulating tumor DNA, CTCs and help inform precision therapy choices (NCT04469556).
The universal availability of genomic profiling has provided the opportunity to understand the biology of tumors on a personal level. With the inevitable progress of the multi-omics testing of tumors and the ability for an in-depth evaluation of tumor characteristics through technologies such as single-cell RNA, a multidisciplinary molecular tumor board including clinical/translational/basic investigators, geneticists, bioinformatics, pharmacist, mediation acquisition specialist, and clinical trial navigators needs to be available to determine the choice of initial treatment and subsequent therapies based on a clonal evolution of the tumor. Such an approach has been previously implemented [113]. Ideally, this should be open to all treatment oncologists irrespective of the type of the treating institution (academic vs. community) and locally available resources. A successful example of such an approach is available (https://www.nature.com/natcancer, accessed on 15 April 2022). Utilizing such an approach, testing and optimizing combinatorial targeted therapies in PDAC is reachable. However, it is important to highlight that genomic profiling technology is not ubiquitously available in all parts of the world, which poses a barrier to accessing novel therapies. As technology matures and becomes cheaper, hopefully, it will be within reach of the majority of the world.
The liquid biopsy that encompasses one or more circulating biomarkers, including cell-free DNA (cfDNA), tumor-derived exosomes, and circulating tumor cells, has shown promise as a novel tool to supplement the care continuum for patients with mPDAC, although not yet universally adopted for routine clinical use [114]. The use of cfDNA-based tumor genomic profiling is particularly important in the setting of metastatic disease after progression on standard treatment. Preliminary data suggest that cfDNA has a higher concordance with metastatic lesions than primary tumor tissue [115]. cfDNA analysis may identify the actionable mutations, broadening treatment options in the setting where obtaining an additional tissue biopsy is challenging. However, prospective studies need to be conducted to assess if this theoretical advantage can be translated into a quantifiable clinical benefit.
In conclusion, the modest efficacy of chemotherapy in advanced PDAC has shifted the focus of current research from cytotoxic chemotherapy development to biomarker-driven precision therapy. The remarkable advancements in the understanding of molecular biology of PDAC, especially in the areas of DNA repair mechanisms, tumor microenvironment, and metabolomics, have brought new opportunities for novel therapy development. However, several important barriers have to be overcome before precision therapy can be successfully implemented, including intratumor heterogeneity, multiple signaling pathways supporting cancer growth, the signaling pathway redundancy of RAS, and clonal evolution. Both serial liquid biopsy and the multi-omic evaluation of tumors might serve as a new tool to determine the optimum treatment for each patient.

Author Contributions

Conceptualization, S.C. and A.M.; methodology, all authors; validation, all authors; formal analysis, all authors; investigation, all authors; resources, S.C., M.K. and A.M.; data curation, S.C., M.K. and A.M.; writing—original draft preparation, all authors; writing—review and editing, all authors; visualization, all authors; supervision, A.M.; project administration, all authors; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
  2. Torre, L.A.; Bray, F.; Siegel, R.L.; Ferlay, J.; Lortet-Tieulent, J.; Jemal, A. Global cancer statistics, 2012. CA Cancer J. Clin. 2015, 65, 87–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Ryan, D.P.; Hong, T.S.; Bardeesy, N. Pancreatic Adenocarcinoma. N. Engl. J. Med. 2014, 371, 1039–1049. [Google Scholar] [CrossRef]
  4. Surveillance, E.; End Results (SEER) Program. Cancer Stat Facts: Pancreatic Cancer. National Cancer Institute. Available online: https://seer.cancer.gov/statfacts/html/pancreas.html (accessed on 27 December 2021).
  5. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Yamada, M.; Sugiura, T.; Okamura, Y.; Ito, T.; Yamamoto, Y.; Ashida, R.; Ohgi, K.; Aramaki, T.; Endo, M.; Uesaka, K. Clinical Implication of Node-negative Resectable Pancreatic Cancer. Ann. Surg. Oncol. 2021, 28, 2257–2264. [Google Scholar] [CrossRef] [PubMed]
  7. Conroy, T.; Desseigne, F.; Ychou, M.; Bouché, O.; Guimbaud, R.; Bécouarn, Y.; Adenis, A.; Raoul, J.L.; Gourgou-Bourgade, S.; de la Fouchardière, C.; et al. FOLFIRINOX vs. gemcitabine for metastatic pancreatic cancer. N. Engl. J. Med. 2011, 364, 1817–1825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Von Hoff, D.D.; Ervin, T.; Arena, F.P.; Chiorean, E.G.; Infante, J.; Moore, M.; Seay, T.; Tjulandin, S.A.; Ma, W.W.; Saleh, M.N.; et al. Increased Survival in Pancreatic Cancer with nab-Paclitaxel plus Gemcitabine. N. Engl. J. Med. 2013, 369, 1691–1703. [Google Scholar] [CrossRef] [Green Version]
  9. Berlin, J.D.; Catalano, P.; Thomas, J.P.; Kugler, J.W.; Haller, D.G.; Benson, A.B., 3rd. Phase III study of gemcitabine in combination with fluorouracil vs. gemcitabine alone in patients with advanced pancreatic carcinoma: Eastern Cooperative Oncology Group Trial E2297. J. Clin. Oncol. 2002, 20, 3270–3275. [Google Scholar] [CrossRef]
  10. Burris, H.A., 3rd; Moore, M.J.; Andersen, J.; Green, M.R.; Rothenberg, M.L.; Modiano, M.R.; Cripps, M.C.; Portenoy, R.K.; Storniolo, A.M.; Tarassoff, P.; et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: A randomized trial. J. Clin. Oncol. 1997, 15, 2403–2413. [Google Scholar] [CrossRef] [Green Version]
  11. Cunningham, D.; Chau, I.; Stocken, D.D.; Valle, J.W.; Smith, D.; Steward, W.; Harper, P.G.; Dunn, J.; Tudur-Smith, C.; West, J.; et al. Phase III randomized comparison of gemcitabine vs. gemcitabine plus capecitabine in patients with advanced pancreatic cancer. J. Clin. Oncol. 2009, 27, 5513–5518. [Google Scholar] [CrossRef]
  12. Heinemann, V.; Quietzsch, D.; Gieseler, F.; Gonnermann, M.; Schönekäs, H.; Rost, A.; Neuhaus, H.; Haag, C.; Clemens, M.; Heinrich, B.; et al. Randomized phase III trial of gemcitabine plus cisplatin compared with gemcitabine alone in advanced pancreatic cancer. J. Clin. Oncol. 2006, 24, 3946–3952. [Google Scholar] [CrossRef] [PubMed]
  13. Sultana, A.; Smith, C.T.; Cunningham, D.; Starling, N.; Neoptolemos, J.P.; Ghaneh, P. Meta-analyses of chemotherapy for locally advanced and metastatic pancreatic cancer. J. Clin. Oncol. 2007, 25, 2607–2615. [Google Scholar] [CrossRef] [PubMed]
  14. Yip, D.; Karapetis, C.; Strickland, A.; Steer, C.B.; Goldstein, D. Chemotherapy and radiotherapy for inoperable advanced pancreatic cancer. Cochrane Database Syst. Rev. 2006, CD002093. [Google Scholar] [CrossRef]
  15. Moore, M.J.; Goldstein, D.; Hamm, J.; Figer, A.; Hecht, J.R.; Gallinger, S.; Au, H.J.; Murawa, P.; Walde, D.; Wolff, R.A.; et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: A phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J. Clin. Oncol. 2007, 25, 1960–1966. [Google Scholar] [CrossRef] [PubMed]
  16. Jameson, G.S.; Borazanci, E.; Babiker, H.M.; Poplin, E.; Niewiarowska, A.A.; Gordon, M.S.; Barrett, M.T.; Rosenthal, A.; Stoll-D’Astice, A.; Crowley, J.; 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. 2020, 6, 125–132. [Google Scholar] [CrossRef] [PubMed]
  17. O’Reilly, E.M.; Lee, J.W.; Zalupski, M.; Capanu, M.; Park, J.; Golan, T.; Tahover, E.; Lowery, M.A.; Chou, J.F.; Sahai, V.; 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. [Google Scholar] [CrossRef]
  18. Wainberg, Z.A.; Bekaii-Saab, T.; Boland, P.M.; Dayyani, F.; Macarulla, T.; Mody, K.; Belanger, B.; Maxwell, F.; Moore, Y.; Thiagalingam, A.; et al. First-line liposomal irinotecan with oxaliplatin, 5-fluorouracil and leucovorin (NALIRIFOX) in pancreatic ductal adenocarcinoma: A phase I/II study. Eur. J. Cancer 2021, 151, 14–24. [Google Scholar] [CrossRef]
  19. Stein, S.M.; James, E.S.; Deng, Y.; Cong, X.; Kortmansky, J.S.; Li, J.; Staugaard, C.; Indukala, D.; Boustani, A.M.; Patel, V.; et al. Final analysis of a phase II study of modified FOLFIRINOX in locally advanced and metastatic pancreatic cancer. Br. J. Cancer 2016, 114, 737–743. [Google Scholar] [CrossRef] [Green Version]
  20. Mahaseth, H.; Brutcher, E.; Kauh, J.; Hawk, N.; Kim, S.; Chen, Z.; Kooby, D.A.; Maithel, S.K.; Landry, J.; El-Rayes, B.F. Modified FOLFIRINOX regimen with improved safety and maintained efficacy in pancreatic adenocarcinoma. Pancreas 2013, 42, 1311–1315. [Google Scholar] [CrossRef]
  21. Usón Junior, P.L.S.; Rother, E.T.; Maluf, F.C.; Bugano, D.D.G. Meta-analysis of Modified FOLFIRINOX Regimens for Patients with Metastatic Pancreatic Cancer. Clin. Colorectal Cancer 2018, 17, 187–197. [Google Scholar] [CrossRef]
  22. Kang, J.; Hwang, I.; Yoo, C.; Kim, K.P.; Jeong, J.H.; Chang, H.M.; Lee, S.S.; Park, D.H.; Song, T.J.; Seo, D.W.; et al. Nab-paclitaxel plus gemcitabine vs. FOLFIRINOX as the first-line chemotherapy for patients with metastatic pancreatic cancer: Retrospective analysis. Investig. New Drugs 2018, 36, 732–741. [Google Scholar] [CrossRef] [PubMed]
  23. Kim, S.; Signorovitch, J.E.; Yang, H.; Patterson-Lomba, O.; Xiang, C.Q.; Ung, B.; Parisi, M.; Marshall, J.L. Comparative Effectiveness of nab-Paclitaxel Plus Gemcitabine vs. FOLFIRINOX in Metastatic Pancreatic Cancer: A Retrospective Nationwide Chart Review in the United States. Adv. Ther. 2018, 35, 1564–1577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Pusceddu, S.; Ghidini, M.; Torchio, M.; Corti, F.; Tomasello, G.; Niger, M.; Prinzi, N.; Nichetti, F.; Coinu, A.; Di Bartolomeo, 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 2019, 11, 484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Friedenson, B. BRCA1 and BRCA2 pathways and the risk of cancers other than breast or ovarian. Medscape Gen. Med. 2005, 7, 60. [Google Scholar]
  26. Farmer, H.; McCabe, N.; Lord, C.J.; Tutt, A.N.; Johnson, D.A.; Richardson, T.B.; Santarosa, M.; Dillon, K.J.; Hickson, I.; Knights, C.; et al. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005, 434, 917–921. [Google Scholar] [CrossRef]
  27. Lowery, M.A.; Kelsen, D.P.; Capanu, M.; Smith, S.C.; Lee, J.W.; Stadler, Z.K.; Moore, M.J.; Kindler, H.L.; Golan, T.; Segal, A.; et al. Phase II trial of veliparib in patients with previously treated BRCA-mutated pancreas ductal adenocarcinoma. Eur. J. Cancer 2018, 89, 19–26. [Google Scholar] [CrossRef]
  28. O’Reilly, E.M.; Lee, J.W.; Lowery, M.A.; Capanu, M.; Stadler, Z.K.; Moore, M.J.; Dhani, N.; Kindler, H.L.; Estrella, H.; Maynard, H.; et al. Phase 1 trial evaluating cisplatin, gemcitabine, and veliparib in 2 patient cohorts: Germline BRCA mutation carriers and wild-type BRCA pancreatic ductal adenocarcinoma. Cancer 2018, 124, 1374–1382. [Google Scholar] [CrossRef] [Green Version]
  29. Assenat, E.; de la Fouchardière, C.; Portales, F.; Ychou, M.; Debourdeau, A.; Desseigne, F.; Iltache, S.; Fiess, C.; Mollevi, C.; Mazard, T. Sequential first-line treatment with nab-paclitaxel/gemcitabine and FOLFIRINOX in metastatic pancreatic adenocarcinoma: GABRINOX phase Ib-II controlled clinical trial. ESMO Open 2021, 6, 100318. [Google Scholar] [CrossRef]
  30. Golan, T.; Hammel, P.; Reni, M.; Van Cutsem, E.; Macarulla, T.; Hall, M.J.; Park, J.-O.; Hochhauser, D.; Arnold, D.; Oh, D.-Y.; et al. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. N. Engl. J. Med. 2019, 381, 317–327. [Google Scholar] [CrossRef]
  31. Golan, T.; Hammel, P.; Reni, M.; Cutsem, E.V.; Macarulla, T.; Hall, M.J.; Park, J.O.; Hochhauser, D.; Arnold, D.; Oh, D.-Y.; et al. Overall survival from the phase 3 POLO trial: Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. J. Clin. Oncol. 2021, 39, 378. [Google Scholar] [CrossRef]
  32. Reiss, K.A.; Mick, R.; O’Hara, M.H.; Teitelbaum, U.; Karasic, T.B.; Schneider, C.; Cowden, S.; Southwell, T.; Romeo, J.; Izgur, N.; et al. Phase II Study of Maintenance Rucaparib in Patients with Platinum-Sensitive Advanced Pancreatic Cancer and a Pathogenic Germline or Somatic Variant in BRCA1, BRCA2, or PALB2. J. Clin. Oncol. 2021, 39, 2497–2505. [Google Scholar] [CrossRef] [PubMed]
  33. Chevalier, H.; Vienot, A.; Lièvre, A.; Edeline, J.; El Hajbi, F.; Peugniez, C.; Vernerey, D.; Meurisse, A.; Hammel, P.; Neuzillet, C.; et al. FOLFIRINOX De-Escalation in Advanced Pancreatic Cancer: A Multicenter Real-Life Study. Oncologist 2020, 25, e1701–e1710. [Google Scholar] [CrossRef] [PubMed]
  34. Dahan, L.; Williet, N.; Malicot, K.L.; Phelip, J.-M.; Desrame, J.; Bouché, O.; Petorin, C.; Malka, D.; Rebischung, C.; Aparicio, T.; et al. Randomized Phase II Trial Evaluating Two Sequential Treatments in First Line of Metastatic Pancreatic Cancer: Results of the PANOPTIMOX-PRODIGE 35 Trial. J. Clin. Oncol. 2021, 39, 3242–3250. [Google Scholar] [CrossRef]
  35. Petrioli, R.; Torre, P.; Pesola, G.; Paganini, G.; Paolelli, L.; Miano, S.T.; Martellucci, I.; Francini, G.; Francini, E. Gemcitabine plus nab-paclitaxel followed by maintenance treatment with gemcitabine alone as first-line treatment for older adults with locally advanced or metastatic pancreatic cancer. J. Geriatr. Oncol. 2020, 11, 647–651. [Google Scholar] [CrossRef] [PubMed]
  36. Reni, M.; Cereda, S.; Milella, M.; Novarino, A.; Passardi, A.; Mambrini, A.; Di Lucca, G.; Aprile, G.; Belli, C.; Danova, M.; et al. Maintenance sunitinib or observation in metastatic pancreatic adenocarcinoma: A phase II randomised trial. Eur. J. Cancer 2013, 49, 3609–3615. [Google Scholar] [CrossRef] [PubMed]
  37. Gränsmark, E.; Bylin, N.B.; Blomstrand, H.; Fredrikson, M.; Åvall-Lundqvist, E.; Elander, N.O. Real World Evidence on Second-Line Palliative Chemotherapy in Advanced Pancreatic Cancer. Front. Oncol. 2020, 10, 1176. [Google Scholar] [CrossRef] [PubMed]
  38. Mita, N.; Iwashita, T.; Uemura, S.; Yoshida, K.; Iwasa, Y.; Ando, N.; Iwata, K.; Okuno, M.; Mukai, T.; Shimizu, M. Second-Line Gemcitabine Plus Nab-Paclitaxel for Patients with Unresectable Advanced Pancreatic Cancer after First-Line FOLFIRINOX Failure. J. Clin. Med. 2019, 8, 761. [Google Scholar] [CrossRef] [Green Version]
  39. Portal, A.; Pernot, S.; Tougeron, D.; Arbaud, C.; Bidault, A.T.; de la Fouchardière, C.; Hammel, P.; Lecomte, T.; Dréanic, J.; Coriat, R.; 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. [Google Scholar] [CrossRef]
  40. Viaud, J.; Brac, C.; Artru, P.; Le Pabic, E.; Leconte, B.; Bodère, A.; Pracht, M.; Le Sourd, S.; Edeline, J.; Lièvre, A. Gemcitabine as second-line chemotherapy after Folfirinox failure in advanced pancreatic adenocarcinoma: A retrospective study. Dig. Liver Dis. 2017, 49, 692–696. [Google Scholar] [CrossRef]
  41. Wang-Gillam, A.; Li, C.P.; Bodoky, G.; Dean, A.; Shan, Y.S.; Jameson, G.; Macarulla, T.; Lee, K.H.; Cunningham, D.; Blanc, J.F.; 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. [Google Scholar] [CrossRef]
  42. Oettle, H.; Riess, H.; Stieler, J.M.; Heil, G.; Schwaner, I.; Seraphin, J.; Görner, M.; Mölle, M.; Greten, T.F.; Lakner, V.; et al. Second-line oxaliplatin, folinic acid, and fluorouracil vs. folinic acid and fluorouracil alone for gemcitabine-refractory pancreatic cancer: Outcomes from the CONKO-003 trial. J. Clin. Oncol. 2014, 32, 2423–2429. [Google Scholar] [CrossRef] [PubMed]
  43. Gill, S.; Ko, Y.J.; Cripps, C.; Beaudoin, A.; Dhesy-Thind, S.; Zulfiqar, M.; Zalewski, P.; Do, T.; Cano, P.; Lam, W.Y.H.; et al. PANCREOX: A Randomized Phase III Study of Fluorouracil/Leucovorin with or without Oxaliplatin for Second-Line Advanced Pancreatic Cancer in Patients Who Have Received Gemcitabine-Based Chemotherapy. J. Clin. Oncol. 2016, 34, 3914–3920. [Google Scholar] [CrossRef] [PubMed]
  44. Kim, J.H.; Lee, S.-C.; Oh, S.Y.; Song, S.-Y.; Lee, N.; Nam, E.M.; Lee, S.; Hwang, I.G.; Lee, H.R.; Lee, K.T.; et al. Attenuated FOLFIRINOX in the salvage treatment of gemcitabine-refractory advanced pancreatic cancer: A phase II study. Cancer Commun. 2018, 38, 32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Sawada, M.; Kasuga, A.; Mie, T.; Furukawa, T.; Taniguchi, T.; Fukuda, K.; Yamada, Y.; Takeda, T.; Kanata, R.; Matsuyama, M.; et al. Modified FOLFIRINOX as a second-line therapy following gemcitabine plus nab-paclitaxel therapy in metastatic pancreatic cancer. BMC Cancer 2020, 20, 449. [Google Scholar] [CrossRef]
  46. Zaniboni, A.; Aitini, E.; Barni, S.; Ferrari, D.; Cascinu, S.; Catalano, V.; Valmadre, G.; Ferrara, D.; Veltri, E.; Codignola, C.; et al. FOLFIRI as second-line chemotherapy for advanced pancreatic cancer: A GISCAD multicenter phase II study. Cancer Chemother. Pharmacol. 2012, 69, 1641–1645. [Google Scholar] [CrossRef]
  47. Xiong, H.Q.; Varadhachary, G.R.; Blais, J.C.; Hess, K.R.; Abbruzzese, J.L.; Wolff, R.A. Phase 2 trial of oxaliplatin plus capecitabine (XELOX) as second-line therapy for patients with advanced pancreatic cancer. Cancer 2008, 113, 2046–2052. [Google Scholar] [CrossRef]
  48. Ettrich, T.J.; Perkhofer, L.; von Wichert, G.; Gress, T.M.; Michl, P.; Hebart, H.F.; Büchner-Steudel, P.; Geissler, M.; Muche, R.; Danner, B.; et al. DocOx (AIO-PK0106): A phase II trial of docetaxel and oxaliplatin as a second line systemic therapy in patients with advanced pancreatic ductal adenocarcinoma. BMC Cancer 2016, 16, 21. [Google Scholar] [CrossRef] [Green Version]
  49. Sonbol, M.B.; Firwana, B.; Wang, Z.; Almader-Douglas, D.; Borad, M.J.; Makhoul, I.; Ramanathan, R.K.; Ahn, D.H.; Bekaii-Saab, T. Second-line treatment in patients with pancreatic ductal adenocarcinoma: A meta-analysis. Cancer 2017, 123, 4680–4686. [Google Scholar] [CrossRef]
  50. Damanakis, A.I.; Ostertag, L.; Waldschmidt, D.; Kütting, F.; Quaas, A.; Plum, P.; Bruns, C.J.; Gebauer, F.; Popp, F. Proposal for a definition of “Oligometastatic disease in pancreatic cancer”. BMC Cancer 2019, 19, 1261. [Google Scholar] [CrossRef] [Green Version]
  51. Puckett, Y.; Eskander, M.F.; Aquina, C.T.; Pawlik, T.M. Is there a role for resection of oligometastatic disease in pancreatic ductal adenocarcinoma? J. Cancer Metastasis Treat. 2020, 6, 44. [Google Scholar] [CrossRef]
  52. De Simoni, O.; Scarpa, M.; Tonello, M.; Pilati, P.; Tolin, F.; Spolverato, Y.; Gruppo, M. Oligometastatic Pancreatic Cancer to the Liver in the Era of Neoadjuvant Chemotherapy: Which Role for Conversion Surgery? A Systematic Review and Meta-Analysis. Cancers 2020, 12, 3402. [Google Scholar] [CrossRef] [PubMed]
  53. Hackert, T.; Niesen, W.; Hinz, U.; Tjaden, C.; Strobel, O.; Ulrich, A.; Michalski, C.W.; Büchler, M.W. Radical surgery of oligometastatic pancreatic cancer. Eur. J. Surg. Oncol. 2017, 43, 358–363. [Google Scholar] [CrossRef] [PubMed]
  54. Downs-Canner, S.; Zenati, M.; Boone, B.A.; Varley, P.R.; Steve, J.; Hogg, M.E.; Zureikat, A.; Zeh, H.J.; Lee, K.K. The indolent nature of pulmonary metastases from ductal adenocarcinoma of the pancreas. J. Surg. Oncol. 2015, 112, 80–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Bryant, K.L.; Mancias, J.D.; Kimmelman, A.C.; Der, C.J. KRAS: Feeding pancreatic cancer proliferation. Trends Biochem. Sci. 2014, 39, 91–100. [Google Scholar] [CrossRef] [Green Version]
  56. Luo, J. KRAS mutation in pancreatic cancer. Semin. Oncol. 2021, 48, 10–18. [Google Scholar] [CrossRef]
  57. Bekaii-Saab, T.S.; Spira, A.I.; Yaeger, R.; Buchschacher, G.L.; McRee, A.J.; Sabari, J.K.; Johnson, M.L.; Barve, M.A.; Hafez, N.; Velastegui, K.; et al. KRYSTAL-1: Updated activity and safety of adagrasib (MRTX849) in patients (Pts) with unresectable or metastatic pancreatic cancer (PDAC) and other gastrointestinal (GI) tumors harboring a KRASG12C mutation. J. Clin. Oncol. 2022, 40, 519. [Google Scholar] [CrossRef]
  58. Strickler, J.H.; Satake, H.; Hollebecque, A.; Sunakawa, Y.; Tomasini, P.; Bajor, D.L.; Schuler, M.H.; Yaeger, R.; George, T.J.; Garrido-Laguna, I.; et al. First data for sotorasib in patients with pancreatic cancer with KRAS p.G12C mutation: A phase I/II study evaluating efficacy and safety. J. Clin. Oncol. 2022, 40, 360490. [Google Scholar] [CrossRef]
  59. Ning, W.; Yang, Z.; Kocher, G.J.; Dorn, P.; Peng, R.W. A Breakthrough Brought about by Targeting KRAS(G12C): Nonconformity is Punished. Cancers 2022, 14, 390. [Google Scholar] [CrossRef]
  60. Bournet, B.; Muscari, F.; Buscail, C.; Assenat, E.; Barthet, M.; Hammel, P.; Selves, J.; Guimbaud, R.; Cordelier, P.; Buscail, L. KRAS G12D Mutation Subtype Is A Prognostic Factor for Advanced Pancreatic Adenocarcinoma. Clin. Transl. Gastroenterol. 2016, 7, e157. [Google Scholar] [CrossRef]
  61. Vatansever, S.; Erman, B.; Gümüş, Z.H. Comparative effects of oncogenic mutations G12C, G12V, G13D, and Q61H on local conformations and dynamics of K-Ras. Comput. Struct. Biotechnol. J. 2020, 18, 1000–1011. [Google Scholar] [CrossRef]
  62. Infante, J.R.; Somer, B.G.; Park, J.O.; Li, C.P.; Scheulen, M.E.; Kasubhai, S.M.; Oh, D.Y.; Liu, Y.; Redhu, S.; Steplewski, K.; et al. A randomised, double-blind, placebo-controlled trial of trametinib, an oral MEK inhibitor, in combination with gemcitabine for patients with untreated metastatic adenocarcinoma of the pancreas. Eur. J. Cancer 2014, 50, 2072–2081. [Google Scholar] [CrossRef] [PubMed]
  63. Bryant, K.L.; Stalnecker, C.A.; Zeitouni, D.; Klomp, J.E.; Peng, S.; Tikunov, A.P.; Gunda, V.; Pierobon, M.; Waters, A.M.; George, S.D.; et al. Combination of ERK and autophagy inhibition as a treatment approach for pancreatic cancer. Nat. Med. 2019, 25, 628–640. [Google Scholar] [CrossRef] [PubMed]
  64. Kinsey, C.G.; Camolotto, S.A.; Boespflug, A.M.; Guillen, K.P.; Foth, M.; Truong, A.; Schuman, S.S.; Shea, J.E.; Seipp, M.T.; Yap, J.T.; et al. Protective autophagy elicited by RAF→MEK→ERK inhibition suggests a treatment strategy for RAS-driven cancers. Nat. Med. 2019, 25, 620–627. [Google Scholar] [CrossRef] [PubMed]
  65. Henry, K.E.; Mack, K.N.; Nagle, V.L.; Cornejo, M.; Michel, A.O.; Fox, I.L.; Davydova, M.; Dilling, T.R.; Pillarsetty, N.; Lewis, J.S. ERK Inhibition Improves Anti-PD-L1 Immune Checkpoint Blockade in Preclinical Pancreatic Ductal Adenocarcinoma. Mol. Cancer Ther. 2021, 20, 2026–2034. [Google Scholar] [CrossRef]
  66. Loi, S.; Dushyanthen, S.; Beavis, P.A.; Salgado, R.; Denkert, C.; Savas, P.; Combs, S.; Rimm, D.L.; Giltnane, J.M.; Estrada, M.V.; et al. RAS/MAPK Activation is Associated with Reduced Tumor-Infiltrating Lymphocytes in Triple-Negative Breast Cancer: Therapeutic Cooperation between MEK and PD-1/PD-L1 Immune Checkpoint Inhibitors. Clin. Cancer Res. 2016, 22, 1499–1509. [Google Scholar] [CrossRef] [Green Version]
  67. Singhi, A.D.; George, B.; Greenbowe, J.R.; Chung, J.; Suh, J.; Maitra, A.; Klempner, S.J.; Hendifar, A.; Milind, J.M.; Golan, T.; et al. Real-Time Targeted Genome Profile Analysis of Pancreatic Ductal Adenocarcinomas Identifies Genetic Alterations That Might Be Targeted with Existing Drugs or Used as Biomarkers. Gastroenterology 2019, 156, 2242–2253.e2244. [Google Scholar] [CrossRef] [Green Version]
  68. Hyman, D.M.; Puzanov, I.; Subbiah, V.; Faris, J.E.; Chau, I.; Blay, J.Y.; Wolf, J.; Raje, N.S.; Diamond, E.L.; Hollebecque, A.; et al. Vemurafenib in Multiple Nonmelanoma Cancers with BRAF V600 Mutations. N. Engl. J. Med. 2015, 373, 726–736. [Google Scholar] [CrossRef]
  69. Heining, C.; Horak, P.; Uhrig, S.; Codo, P.L.; Klink, B.; Hutter, B.; Fröhlich, M.; Bonekamp, D.; Richter, D.; Steiger, K.; et al. NRG1 Fusions in KRAS Wild-Type Pancreatic Cancer. Cancer Discov. 2018, 8, 1087–1095. [Google Scholar] [CrossRef] [Green Version]
  70. Park, W.; Chen, J.; Chou, J.F.; Varghese, A.M.; Yu, K.H.; Wong, W.; Capanu, M.; Balachandran, V.; McIntyre, C.A.; El Dika, I.; et al. Genomic Methods Identify Homologous Recombination Deficiency in Pancreas Adenocarcinoma and Optimize Treatment Selection. Clin. Cancer Res. 2020, 26, 3239–3247. [Google Scholar] [CrossRef]
  71. Casolino, R.; Paiella, S.; Azzolina, D.; Beer, P.A.; Corbo, V.; Lorenzoni, G.; Gregori, D.; Golan, T.; Braconi, C.; Froeling, F.E.M.; et al. Homologous Recombination Deficiency in Pancreatic Cancer: A Systematic Review and Prevalence Meta-Analysis. J. Clin. Oncol. 2021, 39, 2617–2631. [Google Scholar] [CrossRef]
  72. Lin, J.C.; Liu, T.P.; Yang, P.M. CDKN2A-Inactivated Pancreatic Ductal Adenocarcinoma Exhibits Therapeutic Sensitivity to Paclitaxel: A Bioinformatics Study. J. Clin. Med. 2020, 9, 4019. [Google Scholar] [CrossRef] [PubMed]
  73. Al Baghdadi, T.; Halabi, S.; Garrett-Mayer, E.; Mangat, P.K.; Ahn, E.R.; Sahai, V.; Alvarez, R.H.; Kim, E.S.; Yost, K.J.; Rygiel, A.L.; et al. Palbociclib in Patients with Pancreatic and Biliary Cancer with CDKN2A Alterations: Results from the Targeted Agent and Profiling Utilization Registry Study. JCO Precis. Oncol. 2019, 3, 1–8. [Google Scholar] [CrossRef] [PubMed]
  74. Kato, S.; Adashek, J.J.; Shaya, J.; Okamura, R.; Jimenez, R.E.; Lee, S.; Sicklick, J.K.; Kurzrock, R. Concomitant MEK and Cyclin Gene Alterations: Implications for Response to Targeted Therapeutics. Clin. Cancer Res. 2021, 27, 2792–2797. [Google Scholar] [CrossRef] [PubMed]
  75. Mavrakis, K.J.; McDonald, E.R., 3rd; Schlabach, M.R.; Billy, E.; Hoffman, G.R.; deWeck, A.; Ruddy, D.A.; Venkatesan, K.; Yu, J.; McAllister, G.; et al. Disordered methionine metabolism in MTAP/CDKN2A-deleted cancers leads to dependence on PRMT5. Science 2016, 351, 1208–1213. [Google Scholar] [CrossRef]
  76. Marjon, K.; Cameron, M.J.; Quang, P.; Clasquin, M.F.; Mandley, E.; Kunii, K.; McVay, M.; Choe, S.; Kernytsky, A.; Gross, S.; et al. MTAP Deletions in Cancer Create Vulnerability to Targeting of the MAT2A/PRMT5/RIOK1 Axis. Cell Rep. 2016, 15, 574–587. [Google Scholar] [CrossRef] [Green Version]
  77. Hu, Z.I.; Shia, J.; Stadler, Z.K.; Varghese, A.M.; Capanu, M.; Salo-Mullen, E.; Lowery, M.A.; Diaz, L.A., Jr.; Mandelker, D.; Yu, K.H.; et al. Evaluating Mismatch Repair Deficiency in Pancreatic Adenocarcinoma: Challenges and Recommendations. Clin. Cancer Res. 2018, 24, 1326–1336. [Google Scholar] [CrossRef] [Green Version]
  78. Luchini, C.; Brosens, L.A.A.; Wood, L.D.; Chatterjee, D.; Shin, J.I.; Sciammarella, C.; Fiadone, G.; Malleo, G.; Salvia, R.; Kryklyva, V.; et al. Comprehensive characterisation of pancreatic ductal adenocarcinoma with microsatellite instability: Histology, molecular pathology and clinical implications. Gut 2021, 70, 148–156. [Google Scholar] [CrossRef]
  79. Chakrabarti, S.; Bucheit, L.A.; Starr, J.S.; Innis-Shelton, R.; Shergill, A.; Resta, R.; Wagner, S.A.; Kasi, P.M. Does detection of microsatellite instability-high (MSI-H) by plasma-based testing predict tumor response to immunotherapy (IO) in patients with pancreatic cancer (PC)? J. Clin. Oncol. 2022, 40, 607. [Google Scholar] [CrossRef]
  80. Cox, R.E., Jr.; Mahipal, A.; Chakrabarti, S. A Patient with Locally Advanced Mismatch-Repair-Deficient Pancreatic Ductal Adenocarcinoma Successfully Treated with Neoadjuvant Immunotherapy. Cureus 2021, 13, e14640. [Google Scholar] [CrossRef]
  81. Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [Green Version]
  82. Marabelle, A.; Le, D.T.; Ascierto, P.A.; Di Giacomo, A.M.; De Jesus-Acosta, A.; Delord, J.P.; Geva, R.; Gottfried, M.; Penel, N.; Hansen, A.R.; 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. [Google Scholar] [CrossRef]
  83. Huber, M.; Brehm, C.U.; Gress, T.M.; Buchholz, M.; Alhamwe, B.A.; von Strandmann, E.P.; Slater, E.P.; Bartsch, J.W.; Bauer, C.; Lauth, M. The Immune Microenvironment in Pancreatic Cancer. Int. J. Mol. Sci. 2020, 21, 7307. [Google Scholar] [CrossRef] [PubMed]
  84. Royal, R.E.; Levy, C.; Turner, K.; Mathur, A.; Hughes, M.; Kammula, U.S.; Sherry, R.M.; Topalian, S.L.; Yang, J.C.; Lowy, I.; 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. [Google Scholar] [CrossRef] [PubMed]
  85. O’Reilly, E.M.; Oh, D.Y.; Dhani, N.; Renouf, D.J.; Lee, M.A.; Sun, W.; Fisher, G.; Hezel, A.; Chang, S.C.; Vlahovic, G.; et al. Durvalumab with or without Tremelimumab for Patients with Metastatic Pancreatic Ductal Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2019, 5, 1431–1438. [Google Scholar] [CrossRef] [PubMed]
  86. Terrero, G.; Datta, J.; Dennison, J.; Sussman, D.A.; Lohse, I.; Merchant, N.B.; Hosein, P.J. Ipilimumab/Nivolumab Therapy in Patients with Metastatic Pancreatic or Biliary Cancer with Homologous Recombination Deficiency Pathogenic Germline Variants. JAMA Oncol. 2022, e220611. [Google Scholar] [CrossRef]
  87. Vonderheide, R.H. CD40 Agonist Antibodies in Cancer Immunotherapy. Annu. Rev. Med. 2020, 71, 47–58. [Google Scholar] [CrossRef] [Green Version]
  88. Beatty, G.L.; Chiorean, E.G.; Fishman, M.P.; Saboury, B.; Teitelbaum, U.R.; Sun, W.; Huhn, R.D.; Song, W.; Li, D.; Sharp, L.L.; et al. CD40 agonists alter tumor stroma and show efficacy against pancreatic carcinoma in mice and humans. Science 2011, 331, 1612–1616. [Google Scholar] [CrossRef] [Green Version]
  89. Le, D.T.; Wang-Gillam, A.; Picozzi, V.; Greten, T.F.; Crocenzi, T.; Springett, G.; Morse, M.; Zeh, H.; Cohen, D.; Fine, R.L.; et al. Safety and survival with GVAX pancreas prime and Listeria Monocytogenes-expressing mesothelin (CRS-207) boost vaccines for metastatic pancreatic cancer. J. Clin. Oncol. 2015, 33, 1325–1333. [Google Scholar] [CrossRef] [Green Version]
  90. Le, D.T.; Picozzi, V.J.; Ko, A.H.; Wainberg, Z.A.; Kindler, H.; Wang-Gillam, A.; Oberstein, P.; Morse, M.A.; Zeh, H.J., 3rd; Weekes, C.; et al. Results from a Phase IIb, Randomized, Multicenter Study of GVAX Pancreas and CRS-207 Compared with Chemotherapy in Adults with Previously Treated Metastatic Pancreatic Adenocarcinoma (ECLIPSE Study). Clin. Cancer Res. 2019, 25, 5493–5502. [Google Scholar] [CrossRef]
  91. Tsujikawa, T.; Crocenzi, T.; Durham, J.N.; Sugar, E.A.; Wu, A.A.; Onners, B.; Nauroth, J.M.; Anders, R.A.; Fertig, E.J.; Laheru, D.A.; et al. Evaluation of Cyclophosphamide/GVAX Pancreas Followed by Listeria-Mesothelin (CRS-207) with or without Nivolumab in Patients with Pancreatic Cancer. Clin. Cancer Res. 2020, 26, 3578–3588. [Google Scholar] [CrossRef] [Green Version]
  92. Beatty, G.L.; O’Hara, M.H.; Lacey, S.F.; Torigian, D.A.; Nazimuddin, F.; Chen, F.; Kulikovskaya, I.M.; Soulen, M.C.; McGarvey, M.; Nelson, A.M.; et al. Activity of Mesothelin-Specific Chimeric Antigen Receptor T Cells against Pancreatic Carcinoma Metastases in a Phase 1 Trial. Gastroenterology 2018, 155, 29–32. [Google Scholar] [CrossRef] [PubMed]
  93. Dickson, I. Improved CAR T therapy for PDAC. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 456. [Google Scholar] [CrossRef] [PubMed]
  94. Vander Heiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg effect: The metabolic requirements of cell proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Bachet, J.-B.; Gay, F.; Maréchal, R.; Galais, M.-P.; Adenis, A.; MsC, D.S.; Cros, J.; Demetter, P.; Svrcek, M.; Bardier-Dupas, A.; et al. Asparagine Synthetase Expression and Phase I Study with L-Asparaginase Encapsulated in Red Blood Cells in Patients with Pancreatic Adenocarcinoma. Pancreas 2015, 44, 1141–1147. [Google Scholar] [CrossRef]
  96. Schrek, R.; Dolowy, W.C.; Ammeraal, R.N. L-asparaginase: Toxicity to normal and leukemic human lymphocytes. Science 1967, 155, 329–330. [Google Scholar] [CrossRef]
  97. Hammel, P.; El-Hariry, I.; Macarulla, T.; Garcia-Carbonero, R.; Metges, J.-P.; Bouché, O.; Portales, F.; Cid, R.A.P.; Mineur, L.; Gracian, A.M.C.; et al. Trybeca-1: A randomized, phase 3 study of eryaspase in combination with chemotherapy vs. chemotherapy alone as second-line treatment in patients with advanced pancreatic adenocarcinoma (NCT03665441). J. Clin. Oncol. 2022, 40, 518. [Google Scholar] [CrossRef]
  98. Lowery, M.A.; Yu, K.H.; Kelsen, D.P.; Harding, J.J.; Bomalaski, J.S.; Glassman, D.C.; Covington, C.M.; Brenner, R.; Hollywood, E.; Barba, A.; et al. A phase 1/1B trial of ADI-PEG 20 plus nab-paclitaxel and gemcitabine in patients with advanced pancreatic adenocarcinoma. Cancer 2017, 123, 4556–4565. [Google Scholar] [CrossRef]
  99. Netti, P.A.; Berk, D.A.; Swartz, M.A.; Grodzinsky, A.J.; Jain, R.K. Role of extracellular matrix assembly in interstitial transport in solid tumors. Cancer Res. 2000, 60, 2497–2503. [Google Scholar]
  100. Ramanathan, R.K.; McDonough, S.L.; Philip, P.A.; Hingorani, S.R.; Lacy, J.; Kortmansky, J.S.; Thumar, J.; Chiorean, E.G.; Shields, A.F.; Behl, D.; et al. Phase IB/II Randomized Study of FOLFIRINOX Plus Pegylated Recombinant Human Hyaluronidase vs. FOLFIRINOX Alone in Patients with Metastatic Pancreatic Adenocarcinoma: SWOG S1313. J. Clin. Oncol. 2019, 37, 1062–1069. [Google Scholar] [CrossRef]
  101. Van Cutsem, E.; Tempero, M.A.; Sigal, D.; Oh, D.Y.; Fazio, N.; Macarulla, T.; Hitre, E.; Hammel, P.; Hendifar, A.E.; Bates, S.E.; 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. [Google Scholar] [CrossRef]
  102. Zhen, D.B.; Whittle, M.; Ritch, P.S.; Hochster, H.S.; Coveler, A.L.; George, B.; Hendifar, A.E.; Dragovich, T.; Green, S.; Dion, B.; et al. Phase II study of PEGPH20 plus pembrolizumab for patients (pts) with hyaluronan (HA)-high refractory metastatic pancreatic adenocarcinoma (mPC): PCRT16-001. J. Clin. Oncol. 2022, 40, 576. [Google Scholar] [CrossRef]
  103. Aikawa, T.; Gunn, J.; Spong, S.M.; Klaus, S.J.; Korc, M. Connective tissue growth factor-specific antibody attenuates tumor growth, metastasis, and angiogenesis in an orthotopic mouse model of pancreatic cancer. Mol. Cancer Ther. 2006, 5, 1108–1116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Norton, J.; Foster, D.; Chinta, M.; Titan, A.; Longaker, M. Pancreatic Cancer Associated Fibroblasts (CAF): Under-Explored Target for Pancreatic Cancer Treatment. Cancers 2020, 12, 1347. [Google Scholar] [CrossRef] [PubMed]
  105. Sherman, M.H.; Yu, R.T.; Engle, D.D.; Ding, N.; Atkins, A.R.; Tiriac, H.; Collisson, E.A.; Connor, F.; Van Dyke, T.; Kozlov, S.; et al. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis and enhances pancreatic cancer therapy. Cell 2014, 159, 80–93. [Google Scholar] [CrossRef] [Green Version]
  106. Kocher, H.M.; Basu, B.; Froeling, F.E.M.; Sarker, D.; Slater, S.; Carlin, D.; de Souza, N.M.; De Paepe, K.N.; Goulart, M.R.; Hughes, C.; et al. Phase I clinical trial repurposing all-trans retinoic acid as a stromal targeting agent for pancreatic cancer. Nat. Commun. 2020, 11, 4841. [Google Scholar] [CrossRef]
  107. Aung, K.L.; Fischer, S.E.; Denroche, R.E.; Jang, G.H.; Dodd, A.; Creighton, S.; Southwood, B.; Liang, S.B.; Chadwick, D.; Zhang, A.; et al. Genomics-Driven Precision Medicine for Advanced Pancreatic Cancer: Early Results from the COMPASS Trial. Clin. Cancer Res. 2018, 24, 1344–1354. [Google Scholar] [CrossRef] [Green Version]
  108. Moffitt, R.A.; Marayati, R.; Flate, E.L.; Volmar, K.E.; Loeza, S.G.; Hoadley, K.A.; Rashid, N.U.; Williams, L.A.; Eaton, S.C.; Chung, A.H.; et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat. Genet. 2015, 47, 1168–1178. [Google Scholar] [CrossRef]
  109. O’Kane, G.M.; Grünwald, B.T.; Jang, G.H.; Masoomian, M.; Picardo, S.; Grant, R.C.; Denroche, R.E.; Zhang, A.; Wang, Y.; Lam, B.; et al. GATA6 Expression Distinguishes Classical and Basal-like Subtypes in Advanced Pancreatic Cancer. Clin. Cancer Res. 2020, 26, 4901–4910. [Google Scholar] [CrossRef] [Green Version]
  110. Rashid, N.U.; Peng, X.L.; Jin, C.; Moffitt, R.A.; Volmar, K.E.; Belt, B.A.; Panni, R.Z.; Nywening, T.M.; Herrera, S.G.; Moore, K.J.; et al. Purity Independent Subtyping of Tumors (PurIST), A Clinically Robust, Single-sample Classifier for Tumor Subtyping in Pancreatic Cancer. Clin. Cancer Res. 2020, 26, 82–92. [Google Scholar] [CrossRef]
  111. Bengtsson, A.; Andersson, R.; Rahm, J.; Ganganna, K.; Andersson, B.; Ansari, D. Organoid technology for personalized pancreatic cancer therapy. Cell Oncol. 2021, 44, 251–260. [Google Scholar] [CrossRef]
  112. Yu, K.H.; Ricigliano, M.; McCarthy, B.; Chou, J.F.; Capanu, M.; Cooper, B.; Bartlett, A.; Covington, C.; Lowery, M.A.; O’Reilly, E.M. Circulating Tumor and Invasive Cell Gene Expression Profile Predicts Treatment Response and Survival in Pancreatic Adenocarcinoma. Cancers 2018, 10, 467. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Kato, S.; Kim, K.H.; Lim, H.J.; Boichard, A.; Nikanjam, M.; Weihe, E.; Kuo, D.J.; Eskander, R.N.; Goodman, A.; Galanina, N.; et al. Real-world data from a molecular tumor board demonstrates improved outcomes with a precision N-of-One strategy. Nat. Commun. 2020, 11, 4965. [Google Scholar] [CrossRef] [PubMed]
  114. Grunvald, M.W.; Jacobson, R.A.; Kuzel, T.M.; Pappas, S.G.; Masood, A. Current Status of Circulating Tumor DNA Liquid Biopsy in Pancreatic Cancer. Int. J. Mol. Sci. 2020, 21, 7651. [Google Scholar] [CrossRef] [PubMed]
  115. Pishvaian, M.J.; Bender, R.J.; Matrisian, L.M.; Rahib, L.; Hendifar, A.; Hoos, W.A.; Mikhail, S.; Chung, V.; Picozzi, V.; Heartwell, C.; et al. A pilot study evaluating concordance between blood-based and patient-matched tumor molecular testing within pancreatic cancer patients participating in the Know Your Tumor (KYT) initiative. Oncotarget 2017, 8, 83446–83456. [Google Scholar] [CrossRef] [Green Version]
Table 1. Select trials evaluating efficacy and safety of first-line chemotherapy in metastatic pancreatic cancer.
Table 1. Select trials evaluating efficacy and safety of first-line chemotherapy in metastatic pancreatic cancer.
RegimenStudy DesignnMedian OSORRToxicityCommentsReference
Gemcitabine vs. 5-FUPhase III1265.6 months vs. 4.4 months
(p = 0.0025)
5.4% vs. 0%Neutropenia ≥ grade 3: 25.9 % vs.4.9% (p < 0.001)Survival beyond 12 months: 18% vs. 2%.
Clinical benefit, the primary efficacy measure of the study, 23.8% vs. 4.8% (p = 0.0022).
Burris (1997) [10]
Gemcitabine plus cisplatin vs. gemcitabinePhase III1957.5 months vs. 6.0 months
(p = 0.15)
10.2% vs. 8.2%Nausea and vomiting 22.2% vs. 5.9%; p = 0.0002Median PFS 5.3 vs. 3.1 months (p = 0.053)
Rate of SD 60.2% vs. 40.2%; p < 0.001
Heinemann (2006) [12]
Erlotinib plus gemcitabine vs. gemcitabinePhase III5696.24 months vs. 5.91 months (p = 0.038)8.6% vs. 8%Higher frequencies of rash, diarrhea, infection, and stomatitis in the erlotinib + gemcitabine arm.One-year survival was also greater with erlotinib plus gemcitabine (23% vs. 17%; p = 0.023)Moore
(2007) [15]
Gemcitabine + bolus 5-FU vs. GemcitabinePhase III3226.7 months vs. 5.4 months (p = 0.09).3.4 months vs. 2.2 months
(p = 0.02)
No significant differences between the two armsORR 6.9 % vs. 5.6%.Berlin (2002) [9]
Gemcitabine plus capecitabine vs. gemcitabinePhase III5337.1 months vs. 6.2 months
(p = 0.08)
19.1% vs.12.4%Grade ≥ 3 neutropenia 35% vs. 22%The median PFS: 5.3 vs. 3.8 months (p = 0.004)Cunningham (2009) [11]
FOLFIRINOX vs. gemcitabinePhase III34211.1 months vs. 6.8 months
(p < 0.001)
31.6% vs. 9.4%Febrile neutropenia −5.4% in FOLFIRINOX groupThe median PFS: 6.4 vs. 3.3 months (p < 0.001)Conroy
(2011) [7]
Nab-paclitaxel plus gemcitabine vs. gemcitabinePhase III8618.5 months vs. 6.7 months
(p <0.001)
23% vs. 7%Grade ≥ 3 toxicities: neutropenia: 38% vs. 27%.
Neuropathy: 17% vs. 1%.
The median PFS: 5.5 vs. 3.7 months (p < 0.001)Von Hoff
(2013) [8]
Nab-paclitaxel plus gemcitabine plus cisplatin Phase 1b/22516.4 months71%Grade ≥ 3 toxicities: Thrombocytopenia (68%), anemia (32%), and neutropenia (24%).The median PFS was 10.1 months.Jameson (2019) [16]
Gemcitabine, cisplatin plus veliparib vs. gemcitabine and cisplatinPhase II5015.5 vs. 16.4 months
(p = 0.6)
74.1 % vs. 65.2%Grade 3 to 4 hematologic toxicities:
neutropenia 48% vs. 30%.
thrombocytopenia 55% vs. 9%
Median PFS: 10.1 vs. 9.7 months.
The two-year OS rate for the entire cohort was 30.6% (95% CI, 17.8% to 44.4%).
O’Reilly (2020)
[17]
NALIRIFOX
(liposomal irinotecan + oxaliplatin + 5-FU/LV)
Phase I/II5612.6 months34.4%22 of 32 patients had grade ≥ 3 TRAEs, including neutropenia (31.3%), febrile neutropenia (12.5%) and hypokalemia (12.5%).Median PFS 9.2 months.Wainberg (2021) [18]
Abbreviations: OS, overall survival; ORR, overall response rate; 5-FU, 5-fluorouracil; PFS, progression-free survival; SD, stable disease; FOLFIRINOX, 5-FU, irinotecan and oxaliplatin; TRAE, treatment-related adverse events; LV, leucovorin.
Table 2. Published studies with maintenance treatment regimens in metastatic pancreatic ductal adenocarcinoma.
Table 2. Published studies with maintenance treatment regimens in metastatic pancreatic ductal adenocarcinoma.
StudyPatient PopulationnStudy DesignInduction TherapyMaintenance TherapyOutcomeComments
POLO [31]mPDAC with BRCA1 or 2 germline mutation154Randomized phase IIIAt least 16 weeks of first-line platinum-based chemotherapyOlaparib or placebomPFS 7.4 (olaparib) vs. 3.8 months (placebo) [HR, 0.53; 95% CI, 0.35–0.82; p = 0.004]Most patients received induction FOLFIRINOX (79.3 % in the Olaparib group and 71% in the placebo group)
PACT-12 [36]Unselected patients with mPDAC56Randomized Phase II6 months of first-line chemotherapyArm A = Observation
Arm B = sunitinib at 37.5 mg daily until progression or a maximum of 6 months
mPFS significantly longer with sunitinib maintenance (3.2 vs. 2 months, HR 0.51; 95% CI, 0.29–0. 89; p < 0.012-year OS was remarkably high with sunitinib (7.1% vs. 22.9%; p = 0.11)
Petrioli et al. [35]Locally advanced or mPDAC >70 years old36Prospective observational study3 cycles of gemcitabine and nab-paclitaxelGemcitabineSix-month DCR- 61%, mPFS-6.4 months,
and mOS-13.4 months.
Chevalier et al.
[33]
Unselected patients with mPDAC321Multicenter retrospective studyFOLFIRINOX
(Median 9 cycles)
FOLFIRI-45%,
5-FU or capecitabine-35%, FOLFOX 17%
mOS- 16.1 months.mOS and mPFS were similar between the groups receiving FOLFIRI or 5-FU
PANOPTIMOX-PRODIGE 35
[34]
Unselected patients with mPDAC276Randomized Phase II6 months of FOLFIRINOX (arm A), 4 months of FOLFIRINOX followed by 5-FU/LV maintenance treatment (arm B), or a sequential treatment alternating gemcitabine and FOLFIRI every 2 months (arm C).mOS: 10.1 months in arm A, 11.2 in arm B, and 7.3 in arm C.Median survival without deterioration in quality-of-life scores was higher in the maintenance arm (B) at 11.4 months than in arms A (7.2 months) and C (7.5 months).
Abbreviations: mPDAC, metastatic pancreatic ductal adenocarcinoma; mPFS, median progression-free survival; HR, hazard ratio; mOS, median overall survival; DCR, disease control rate; 5-FU, 5-fluorouracil; LV, leucovorin; FOLFIRINOX, 5-FU, irinotecan, and oxaliplatin; FOLFIRI, 5-FU, and irinotecan; FOLFOX, 5-FU, and oxaliplatin.
Table 3. Studies with second-line chemotherapy in patients with metastatic pancreatic ductal adenocarcinoma.
Table 3. Studies with second-line chemotherapy in patients with metastatic pancreatic ductal adenocarcinoma.
StudyStudy DesignnTreatment RegimenORR (%)Median PFS (Months)Median OS (Months)Additional Information
After progression on first-line FOLFIRINOX
Portal et al. [39]Prospective multicenter study57GA185.18.8Garde 3 or higher neurotoxicity 13%
Mita et al. [38]Phase II30GA133.87.670% of patients experienced grade 3 or 4 AEs
Viaud et al. [40]Retrospective study96Gemcitabine102.13.7DCR 40%
After progression on first-line gemcitabine
Wang-Gillam
et al. [41]
Phase III4175-FU/LV/Nal-IRI vs.
5-FU/LV
17 vs. 13.1 vs. 1.5
(p < 0.001).
6.1 vs. 4.2
(p = 0.012)
Estimated one-year overall survival rate was 26% with nal-IRI + 5-FU/LV.
Oettle et al. [42]Phase III160OFF vs. 5-FU/LV-2.9 vs. 2
(HR, 0.68;
p = 0.019)
5.9 vs. 3.3
(p = 0.01)
Grade 1/2 neurotoxicity, 38.2% vs. 7.1%
Gill et al. [43]Phase III108mFOLFOX6 vs.
5-FU/LV
13.2 vs. 8.53.1 vs. 2.9
(p = 0.99)
6.1 vs. 9.9
(p = 0.02)
Adverse events leading to study withdrawal- 20% vs. 2%
Kim et al. [44]Phase II39mFOLFIRINOX10.33.88.5Grade 3–4 neutropenia occurred
in 40% of patients
Sawada et al. [45]Retrospective104mFOLFIRINOX10.63.97 First-line regimen-GA.
Zaniboni et al. [46]Phase II50FOLFIRI83.256-mo survival rate-32%.
Xiong et al. [47]Phase II41XELOX2.52.56The survival rate at 1 year was 21%.
Ettrich et al. [48]Phase II44Docetaxel and oxaliplatin15.91.810.1DCR 47.7%
Abbreviations: ORR, overall response rate; PFS, progression-free survival; OS, overall survival; FOLFIRINOX, 5-FU/leucovorin/oxaliplatin/irinotecan;.GA, gemcitabine/abraxane; AE, adverse event; DCR, disease control rate; 5-FU, 5-fluorouracil; LV, leucovorin; Nal-IRI, nanoliposomal irinotecan; OFF, oxaliplatin/5-FU/leucovorin; HR, hazard ratio; m (modified)FOLFOX, 5-FU/leucovorin/oxaliplatin; mFOLFIRINOX, modified FOLFIRINOX; FOLFIRI, 5-FU/leucovorin/irinotecan; XELOX, capecitabine/oxaliplatin.
Table 4. Novel clinical trials with targeted therapy combinations.
Table 4. Novel clinical trials with targeted therapy combinations.
Target Pathway/GeneStudy Agents with the Mechanism of ActionStudy PhaseClinical Trial Identifier
KRAS G12CAdagrasib (KRAS G12C inhibitor) +
Cetuximab (anti-EGFR agent)
1bNCT03785249
Adagrasib +
BI-1701963 (SOS-1 inhibitor)
1/1bNCT04975256
(KRYSTAL-14)
Adagrasib +
TNO155 (SHP-2 inhibitor)
1/2NCT04330664
(KRYSTAL-2)
Adagrasib
Palbociclib (CDK 4/6 inhibitor)
1NCT05178888
(KRYSTAL-16)
KRAS G12DsiG12D-LODER in combination with chemotherapy2NCT01676259
(PROTACT)
Pan-KRASBI-1701963 (SOS-1 inhibitor)
+/− Trametinib (MEK inhibitor)
1NCT04111458
RMC-4630 (SHP-2 inhibitor) +
Cobimetinib (MEK inhibitor)
1/2NCT03989115
KRAS-based immunotherapyV941 (KRAS-targeted vaccine)
+/−Pembrolizumab
1NCT03948763
Poly-ICLC (a TLR3 Agonist) +
nivolumab & ipilimumab
1NCT04117087
LY3214996 (ERK inhibitor) +
Hydroxychloroquine (targets autophagy)
2NCT04386057
Trametinib
Hydroxychloroquine
1NCT04132505
(HOPE)
Binimetinib
Hydroxychloroquine
1NCT03825289
(THREAD)
Cobimetinib
Hydroxychloroquine
Atezolizumab
1/2NCT04214418
(MEKiAUTO)
Targeting HRDNiraparib
Dostarlimab
2NCT04493060
Olaparib
Pembrolizumab
2NCT04666740
(POLAR)
CX-5461 (RNA polymerase I transcription inhibitor)1NCT04890613
Targeting CDKN2A and MTAP deletionsAG-270 + GnP1NCT03435250
Targeting tumor metabolismSBP-101 + GnP1NCT03412799
SBP-101 + GnP2/3NCT05254171
L-glutamine + GnP1NCT04634539
GP-2250 + gemcitabine1/2NCT03854110
Targeting tumor stromaPamrevlumab (antibody against connective tissue growth factor)
+ GnP as first or second-line
2/3NCT04229004
(Precision Promise)
Paricalcitol
(Vitamin D analog)
2NCT04054362
Paricalcitol
Nivolumab
2NCT02754726
Paricalcitol
hydroxychloroquine
2NCT04524702
Paricalcitol + 5-Fu/LV/Nal-IRI1NCT03883919
+/− ATRA + GnP2NCT04241276
Defactinib
(FAK inhibitor) +
Pembrolizumab
2NCT03727880
Abbreviations: EGFR, epidermal growth factor receptor; HRD, homologous recombination deficiency; 5-FU/LV/Nal-IRI: 5-Fluorouracil/leucovorin/liposomal irinotecan; GnP, gemcitabine and nab-paclitaxel; ATRA: All-trans retinoic acid; FAK, focal adhesion kinase.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Chakrabarti, S.; Kamgar, M.; Mahipal, A. Systemic Therapy of Metastatic Pancreatic Adenocarcinoma: Current Status, Challenges, and Opportunities. Cancers 2022, 14, 2588. https://doi.org/10.3390/cancers14112588

AMA Style

Chakrabarti S, Kamgar M, Mahipal A. Systemic Therapy of Metastatic Pancreatic Adenocarcinoma: Current Status, Challenges, and Opportunities. Cancers. 2022; 14(11):2588. https://doi.org/10.3390/cancers14112588

Chicago/Turabian Style

Chakrabarti, Sakti, Mandana Kamgar, and Amit Mahipal. 2022. "Systemic Therapy of Metastatic Pancreatic Adenocarcinoma: Current Status, Challenges, and Opportunities" Cancers 14, no. 11: 2588. https://doi.org/10.3390/cancers14112588

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop