30,220
Views
28
CrossRef citations to date
0
Altmetric
Review Article

Pancreatic Cancer: A Review of Current Treatment and Novel Therapies

, , &
Article: 2129884 | Received 09 Jul 2022, Accepted 21 Sep 2022, Published online: 03 Oct 2022

Abstract

Pancreatic cancer is one of the leading causes for cancer-related deaths in the United States. Majority of patients present with unresectable or metastatic disease. For those that present with localized disease, a multidisciplinary approach is necessary to maximize survival and optimize outcomes. The quality and safety of surgery for pancreatic cancer have improved in recent years with increasing adoption of minimally invasive techniques and surgical adjuncts. Systemic chemotherapy has also evolved to impact survival. It is now increasingly being utilized in the neoadjuvant setting, often with concomitant radiation. Increased utilization of genomic testing in metastatic pancreatic cancer has led to better understanding of their biology, thereby allowing clinicians to consider potential targeted therapies. Similarly, targeted agents such as PARP inhibitors and immune checkpoint- inhibitors have emerged with promising results. In summary, pancreatic cancer remains a disease with poor long-term survival. However, recent developments have led to improved outcomes and have changed practice in the past decade. This review summarizes current practices in pancreatic cancer treatment and the milestones that brought us to where we are today, along with emerging therapies.

Background

Pancreatic ductal adenocarcinoma (PDAC) is the third overall leading cause for cancer-related mortality in the US [Citation1]. According to Surveillance, Epidemiology and End Result Program (SEER), roughly 57,600 new cases were diagnosed in 2020, resulting in 47,050 deaths [Citation2]. The poor prognosis associated with this disease is attributable to its early systemic spread and aggressive local growth. Nearly 50–60% of patients present with distant metastatic disease, 25–30% with regional disease and only 10–15% of patients present with local disease [Citation3]. Treatment and prognosis for patients with local or regional disease depend on resectability of the tumor. Incomplete resection (R2) is known to have a detrimental effect on survival, approaching those presenting with metastatic disease [Citation4–6]. Therefore, the selection of patients eligible for complete and curative resection is crucial. However, the definition of resectability – easily resectable versus borderline resectable – has varied amongst surgeons which led to a degree of heterogeneity in earlier study populations undergoing resection [Citation7]. Still today, definitions by expert panels differ slightly on what constitutes resectable disease () [Citation7, Citation8].

Table 1. Criteria for resectability per the National Comprehensive Cancer Network (NCCN) and MD Anderson Cancer Center (MDACC) [Citation7].

Although our understanding of this disease is still limited and overall prognosis poor, improvements in survival have been made in recent years. The quality of surgical care has improved, owing to increased expertise and the development of high-volume centers. More patients are candidates for resection owing to techniques that include arterial and venous resections and reconstruction. Minimally invasive surgery has found a role, with 15% of all pancreaticoduodenectomies being performed laparoscopically. Similarly, systemic therapy has evolved toward improved survival with growing interest in tumor genomics and targeted therapies. In this review, we will discuss the evidence behind current therapies along with novel treatments that are underway.

Surgical outcomes and adjuncts

Short term outcomes

The choice of surgery for pancreatic cancer depends on the location of the tumor in the pancreas. While tumors distal to the pancreatic head are usually amenable to a distal pancreatectomy, the majority of pancreas tumors, located in the head, require a pancreaticoduodenectomy for complete resection [Citation5]. For the purpose of this review, we will focus on outcomes related to the classic pancreaticoduodenectomy.

Pancreaticoduodenectomy (PD) has evolved since its debut in 1889 by the Italian dr. Codivilla [Citation9]. This complex procedure was not fully established until Dr. Whipple described it in his two-step approach for ampullary malignancies in a seminal article in 1935 [Citation10]. However, perioperative mortality remained high for decades. Early reports of post-operative mortality were as high as 30-45% [Citation11–13]. Today, mortality has vastly improved and is now at an acceptable 1-3% in high-volume centers [Citation13–16]. Nevertheless, short-term morbidity remains a significant issue [Citation11]. The incidence of pancreatic fistula has varied in published literature, related to lack of clear definition. Commonly quoted at 5-15%, this complication is now defined into grades A-C as per the international Study Group On Pancreatic Fistulas (ISGPF) [Citation17]. Delayed gastric emptying (DGE) is another serious complication that affects approximately 20% of patients [Citation18]. Similar to pancreatic fistulas, the International Study Group on Pancreatic Surgery has defined DGE according to severity into grades A-C [Citation19]. Both surgical technique and patient-related factors have been investigated for association with DGE. Eisenberg et al found intra-abdominal fluid collections and smoking history to be associated with DGE in their retrospective series of 721 patients [Citation18]. A meta-analysis by Hanna et al on surgical technique found antecolic position of the gastrojejunostomy limb as well as subtotal stomach preserving PD to be associated with lower incidence of DGE [Citation20]. Overall, DGE after PD remains a challenging issue in need of further study. With the advent of high-volume centers, outcomes have improved, prompting a recent attempt to define outcome benchmarks for pancreaticoduodenectomies [Citation21]. Sanchez-Vazquez et al [Citation21] derived benchmark values from operative and peri-operative data from 2,375 pancreaticoduodenectomies performed at high-volume centers around the world. Among those proposed standards included operative time ≤ 7.5 hours, ≤ 30% of patients suffering Clavien-Dindo [Citation22] grade ≥3 complications, in hospital mortality ≤1.6% and ≤9% readmission rate. These benchmarks represent a modern standard in surgical care for patients undergoing pancreaticoduodenectomy.

Oncologic outcomes

The only hope of curing pancreatic cancer involves a combination of complete resection and systemic multi-agent chemotherapy [Citation23]. Surgical outcomes have improved in the past two decades, largely due to increased surgical expertise at higher volume centers [Citation13, Citation15]. More patients are now considered for resection thanks to aggressive surgical techniques that include vascular resection and reconstruction [Citation24]. The median 5-year survival after surgery in modern reports is around 20% [Citation13, Citation23]. Some have reported higher survival rates of up to 30% with the latest multimodal therapy [Citation25, Citation26]. Overall recurrence rate, however, remains high at 70-80% [Citation27, Citation28]. Distant metastases after surgery are the main mode of disease recurrence with the liver being the most common site [Citation27–29]. Many believe this to be due to micro-metastases present at time of surgery. This is driving current practice toward treating patients with neoadjuvant chemotherapy before surgery, regardless of resectability [Citation30, Citation31]. More data are pending that will hopefully establish the true benefits, if any, of this approach in patients with resectable disease [Citation32]. Those with borderline resectable and locally advanced disease should receive neoadjuvant treatment before considering surgery [Citation33]. This approach has been reported to achieve more negative margin resection rates, as well as to convert few unresectable patients to resectable state.

Minimally invasive pancreaticoduodenectomy

The first report of a laparoscopic pancreaticoduodenectomy (LPD) was by Gagner and Pomp in 1994, although not for the indication of malignancy [Citation34]. Initial concerns regarding the laparoscopic approach centered around a long learning curve, increased operative time and potentially, complication rate. However, as this method gained popularity, early retrospective reviews generally reported favorable outcomes. A review summary of all reported LPDs (n = 146) in 2009 reported overall mortality at 1.3%, 16% complication rate, and average blood loss of 143 mL albeit a relatively long median hospital stay of 18 days, and a high conversion rate to open surgery at 43% [Citation35]. As experience with the procedure grew, indications expanded to include mesenteric vascular resections with reconstructions [Citation36, Citation37]. The only randomized clinical trial performed to date, addressed perioperative outcomes in their study of 66 patients (34 LPD, 32 OPD) in 2018. Among their significant findings was a benefit of the laparoscopic approach in terms of length of stay (13.5 vs 17 days) and post-operative complication burden [Citation38]. Two meta-analyses have since been published comparing LPD to open PD in 9,144 and 15,278 patients with pancreatic cancer respectively [Citation39, Citation40]. They reported rates for surgical mortality and pancreatic fistulas as similar between the two approaches while R0 resections and harvested lymph nodes were significantly better in the LPD group in both studies. Overall survival and 5-year survival were found to be similar between these approaches which have been supported by other studies as well [Citation39, Citation41–43].

Much like LPD, robotic pancreaticoduodenectomy (RPD) has mostly observational evidence to justify its use. A multi-institutional retrospective study by Zureikat and colleagues in 2016 compared perioperative outcomes from 211 RPDs and 817 OPDs [Citation44]. Their findings included no difference in mortality, hospital stay or re-admission rate. RPD was associated with less blood loss, less rate of severe complications and longer operative time. A meta-analysis of 11 comparative studies by Chen and colleagues further supported lower overall complication rate for patient undergoing RPD compared to OPD as well as lower operative blood loss, rate of wound infections and earlier activity after surgery without a difference in mortality.

Not all reports on minimally invasive PD (MIPD) have been favorable. Adams and colleagues compared actual national outcomes of patients undergoing LPD/RPD and OPD utilizing data from the National Cancer Database (NCDB) [Citation45]. Their findings showed that the majority of hospitals performing minimally invasive PDs were low-volume (<10 cases/2 years) and 30-day unadjusted mortality was 5.1% vs 3.1% for MIPD vs OPD, respectively [Citation45]. Additionally, Nussbaum et al investigated whether minimally invasive PD was superior to OPD in terms of patients receiving adjuvant therapy earlier, a theoretic advantage of the minimally invasive technique. Utilizing NCDB data from 2010-2012 they found no difference in time to adjuvant therapy or overall use of adjuvant therapy between those undergoing MIPD and OPD [Citation46]. Finally, the majority of available data has been from expert centers, limiting generalizability of reported outcomes. Nevertheless, enthusiasm for minimally invasive PD is growing, in 2012, approximately 15% of all PDs were being performed using minimally invasive techniques [Citation46].

Adjuncts to surgery

Given the need to achieve clear surgical margins for acceptable oncologic outcomes, mesenteric and portal vein resections have been investigated in multiple studies [Citation47–49]. Venous resections are often necessary in borderline resectable and locally advanced pancreatic cancer patients with vascular involvement, if a curative resection is to be achieved. Currently, approximately 4–20% of pancreaticoduodenectomies involve venous resections. Published data speaks to its safety in terms of short-term morbidity while incurring increased operating times and blood loss [Citation13, Citation24, Citation49]. From an oncologic standpoint, similar overall survival after venous resections has been described when compared to traditional PD despite tumor invasion into the portal vein/mesenteric vein on pathology [Citation48]. Involvement of adjacent arterial structures, such as the celiac axis (CA), common hepatic artery (CHA), and superior mesenteric artery (SMA) has traditionally constituted unresectable disease. However, data have been accumulating from expert centers on combined arterial resections, that speak to its feasibility in select cases [Citation49]. In the case of bulky pancreatic body tumors, the celiac axis can be resected either with or without reconstruction with acceptable outcomes [Citation50]. In fact, available data suggests that achieving an R0 resection impacts survival more than the extent of resection [Citation49, Citation50]. Chua et al, in their systematic review of all reports on vascular resections, found median overall and 3-year survival in patients undergoing combined venous and arterial resections to be 18 months and 13%, respectively. For optimal outcomes, vascular resections should be performed by surgeons at high-volume centers with experience in performing these procedures.

Irreversible electroporation (IRE), or Nanoknife ablation, is a non-thermal ablative technique that creates high voltage, short pulse electrical fields that induces local cellular apoptosis via micro-pores formed in the cell membrane [Citation51]. This method is emerging as a potential palliative option in unresectable, locally advanced PDAC given its ability to cause tumor destruction without compromising adjacent vessels or bile ducts [Citation52, Citation53]. A pilot study by Martin et al enrolled 27 patients with unresectable locally advanced pancreatic cancer and performed either resection of the primary tumor with IRE (NanoKnife®) margin accentuation (n = 8) or IRE alone (n = 19). Their findings at 3 months follow-up were consistent with complete tumor destruction in all patients without local recurrence, one patient (3.7%) died within that time and 33% morbidity was described [Citation52]. Other authors have reported improved outcomes when used as an adjunct to PD as well [Citation54]. We currently await the results of a randomized clinical trial that will compare survival of patients with stage 3 unresectable pancreatic cancer randomized to either FOLFIRINOX or FOLFIRINOX and IRE (NCT03899636).

Chemotherapy

Palliative chemotherapy for unresectable and metastatic PDAC patients

Patients with locally advanced or metastatic PDAC are generally considered non-curative and managed with palliative intent. Several trials have showed systemic chemotherapy improves overall survival in this setting. FOLFIRINOX was evaluated in the PRODIGE 4-ACCORD11 trial, comparing standard dose gemcitabine to FOLFIRINOX (5-Fluorouracil, Folinic acid, Irinotecan and Oxaliplatin) in patients with advanced PDAC. This phase 3, multicenter trial randomized 342 patients showed a median overall survival (mOS) benefit of 11.1 months vs. 6.8 months in favor of FOLFIRINOX [Citation55]. In another phase 3 trial (MPACT), 861 patients were randomized to either nab-paclitaxel and gemcitabine (Gem-nabP) or gemcitabine alone [Citation56]. mOS was superior in the Gem-nabP group (8.5 months vs 6.7 months) as well as progression-free survival (5.5 vs 3.7 months). There is no randomized head-to-head comparison of FOLFIRINOX and Gem-nabP, both are considered reasonable first-line treatments for unresectable PDAC. In clinical practice, FOLFIRINOX is generally reserved for fit patients whereas Gem-nabP is preserved for patients with mediocre performance, comorbidities or old age. There is evidence from retrospective data that FOLFIRINOX could improve OS, but causes more toxicity in vulnerable population [Citation57]. Therefore, gemcitabine monotherapy or FOLFOX are considered reasonable treatments for PDAC patients with poorer performance status (ECOG ≥2). After progression on first-line gemcitabine-based therapy, nanoliposomal irinotecan in combination with 5FU compared to 5FU alone was shown in the NAPOLI-1 study to have survival advantage of a mOS of 6.1 months vs 4.2 months) in PDAC patients [Citation58]. Now this combination is standard second-line therapy. About 5% of PDAC patients harbor germline BRCA mutations. Given the role of synthetic lethality and PARP inhibition in patients with DNA damage response (DDR) pathway aberrations, the POLO trial was conducted to investigate the efficacy of Olaparib (a PARP Inhibitor) as switch maintenance therapy in PDAC patients with germline BRCA mutations [Citation59]. Based on the significant mPFS advantage and QOL results (7.4 months vs. 3.8 months, HR 0.53), Olaparib maintenance is now approved for BRCA mutated PDAC patients. In the randomized phase 2 SEQUENCE study, standard gem-nabP (Days 1,8 15) followed by mFOLFOX (on day 29 of 6 weeks cycles) compared to standard gem-nabP, was studied in 153 metastatic PDAC patients [Citation60]. Results were intriguing with mOS 13.7 months vs 9.4 months, however grade 3 cytopenias were higher in the experimental arm. Patients receiving nab-P/Gem-mFOLFOX showed a significantly higher 12-month (55.3% vs. 35.4%), and 24- month (22.4% vs. 7.6%) survival. This could be a potential first-line option in metastatic PDAC treatment.

Adjuvant chemotherapy

Adjuvant chemotherapy after curative intent surgery in PDAC is evolving. Despite the survival benefit from singe agent adjuvant therapy with 5FU (ESPAC-1) or Gemcitabine (ESPAC-3, CONKO 001) [Citation59–61], two most recent trials dictating current practice are the ESPAC-4 and PRODIGE 24 [Citation61, Citation62]. The ESPAC-4 found gemcitabine/capecitabine combination to provide better OS (28 vs 25.5 months) compared to gemcitabine monotherapy in their trial of 732 patients [Citation61]. PRODIGE 24 was a large multi-national phase 3 trial comparing modified FOLFIRINOX regimen (reduced irinotecan and no bolus 5-FU) to gemcitabine in the adjuvant setting [Citation62]. The results showed improved mOS (54.4 months vs 35 months) in favor of mFOLFIRINOX. As expected, more toxicity was associated with mFOLFIRINOX. Modified FOLFIRINOX for 24 weeks remains the standard of care for good performance patients after surgical resection. Gemcitabine with or without Capecitabine is an option for those unable to tolerate mFOLFIRINOX.

Neoadjuvant therapy

Neoadjuvant chemotherapy offers several theoretical benefits over adjuvant chemotherapy. Those include the ability to address possible micro-metastases present at surgery by reaching target tissues more effectively, given intact perfusion of the tumor. Additionally, it has been shown to be better tolerated than adjuvant chemotherapy in other cancers, resulting in improved completion rates [Citation63, Citation64]. The high rate of margin positivity with resection of borderline resectable tumors (36%-64%) and subsequent worse survival has driven research on neoadjuvant therapies [Citation65]. In recent years, multiple trials have been conducted on patients with borderline resectable/locally advanced PDAC25 [Citation33, Citation66–70], and resectable cancers [Citation71–76]. A summary on the most recent trials on resectability, ability to achieve R0 resection and survival is listed in . In regards to resectable cancer, Lutfi et al evaluated data from the National Cancer Database on early stage PDAC patients (AJCC stage I/II) that underwent resection and received any type and timing of chemotherapy vs those who received no chemotherapy [Citation77]. Their findings showed early stage patients had better survival with neoadjuvant-, adjuvant chemotherapy or both compared to surgery alone. A meta-analysis by Versteijne et al, including 3,484 patients with resectable and borderline resectable disease receiving any type neoadjuvant chemotherapy with/without radiation therapy, found a modest OS benefit for those undergoing neoadjuvant treatment (18.8 vs 14.8 months) despite lower resection rates in the neoadjuvant treatment group (66% vs 81.3%) [Citation78]. Their results support that a subgroup of patients with local PDAC will develop aggressive disease progression and have no benefit from resection. Neoadjuvant treatment is thus emerging as a screening tool for PDAC patient who are truly localized and therefore benefits from surgical resection. Nevertheless, the role for chemotherapy in the neoadjuvant setting is still undefined. Larger, randomized trials are needed to compare the best regimens and further define the benefit in each clinical setting. Current NCCN guidelines recommend considering neoadjuvant treatment for borderline/locally advanced disease and preferred agents are FOLFIRINOXor Gemcitabine in combination with nab-paclitaxel [Citation8]. A large, randomized phase III trial involving 352 patients is active recruiting to investigate perioperative versus adjuvant chemotherapy for resectable PDAC patients (NCT04340141)

Table 2. Recent trials on neoadjuvant chemotherapy, resectability and outcomes.

Radiation

Adjuvant radiation

Adjuvant radiation therapy (RT) is widely utilized despite inconsistent evidence behind its efficacy. The GITSG study in 1985 was the first randomized study conducted to compare adjuvant chemoradiation (40 Gy in a split course with weekly bolus of 5-FU) to observation in 43 (22 vs 21) patients with negative margins after PDAC resection [Citation79]. Their results showed an overall survival benefit with CRT (median 20 months vs 11 months), however the trial closed early due to poor accrual (intended to accrue 100 patients). CRT thus became the new standard in adjuvant treatment. However, multiple randomized trials conducted after this challenged the survival benefit of CRT in the adjuvant setting [Citation80–83]. The ESPAC-1 trial, although highly criticized, went so far as to find chemoradiation detrimental to survival [Citation81]. We await the results of the ongoing RTOG 0848 (NCT01013649) trial that will hopefully add valuable data on whether the addition of radiation to chemotherapy is truly beneficial in the adjuvant setting of PDAC. Currently, the role for adjuvant chemoradiotherapy is unclear and we recommend evaluating patients for clinical trials. The current American Society of Radiation Oncology (ASTRO) guidelines recommend utilizing conventional fractionated adjuvant RT in the setting of high-risk tumor features, such as positive lymph nodes or margin positivity [Citation84].

Neoadjuvant radiation

Similar to chemotherapy, neoadjuvant radiation has its theoretical advantages. Given the high morbidity after PDAC surgery and early systemic recurrence, logic dictates increased efficacy of the neoadjuvant approach to radiation. Since the 1980s, researchers have only managed to find a modest benefit with neoadjuvant radiation. Most of these trials have been small and many aimed at establishing safety and feasibility of the treatments. Administered with either 5-FU or gemcitabine as radiosensitizers, its ability to improve local control and survival have been modest, both in resectable disease [Citation85–88] and borderline resectable/locally advanced [Citation89–91]. The 2018 PREOPANC-1 study provided better evidence for neoadjuvant chemoradiation in borderline resectable patients. They randomized 246 patients with borderline resectable PDAC into either neoadjuvant gemcitabine + 36 Gy radiation administered between 2 cycles of gemcitabine or upfront surgery with both groups receiving adjuvant chemotherapy. Although fewer patients in the chemoradiation arm underwent resection (62% vs 72%), they achieved significantly better R0 resection rate (65% vs 31%), disease-free survival (median 11.2 vs 7.9 months) and overall survival (median 17.1 vs 13.3 months) [Citation31]. The benefits of neoadjuvant chemoradiation have additionally been supported in a 2018 meta-analysis and patient data from the surveillance, epidemiology, and end results (SEER) registry [Citation78, Citation92].

In the 2019 American Society for Radiation Oncology guidelines, conventionally fractionated RT or SBRT is recommended with moderate evidence in borderline/locally advanced PDAC [Citation84]. However, there is low evidence to recommend RT in high-risk patients in the adjuvant setting or in unresectable patients.

Stratifying patients for adjuvant and neoadjuvant therapy in PDAC and tools for early detection of recurrence

We strongly encourage PDAC patients to participate in available clinical trials. Outside clinical trials, we routinely consider neoadjuvant chemotherapy for patients with resectable PDAC (resectable and borderline). In PDAC patients with high risk for recurrence (bulky tumors, lymph node positive, aggressive pathological features), we consider neoadjuvant chemotherapy and chemoradiation (total neoadjuvant therapy, TNT) approach. Based on emergent data and in our experience, we believe this approach will select “truly localized” PDAC patients and avoid a highly invasive procedure that will otherwise be futile. This approach also decreases the rate of disease recurrence based on emerging data. Stratification of localized PDAC patients for risk of relapse is key factor in adjuvant vs neoadjuvant therapy. Several factors such as tumor size, lymph node status, pathological features such as poorly differentiated histology, lymphovascular/perineural invasion, high CA 19-9 levels (>500), were historically used as biomarkers for recurrence risk stratification. Recently, more sophisticated biomarkers such as circulating tumor cells (CTC) and circulating tumor DNA (ctDNA) have been studied [Citation93, Citation94]. Recent advances in ctDNA technology could be a prognostic tool for early recurrence detection after surgery and chemotherapy [Citation95]. In a recent study, using a personalized and tumor-informed multiplex PCR assay (Signatera™), in a cohort of 93 localized PDAC patients with multimodality therapy, ctDNA positivity correlated with patient survival outcomes more strongly than CA19-9 [Citation96].

Metastasis directed treatment

Resection

Metastatic disease is the most common initial presentation of PDAC [Citation3]. For those who undergo curative resection, over 70% will have disease recurrence, most commonly at distant sites such as the liver, lungs, peritoneum [Citation27]. Although surgical resection is thought to be contraindicated in metastatic disease, expert centers around the world have attempted resection in highly selected patients with varying results. The largest published series on resection of PDAC metastases was reported by Hackert et al. [Citation97] They resected both liver (n = 85) and inter-aortocaval lymph node (ILN, n = 43) metastases in 128 patients between 2001 and 2014. Their median survival after resection was 12.3 months, with an actual 5-year survival of 5.9% and 7.0% after liver and ILN resection, respectively. A marginal improvement over palliative chemotherapy alone [Citation55]. Adams et al. retrospectively examined liver resections at 41 centers in France for non-colorectal, non-endocrine metastases, of which 41 patients were of pancreatic exocrine origin [Citation98]. They reported 20% 5-year survival for this subset of patients after liver resection. Additionally, a retrospective German study reported outcomes from 29 patients after synchronous resection of liver, peritoneum- or aortocaval lymph node metastases during pancreatic resections for PDAC [Citation99]. Ten of their patients had liver metastases, 11 lymph nodes and the rest peritoneal nodules. Their estimated median and 1-year survival was 13.8 months and 58.9%, respectively. There are those that have found no benefit to resection of PDAC metastases [Citation100]. Gleisner et al. reported similar survival in patients undergoing palliative bypass and liver resection of pancreatic or biliary cancers (5.9 months vs 5.6 months) [Citation100]. Perhaps the only scenario where resection of PDAC metastases has been consistently found to be beneficial is in the setting of lung metastases. Isolated metachronous lung metastases in PDAC are well known to incur better prognosis compared to other recurrence sites [Citation27]. Furthermore, their resection in select cases has been associated with improved survival in multiple retrospective series [Citation101, Citation102]. With the exception of lung metastases, survival after resection of metastatic disease varies and higher level of evidence is required to make recommendations. Resecting PDAC metastases is currently not routinely performed but can be considered in a trial setting.

Hepatic artery infusion therapy

Hepatic arterial infusion (HAI) therapy involves placing a catheter into the hepatic arterial vasculature (via gastroduodenal artery) supplying the liver and infusing chemotherapy directly into the liver. The benefits of HAI manifest in lower systemic toxicity and higher gradient of chemotherapy in the liver using certain agents [Citation101]. The benefits of HAI therapy in the setting of liver metastases from colorectal cancer have long been recognized [Citation102]. A few small studies have been conducted with HAI utilized as adjuvant treatment [Citation103–105] as well as for metastatic PDAC [Citation106]. They all have shown benefits to some extent. The non-randomized trial by Wang et al. compared 43 patients receiving adjuvant 2 cycles HAI chemotherapy followed by 4 cycles systemic chemotherapy (both consisting of 5-FU and gemcitabine) to 44 patients receiving adjuvant systemic gemcitabine + 5-FU alone [Citation105]. Although 5-year disease-free probability was the same for both groups, the HAI group had significantly better 5-year overall survival probability (hazard ratio (HR) 0.60) and hepatic metastases-free survival (HR 0.50). Ohigashi et al and Beger et al described 53% 3-year survival and 54% 4-year survival, respectively, with the use of HAI chemotherapy in the adjuvant setting [Citation103, Citation104]. Another small study by Tajima et al on patients with hepatic recurrence after resection showed a response rate in 6 out of 7 patients (85%) [Citation106]. There is currently a phase 2 trial ongoing using floxuridine-based HAI chemotherapy for patients with liver metastases after PDAC resection (NCT03856658).

Immuno- and targeted systemic therapy

Over the last decade, there has been a paradigm shift in PDAC therapeutics development. Next-generation sequencing technology and bioinformatics lead to discovery of driver mutations and aberrant pathways in PDAC. This process led to the identification of novel targets and there by therapeutics. Innovative targeted therapies, based on genomic results could potentially improve survival and quality of life in PDAC patients [Citation107]. PDAC patients with somatic/germ line mutations in DNA Damage Repair (DDR) pathway (BRCA 1,2, PALB2) homologous recombination repair (HRR) genes (ATM, BAP1, BARD1, BLM, BRIP1, CHEK2, FAM175A, FANCA, FANCC, NBN, RAD50, RAD51, RAD51C, and RTEL1) would comprise about 25% of PDAC patients. Recent meta-analysis involving six studies, with 21,842 PDAC patients, revealed that whole genome/whole exome sequencing allows the detection of greater proportion of patients with HRD (24–44%) compared to gene level hotspot/targeted NGS (14.5–16.5%). Prevalence of germline and somatic HRD mutations include BRCA1: 0.9%, BRCA2: 3.5%, PALB2: 0.2%, ATM: 2.2%, CHEK2: 0.3%, FANC: 0.5%, RAD51: 0.0%, and ATR: 0.1% [Citation108]. Detection of DDR pathway deficiency, in other words “BRCAness” in PDAC is important, as these patients, not only respond to platinum-based therapy, but also would benefit from PARP inhibitors, such as Olaparib [Citation105]. For comprehensive coverage of this topic and for a list of the ongoing clinical trial, please refer to an excellent review by Dalmasso et al. [Citation109]

Other major target for therapy in PDAC is KRAS, as it is mutated in about 95% of patients. KRAS codon 12 mutations are more common and constitute about 71%. These alterations include G12D (42%), G12V (32%), G12R (15%), G12C (1.5%), G12A (0.4%) and G12S(0.1%) [Citation110]. There is great momentum in KRAS-targeted therapeutics. In phase I/II trial, Sotorasib in heavily pretreated PDAC patients harboring KRAS G12C mutation revealed clinically meaningful benefit and safety (ORR 21.1% and DCR of 84.2%, mPFS of 4 months and mOS of 7 months) [Citation111]. Another G12C inhibitor Adagrasib showed the response rate of 50% and mPFS of 6.6 months (n = 10) in PDAC patients harboring G12C mutation [Citation112]. A phase I/II trial evaluated a KRAS G12D targeting RNAi released over 4 months from a small biodegradable tumor implant. Results were encouraging with a median OS of 15 months [Citation113]. Continued efforts at investigating KRAS as a therapeutic target are underway (NCT03608631, NCT01676259). Leidner et al. recently reported a single patient experience, who had a significant response (72% reduction of targeted tumor) after a single infusion of T cells that had been genetically engineered to clonally express two allogeneic HLA-C*08:02–restricted T-cell receptors (TCRs) targeting mutant KRAS G12D in tumor cells [Citation114]. This response continues to last for over 6 months. A list of major ongoing trials targeting KRAS can be seen in .

Table 3. List of selected early phase clinical trials involving targeted therapies in PDAC.

Historically, PDAC did not show any response to immunotherapy. This is most likely due to immunosuppressive tumor microenvironment (TME). About 2% of PDAC patients have mismatch repair deficiency (dMMR) and/or microsatellite instability (MSI-H). Based on results from KEYNOTE 158 study, Pembrolizumab has tumor agnostic approval for treatment in advanced cancer patients with MSI-H status. In this study, 22 patients with metastatic PDAC with dMMR and/or MSI-H were studied. Responses were encouraging (one complete response, ORR 18% and median duration of response 13.4 months) [Citation115]. Since PDAC is considered immunologically “cold”, there are some studies focusing on making tumors immunologically active by activating innate lymphocytes or creating inflammatory response in TME and there by bringing in cytotoxic T cells. Recently, Selvanesan et al injected non-disease-causing listeria bacteria carrying a gene encoding for tetanus toxin into mice model of pancreatic cancer. The tetanus toxin-induced memory T cells, which activated cytotoxic T cells in TME, which led to 80% decrease in tumor size, when then they added low dose Gemcitabine [Citation116]. Several clinical trials are investigating the role of chemo immunotherapy in PDAC. We are investigating the role of AZD 0171 (anti LIF1 monoclonal antibody- LIF1 an immune suppressor in TME in pancreatic cancer), durvalumab (PDL-1 inhibitor) in combination with Gemcitabine and Nab-paclitaxel in an open label phase II study (NCT04999969). Several immunological surrogate response markers such as tumor mutation burden (TMB), PDL-1, T cell response signatures are being investigated. Since germ line DDR gene mutations induces genomic instability and causes high TMB, PDAC patients with certain DDR mutations could benefit from immunotherapy. Hosein and colleagues conducted a small study with 12 PDAC patient harboring germ line HRD genetic mutations, which include BRCA1, BRCA2, RAD51C, RAD51D, and ATM, who failed on platinum-based chemotherapy and PARP inhibitors. They were treated with ipilimumab and Nivolumab for 4 cycles followed by Nivolumab. ORR was 42% and DCR of 58%. Complete response was observed in four patients (2 with BRCA1, 1 with BRCA2, 1 with RAD51C), partial response was observed in one (with BRCA1. 3 out 4 patients with CR discontinued treatment after 2 years [Citation117].

Next-generation sequencing (NGS) testing also has advantage of identifying genomic tumor drivers and targeted therapies. One such target is tropomyosin receptor kinase (TRK). Even though incidence of TRK fusions are rare in PDAC (about 0.5%), TRK Inhibitors such Larotrectinib and Entrectinib are now being used in the setting of PDAC as second-line therapy based on phase I and case series experience [Citation118, Citation119]. A comprehensive list of early phase clinical trials involving targeted therapies is shown in .

Discussion

In this overview of pancreatic cancer treatments, we have reviewed the latest developments in surgical and medical therapeutics for PDAC. Treatment options depend on the extent of disease at presentation. While those with resectable tumors at presentation have traditionally undergone surgery first, a paradigm shift is moving toward neoadjuvant chemotherapy first with emerging evidence in favor of total neoadjuvant treatment (chemotherapy followed by chemoradiation). Surgical options now include both minimally invasive approaches in select centers where experience has accumulated. Those with borderline resectable disease will benefit from neoadjuvant chemotherapy and chemoradiation. Provided a favorable response, these patients can undergo surgery although a more extensive resection to include vascular structures is often necessary to achieve an R0 resection. Patients with locally advanced or metastatic disease on presentation should undergo definitive chemotherapy with FOLFIRINOX, gem-nabP or gemcitabine + capecitabine +/- radiation. If patients respond to a sufficient degree, resection can be considered. For those with distant metastases, intra-abdominal metastases should generally not undergo resection unless in a trial setting while metachronous isolated lung metastases can be resected with favorable outcomes. For PDAC patients with isolated liver metastases, hepatic artery infusion pump is currently under investigation. Targeted therapeutics in PDAC are under investigation with the help of next-generation sequencing that allows genomic mapping of tumor cells for the identification of treatment targets. Currently, PARP inhibitors are utilized for tumors with DDR deficiency, agents targeting certain KRAS mutations have been shown to be efficacious in select patients as well as TRK inhibitors. Immunotherapy is currently utilized in the small cohort of PDAC patients with dMMR and/or high microsatellite instability (MSI-H) and multiple studies are ongoing on combination immuno- and chemotherapy.

Conclusion

In summary, pancreatic cancer remains a disease with dismal outcomes and benefits from new treatments have been modest so far. Nevertheless, outcomes continue to improve, and exciting new treatment avenues have opened in the last few years. Optimal care of patients with PDAC should be delivered with multidisciplinary input.

Disclosure statement

The authors have no conflicts of interest or financial ties to disclose.

References

  • Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA A Cancer J Clin. 2019;69(1):1–11. doi:10.3322/caac.21551.
  • Howlander N, Noone A, Krapcho M, et al. SEER cancer statistics review, 1975-2017. Bethesda, MD: National Cancer Institute; 2019; Based on November 2019 SEER data submission, posted to the SEER web site April 2020. https://seer.cancer.gov/csr/1975_2017/. Accessed April 1, 2022.
  • National Cancer Institute. Surveillance, Epidemiology and End Results Program. 2020. https://seer.cancer.gov/explorer/application.html?site=40&data_type=1&graph_type=4&compareBy=sex&chk_sex_1=1&race=1&age_range=1&advopt_precision=1. Accessed December 8, 2020.
  • Millikan KW, Deziel DJ, Silverstein JC, et al. Prognostic factors associated with resectable adenocarcinoma of the head of the pancreas. Am Surg. 1999;65(7):618–623.
  • Sohn TA, Yeo CJ, Cameron JL, et al. Resected adenocarcinoma of the pancreas - 616 patients: results, outcomes, and prognostic indicators. J Gastrointest Surg. 2000;4(6):567–579. doi:10.1016/S1091-255X(00)80105-5.
  • Raut CP, Tseng JF, Sun CC, et al. Impact of resection status on pattern of failure and survival after pancreaticoduodenectomy for pancreatic adenocarcinoma. Ann Surg. 2007;246(1):52–60. doi:10.1097/01.sla.0000259391.84304.2b.
  • Katz MHG, Marsh R, Herman JM, et al. Borderline resectable pancreatic cancer: need for standardization and methods for optimal clinical trial design. Ann Surg Oncol. 2013;20(8):2787–2795. doi:10.1245/s10434-013-2886-9.
  • National Comprehensive Cancer Network. Pancreatic Adenocarcinoma, Version 1.2020. www2.tri-kobe/nccn/guideline/pancreas/english/pancreatic.pdf. Published 2019. Accessed May 8 2020.
  • Schnelldorfer T, Sarr MG. Alessandro codivilla and the first pancreatoduodenectomy. Arch Surg. 2009;144(12):1179–1184. doi:10.1001/archsurg.2009.219.
  • Whipple AO, Parsons WB, Mullins CR. Treatment of carcinoma of the ampulla of vater. Ann Surg. 1935;102(4):763–779. doi:10.1097/00000658-193510000-00023.
  • Fernández-del Castillo C, Morales-Oyarvide V, McGrath D, et al. Evolution of the whipple procedure at the Massachusetts general hospital. Surgery. 2012;152(3 Suppl 1):S56–S63. doi:10.1016/j.surg.2012.05.022.
  • Gallitano A, Fransen H, Martin RG. Carcinoma of the pancreas. Results of treatment. Cancer. 1968;22(5):939–944. doi:10.1002/1097-0142(196811)22:5 < 939::aid-cncr2820220507 > 3.0.co;2-e.
  • Winter JM, Cameron JL, Campbell KA, et al. 1423 pancreaticoduodenectomies for pancreatic cancer: A single-institution experience. J Gastrointest Surg. 2006;10(9):1199–1211. doi:10.1016/j.gassur.2006.08.018.
  • Wright GP, Koehler TJ, Davis AT, Chung MH. The drowning whipple: perioperative fluid balance and outcomes following pancreaticoduodenectomy. J Surg Oncol. 2014;110(4):407–411. doi:10.1002/jso.23662.
  • Hata T, Motoi F, Ishida M, et al. Effect of hospital volume on surgical outcomes after pancreaticoduodenectomy: a systematic review and meta-analysis. Ann Surg. 2016;263(4):664–672. doi:10.1097/SLA.0000000000001437.
  • Macedo FIB, Jayanthi P, Mowzoon M, Yakoub D, Dudeja V, Merchant N. The Impact of surgeon volume on outcomes after pancreaticoduodenectomy: a meta-analysis. J Gastrointest Surg. 2017;21(10):1723–1731. doi:10.1007/s11605-017-3498-7.
  • Bassi C, Dervenis C, Butturini G, et al. Postoperative pancreatic fistula: an international study group (ISGPF) definition. Surgery. 2005;138(1):8–13. doi:10.1016/j.surg.2005.05.001.
  • Eisenberg JD, Rosato EL, Lavu H, Yeo CJ, Winter JM. Delayed gastric emptying after pancreaticoduodenectomy: an analysis of risk factors and cost. J Gastrointest Surg. 2015;19(9):1572–1580. doi:10.1007/s11605-015-2865-5.
  • Wente MN, Bassi C, Dervenis C, et al. Delayed gastric emptying (DGE) after pancreatic surgery: a suggested definition by the International Study Group of Pancreatic Surgery (ISGPS). Surgery. 2007;142(5):761–768. doi:10.1016/j.surg.2007.05.005.
  • Hanna MM, Gadde R, Allen CJ, et al. Delayed gastric emptying after pancreaticoduodenectomy. J Surg Res. 2016;202(2):380–388. doi:10.1016/j.jss.2015.12.053.
  • Sánchez-Velázquez P, Muller X, Malleo G, et al. Benchmarks in pancreatic surgery: a novel tool for unbiased outcome comparisons. Ann Surg. 2019;270(2):211–218. https://journals.lww.com/annalsofsurgery/Fulltext/2019/08000/Benchmarks_in_Pancreatic_Surgery__A_Novel_Tool_for.4.aspx.
  • Dindo D, Demartines N, Clavien PA. Classification of surgical complications: A new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg. 2004;240(2):205–213. doi:10.1097/01.sla.0000133083.54934.ae.
  • Strobel O, Lorenz P, Hinz U, et al. Actual five-year survival after upfront resection for pancreatic ductal adenocarcinoma: who beats the odds? Ann Surg. 2020;275(5):962–971. https://journals.lww.com/annalsofsurgery/Fulltext/9000/Actual_Five_year_Survival_After_Upfront_Resection.94393.aspx.
  • Müller SA, Hartel M, Mehrabi A, et al. Vascular resection in pancreatic cancer surgery: survival determinants. J Gastrointest Surg. 2009;13(4):784–792. doi:10.1007/s11605-008-0791-5.
  • Macedo FI, Ryon E, Maithel SK, et al. Survival outcomes associated with clinical and pathological response following neoadjuvant FOLFIRINOX or gemcitabine/nab-paclitaxel chemotherapy in resected pancreatic cancer. Ann Surg. 2019;270(3):400–413. doi:10.1097/SLA.0000000000003468.
  • Katz MHG, Wang H, Fleming JB, et al. Long-term survival after multidisciplinary management of resected pancreatic adenocarcinoma. Ann Surg Oncol. 2009;16(4):836–847. doi:10.1245/s10434-008-0295-2.
  • Kolbeinsson H, Hoppe A, Bayat A, et al. Recurrence patterns and postrecurrence survival after curative intent resection for pancreatic ductal adenocarcinoma. Surgery. 2021;169(3):649–654. doi:10.1016/j.surg.2020.06.042.
  • Groot VP, Rezaee N, Wu W, et al. Patterns, timing, and predictors of recurrence following pancreatectomy for pancreatic ductal adenocarcinoma. Ann Surg. 2018;267(5):936–945. doi:10.1097/SLA.0000000000002234.
  • Sperti C, Pasquali C, Piccoli A, Pedrazzoli S. Recurrence after resection for ductal adenocarcinoma of the pancreas. World J Surg. 1997;21(2):195–200. doi:10.1007/s002689900215.
  • Kim RY, Christians KK, Aldakkak M, et al. Total neoadjuvant therapy for operable pancreatic cancer. Ann Surg Oncol. 2021;28(4):2246–2256. doi:10.1245/s10434-020-09149-3.
  • Van Tienhoven G, Versteijne E, Suker M, et al. Preoperative chemoradiotherapy versus immediate surgery for resectable and borderline resectable pancreatic cancer (PREOPANC-1): A randomized, controlled, multicenter phase III trial. JCO. 2018;36(18_suppl):LBA4002–LBA4002. doi:10.1200/JCO.2018.36.18_suppl.LBA4002.
  • Janssen QP, van Dam JL, Bonsing BA, et al. Total neoadjuvant FOLFIRINOX versus neoadjuvant gemcitabine-based chemoradiotherapy and adjuvant gemcitabine for resectable and borderline resectable pancreatic cancer (PREOPANC-2 trial): study protocol for a nationwide multicenter randomized controlled t. BMC Cancer. 2021;21(1):300. doi:10.1186/s12885-021-08031-z.
  • Miyasaka Y, Ohtsuka T, Kimura R, et al. Neoadjuvant chemotherapy with gemcitabine plus nab-paclitaxel for borderline resectable pancreatic cancer potentially improves survival and facilitates surgery. Ann Surg Oncol. 2019;26(5):1528–1534. doi:10.1245/s10434-019-07309-8.
  • Gagner M, Pomp A. Laparoscopic pylorus-preserving pancreatoduodenectomy. Surg Endosc. 1994;8(5):408–410. doi:10.1007/BF00642443.
  • Gagner M, Palermo M. Laparoscopic Whipple procedure: review of the literature. J Hepatobiliary Pancreat Surg. 2009;16(6):726–730. doi:10.1007/s00534-009-0142-2.
  • Croome KP, Farnell MB, Que FG, et al. Pancreaticoduodenectomy with major vascular resection: a comparison of laparoscopic versus open approaches. J Gastrointest Surg. 2015;19(1):189–194; discussion 194. doi:10.1007/s11605-014-2644-8.
  • Kendrick ML, Sclabas GM. Major venous resection during total laparoscopic pancreaticoduodenectomy. HPB (Oxford). 2011;13(7):454–458. doi:10.1111/j.1477-2574.2011.00323.x.
  • Poves I, Burdío F, Morató O, et al. Comparison of perioperative outcomes between laparoscopic and open approach for pancreatoduodenectomy: the PADULAP randomized controlled trial. Ann Surg. 2018;268(5):731–739. doi:10.1097/SLA.0000000000002893.
  • Jiang Y-L, Zhang R-C, Zhou Y-C. Comparison of overall survival and perioperative outcomes of laparoscopic pancreaticoduodenectomy and open pancreaticoduodenectomy for pancreatic ductal adenocarcinoma: a systematic review and meta-analysis. BMC Cancer. 2019;19(1):781. doi:10.1186/s12885-019-6001-x.
  • Yin Z, Jian Z, Hou B, Jin H. Surgical and oncological outcomes of laparoscopic versus open pancreaticoduodenectomy in patients with pancreatic duct adenocarcinoma. Pancreas. 2019;48(7):861–867. doi:10.1097/MPA.0000000000001363.
  • Song KB, Kim SC, Hwang DW, et al. Matched case-control analysis comparing laparoscopic and open pylorus-preserving pancreaticoduodenectomy in patients with periampullary tumors. Ann Surg. 2015;262(1):146–155. doi:10.1097/SLA.0000000000001079.
  • Croome KP, Farnell MB, Que FG, et al. Total laparoscopic pancreaticoduodenectomy for pancreatic ductal adenocarcinoma: oncologic advantages over open approaches? Ann Surg. 2014;260(4):633–640. doi:10.1097/SLA.0000000000000937.
  • Stauffer JA, Coppola A, Villacreses D, et al. Laparoscopic versus open pancreaticoduodenectomy for pancreatic adenocarcinoma: long-term results at a single institution. Surg Endosc. 2017;31(5):2233–2241. doi:10.1007/s00464-016-5222-1.
  • Zureikat AH, Postlewait LM, Liu Y, et al. A multi-institutional comparison of perioperative outcomes of robotic and open pancreaticoduodenectomy. Ann Surg. 2016;264(4):640–649. doi: https://journals.lww.com/annalsofsurgery/Fulltext/2016/10000/A_Multi_institutional_Comparison_of_Perioperative.12.aspx.
  • Adam MA, Choudhury K, Dinan MA, et al. Minimally invasive versus open pancreaticoduodenectomy for cancer: practice patterns and short-term outcomes among 7061 patients. Ann Surg. 2015;262(2):372–377. doi:10.1097/SLA.0000000000001055.
  • Nussbaum DP, Adam MA, Youngwirth LM, et al. Minimally invasive pancreaticoduodenectomy does not improve use or time to initiation of adjuvant chemotherapy for patients with pancreatic adenocarcinoma. Ann Surg Oncol. 2016;23(3):1026–1033. doi:10.1245/s10434-015-4937-x.
  • Bold RJ, Charnsangavej C, Cleary KR, et al. Major vascular resection as part of pancreaticoduodenectomy for cancer: radiologic, intraoperative, and pathologic analysis. J Gastrointest Surg. 1999;3(3):233–243. doi:10.1016/S1091-255X(99)80065-1.
  • Ravikumar R, Sabin C, Abu Hilal M, et al. Portal vein resection in borderline resectable pancreatic cancer: a United Kingdom multicenter study. J Am Coll Surg. 2014;218(3):401–411. doi:10.1016/j.jamcollsurg.2013.11.017.
  • Chua TC, Saxena A. Extended pancreaticoduodenectomy with vascular resection for pancreatic cancer: a systematic review. J Gastrointest Surg. 2010;14(9):1442–1452. doi:10.1007/s11605-009-1129-7.
  • Okada K, Kawai M, Tani M, et al. Surgical strategy for patients with pancreatic body/tail carcinoma: Who should undergo distal pancreatectomy with en-bloc celiac axis resection? Surgery. 2013;153(3):365–372. doi:10.1016/j.surg.2012.07.036.
  • Rubinsky B, Onik G, Mikus P. Irreversible electroporation: a new ablation modality–clinical implications. Technol Cancer Res Treat. 2007;6(1):37–48. doi:10.1177/153303460700600106.
  • Martin RCG, 2nd, McFarland K, Ellis S, Velanovich V. Irreversible electroporation therapy in the management of locally advanced pancreatic adenocarcinoma. J Am Coll Surg. 2012;215(3):361–369. doi:10.1016/j.jamcollsurg.2012.05.021.
  • Maor E, Ivorra A, Leor J, Rubinsky B. The effect of irreversible electroporation on blood vessels. Technol Cancer Res Treat. 2007;6(4):307–312. doi:10.1177/153303460700600407.
  • Ansari D, Kristoffersson S, Andersson R, Bergenfeldt M. The role of irreversible electroporation (IRE) for locally advanced pancreatic cancer: a systematic review of safety and efficacy. Scand J Gastroenterol. 2017;52(11):1165–1171. doi:10.1080/00365521.2017.1346705.
  • Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364(19):1817–1825. doi:10.1056/NEJMoa1011923.
  • 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(18):1691–1703. doi:10.1056/NEJMoa1304369.
  • Raphael MJ, Raskin W, Habbous S, et al. The association of drug-funding reimbursement with survival outcomes and use of new systemic therapies among patients with advanced pancreatic cancer. JAMA Netw Open. 2021;4(11):e2133388–e2133388. doi:10.1001/jamanetworkopen.2021.33388.
  • Wang-Gillam A, Li C-P, 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 (London, England. 2016;387(10018):545–557. doi:10.1016/S0140-6736(15)00986-1.
  • Golan T, Hammel P, Reni M, et al. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med. 2019;381(4):317–327. doi:10.1056/NEJMoa1903387.
  • Carrato A, Vieitez JM, Benavides M, et al. Phase I/II trial of sequential treatment of nab-paclitaxel in combination with gemcitabine followed by modified FOLFOX chemotherapy in patients with untreated metastatic exocrine pancreatic cancer: Phase I results. Eur J Cancer. 2020;139:51–58. doi:10.1016/j.ejca.2020.07.035.
  • Neoptolemos JP, Palmer DH, Ghaneh P, et al. Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): a multicentre, open-label, randomised, phase 3 trial. Lancet. 2017;389(10073):1011–1024. doi:10.1016/S0140-6736(16)32409-6.
  • Conroy T, Hammel P, Hebbar M, et al. FOLFIRINOX or gemcitabine as adjuvant therapy for pancreatic cancer. N Engl J Med. 2018;379(25):2395–2406. doi:10.1056/NEJMoa1809775.
  • Cunningham D, Allum WH, Stenning SP, et al. Perioperative chemotherapy versus surgery alone for resectable gastroesophageal cancer. N Engl J Med. 2006;355(1):11–20. doi:10.1056/NEJMoa055531.
  • Sauer R, Becker H, Hohenberger W, et al. Preoperative versus postoperative chemoradiotherapy for rectal cancer. N Engl J Med. 2004;351(17):1731–1740. doi:10.1056/NEJMoa040694.
  • Yamada S, Fujii T, Sugimoto H, et al. Aggressive surgery for borderline resectable pancreatic cancer: evaluation of National Comprehensive Cancer Network guidelines. Pancreas. 2013;42(6):1004–1010. doi:10.1097/MPA.0b013e31827b2d7c.
  • Okada K-I, Hirono S, Kawai M, et al. Phase I study of nab-paclitaxel plus gemcitabine as neoadjuvant therapy for borderline resectable pancreatic cancer. Anticancer Res. 2017;37(2):853–858. doi:10.21873/anticanres.11389.
  • Reni M, Zanon S, Balzano G, et al. A randomised phase 2 trial of nab-paclitaxel plus gemcitabine with or without capecitabine and cisplatin in locally advanced or borderline resectable pancreatic adenocarcinoma. Eur J Cancer. 2018;102:95–102. doi:10.1016/j.ejca.2018.07.007.
  • Murphy JE, Wo JY, Ryan DP, et al. Total neoadjuvant therapy with FOLFIRINOX followed by individualized chemoradiotherapy for borderline resectable pancreatic adenocarcinoma: a phase 2 clinical trial. JAMA Oncol. 2018;4(7):963–969. doi:10.1001/jamaoncol.2018.0329.
  • Ferrone CR, Marchegiani G, Hong TS, et al. Radiological and surgical implications of neoadjuvant treatment with FOLFIRINOX for locally advanced and borderline resectable pancreatic cancer. Ann Surg. 2015;261(1):12–17. doi:10.1097/SLA.0000000000000867.
  • Katz MHG, Shi Q, Ahmad SA, et al. Preoperative modified FOLFIRINOX treatment followed by capecitabine-based chemoradiation for borderline resectable pancreatic cancer: alliance for clinical trials in oncology trial A021101. JAMA Surg. 2016;151(8):e161137. doi:10.1001/jamasurg.2016.1137.
  • Barbour AP, Samra JS, Haghighi KS, et al. The AGITG GAP study: a phase ii study of perioperative gemcitabine and nab-paclitaxel for resectable pancreas cancer. Ann Surg Oncol. 2020;27(7):2506–2515. doi:10.1245/s10434-020-08205-2.
  • OʼReilly EM, Perelshteyn A, Jarnagin WR, et al. A single-arm, nonrandomized phase II trial of neoadjuvant gemcitabine and oxaliplatin in patients with resectable pancreas adenocarcinoma. Ann Surg. 2014;260(1):142–148. doi:10.1097/SLA.0000000000000251.
  • Ielpo B, Duran H, Diaz E, et al. Preoperative treatment with gemcitabine plus nab-paclitaxel is a safe and effective chemotherapy for pancreatic adenocarcinoma. Eur J Surg Oncol. 2016;42(9):1394–1400. doi:10.1016/j.ejso.2016.01.006.
  • MacKenzie S, Zeh H, McCahill LE, PCRT, et al. A pilot phase II multicenter study of nab-paclitaxel (Nab-P) and gemcitabine (G) as preoperative therapy for potentially resectable pancreatic cancer (PC). JCO. 2013;31(15_suppl):4038–4038. doi:10.1200/jco.2013.31.15_suppl.4038.
  • Wei AC, Ou F-S, Shi Q, et al. Perioperative gemcitabine + erlotinib plus pancreaticoduodenectomy for resectable pancreatic adenocarcinoma: ACOSOG Z5041 (alliance) phase II trial. Ann Surg Oncol. 2019;26(13):4489–4497. doi:10.1245/s10434-019-07685-1.
  • Sohal D, Duong MT, Ahmad SA, et al. SWOG S1505: Results of perioperative chemotherapy with mFOLFIRINOX versus gemcitabine/nab-paclitaxel for resectable pancreatic ductal adenocarcinoma. JCO. 2020;38(15_suppl):4504–4504. doi:10.1200/JCO.2020.38.15_suppl.4504.
  • Lutfi W, Talamonti MS, Kantor O, et al. Perioperative chemotherapy is associated with a survival advantage in early stage adenocarcinoma of the pancreatic head. Surgery. 2016;160(3):714–724. doi:10.1016/j.surg.2016.05.029.
  • Versteijne E, Vogel JA, Besselink MG, et al. Meta-analysis comparing upfront surgery with neoadjuvant treatment in patients with resectable or borderline resectable pancreatic cancer. Br J Surg. 2018;105(8):946–958. doi:10.1002/bjs.10870.
  • Kalser MH, Ellenberg SS. Pancreatic cancer: adjuvant combined radiation and chemotherapy following curative resection. Arch Surg. 1985;120(8):899–903. doi:10.1001/archsurg.1985.01390320023003.
  • Klinkenbijl JH, Jeekel J, Sahmoud T, et al. Adjuvant radiotherapy and 5-fluorouracil after curative resection of cancer of the pancreas and periampullary region: phase III trial of the EORTC gastrointestinal tract cancer cooperative group. Ann Surg. 1999;230(6):776–776. doi:10.1097/00000658-199912000-00006.
  • Neoptolemos JP, Stocken DD, Friess H, et al. A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med. 2004;350(12):1200–1210. doi:10.1056/NEJMoa032295.
  • Smeenk HG, van Eijck CHJ, Hop WC, et al. Long-term survival and metastatic pattern of pancreatic and periampullary cancer after adjuvant chemoradiation or observation: long-term results of EORTC trial 40891. Ann Surg. 2007;246(5):734–740. doi:10.1097/SLA.0b013e318156eef3.
  • Van Laethem J-L, Hammel P, Mornex F, et al. Adjuvant gemcitabine alone versus gemcitabine-based chemoradiotherapy after curative resection for pancreatic cancer: a randomized EORTC-40013-22012/FFCD-9203/GERCOR phase II study. JCO. 2010;28(29):4450–4456. doi:10.1200/JCO.2010.30.3446.
  • Palta M, Godfrey D, Goodman KA, et al. Radiation therapy for pancreatic cancer: executive summary of an ASTRO clinical practice guideline. Pract Radiat Oncol. 2019;9(5):322–332. doi:10.1016/j.prro.2019.06.016.
  • Kim EJ, Ben-Josef E, Herman JM, et al. A multi-institutional phase 2 study of neoadjuvant gemcitabine and oxaliplatin with radiation therapy in patients with pancreatic cancer. Cancer. 2013;119(15):2692–2700. doi:10.1002/cncr.28117.
  • Shroff RT, Varadhachary GR, Crane CH, et al. Updated survival analysis of preoperative gemcitabine (gem) plus bevacizumab (bev)-based chemoradiation for resectable pancreatic adenocarcinoma. JCO. 2012;30(15_suppl):4051–4051. doi:10.1200/jco.2012.30.15_suppl.4051.
  • Evans DB, Varadhachary GR, Crane CH, et al. Preoperative gemcitabine-based chemoradiation for patients with resectable adenocarcinoma of the pancreatic head. JCO. 2008;26(21):3496–3502. doi:10.1200/JCO.2007.15.8634.
  • Talamonti MS, Small W, Mulcahy MF, et al. A multi-institutional phase II trial of preoperative full-dose gemcitabine and concurrent radiation for patients with potentially resectable pancreatic carcinoma. Ann Surg Oncol. 2006;13(2):150–158. doi:10.1245/ASO.2006.03.039.
  • Epelbaum R, Rosenblatt E, Nasrallah S, et al. Phase II study of gemcitabine combined with radiation therapy in patients with localized, unresectable pancreatic cancer. J Surg Oncol. 2002;81(3):138–143. doi:10.1002/jso.10159.
  • Joensuu TK, Kiviluoto T, Kärkkäinen P, et al. Phase I-II trial of twice-weekly gemcitabine and concomitant irradiation in patients undergoing pancreaticoduodenectomy with extended lymphadenectomy for locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys. 2004;60(2):444–452. doi:10.1016/j.ijrobp.2004.03.026.
  • Small WJ, Berlin J, Freedman GM, et al. Full-dose gemcitabine with concurrent radiation therapy in patients with nonmetastatic pancreatic cancer: a multicenter phase II trial. J Clin Oncol. 2008;26(6):942–947. doi:10.1200/JCO.2007.13.9014.
  • Stessin AM, Meyer JE, Sherr DL. Neoadjuvant radiation is associated with improved survival in patients with resectable pancreatic cancer: an analysis of data from the surveillance, epidemiology, and end results (SEER) registry. Int J Radiat Oncol Biol Phys. 2008;72(4):1128–1133. doi:10.1016/j.ijrobp.2008.02.065.
  • Effenberger KE, Schroeder C, Hanssen A, et al. Improved risk stratification by circulating tumor cell counts in pancreatic cancer. Clin Cancer Res. 2018;24(12):2844–2850. doi:10.1158/1078-0432.CCR-18-0120.
  • Court CM, Ankeny JS, Sho S, et al. Circulating tumor cells predict occult metastatic disease and prognosis in pancreatic cancer. Ann Surg Oncol. 2018;25(4):1000–1008. doi:10.1245/s10434-017-6290-8.
  • Sivapalan L, Kocher HM, Ross-Adams H, Chelala C. Molecular profiling of ctDNA in pancreatic cancer: opportunities and challenges for clinical application. Pancreatology. 2021;21(2):363–378. doi:10.1016/j.pan.2020.12.017.
  • Botta GP, Abdelrahim M, Aushev VN, et al. Association of personalized and tumor-informed ctDNA with patient survival outcomes in pancreatic adenocarcinoma. JCO. 2022;40(4_suppl):517–517. doi:10.1200/JCO.2022.40.4_suppl.517.
  • Hackert T, Niesen W, Hinz U, et al. Radical surgery of oligometastatic pancreatic cancer. Eur J Surg Oncol. 2017;43(2):358–363. 10.1016/j.ejso.2016.10.023.
  • Adam R, Chiche L, Aloia T, et al. Hepatic resection for noncolorectal nonendocrine liver metastases: analysis of 1,452 patients and development of a prognostic model. Ann Surg. 2006;244(4):524–535. doi:10.1097/01.sla.0000239036.46827.5f.
  • Shrikhande SV, Kleeff J, Reiser C, et al. Pancreatic Resection for M1 Pancreatic Ductal Adenocarcinoma. Ann Surg Oncol. 2007;14(1):118–127. doi:10.1245/s10434-006-9131-8.
  • Gleisner AL, Assumpcao L, Cameron JL, et al. Is resection of periampullary or pancreatic adenocarcinoma with synchronous hepatic metastasis justified? Cancer. 2007;110(11):2484–2492. doi:10.1002/cncr.23074.
  • Ilmer M, Schiergens TS, Renz BW, et al. Oligometastatic pulmonary metastasis in pancreatic cancer patients: Safety and outcome of resection. Surg Oncol. 2019;31:16–21. 10.1016/j.suronc.2019.08.010.
  • Yamashita K, Miyamoto A, Hama N, et al. Survival impact of pulmonary metastasis as recurrence of pancreatic ductal adenocarcinoma. Dig Surg. 2015;32(6):464–471. doi:10.1159/000439545.
  • Ensminger WD. Intrahepatic arterial infusion of chemotherapy: pharmacologic principles. Semin Oncol. 2002;29(2):119–125. doi:10.1053/sonc.2002.31679.
  • Groot Koerkamp B, Sadot E, Kemeny NE, et al. Perioperative hepatic arterial infusion pump chemotherapy is associated with longer survival after resection of colorectal liver metastases: a propensity score analysis. JCO. 2017;35(17):1938–1944. doi:10.1200/JCO.2016.71.8346.
  • Beger HG, Gansauge F, Büchler MW, Link KH. Intraarterial adjuvant chemotherapy after pancreaticoduodenectomy for pancreatic cancer: Significant reduction in occurrence of liver metastasis. In. World Journal of Surgery. 1999;23(9):946–949. doi:10.1007/s002689900604.
  • Ohigashi H, Ishikawa O, Eguchi H, et al. Feasibility and efficacy of combination therapy with preoperative and postoperative chemoradiation, extended pancreatectomy, and postoperative liver perfusion chemotherapy for locally advanced cancers of the pancreatic head. Ann Surg Oncol. 2005;12(8):629–636. doi:10.1245/ASO.2005.05.028.
  • Wang Y, Xu Y, Zheng Y, Bao Y, Wang P. Postoperative hepatic arterial infusion chemotherapy improved survival of pancreatic cancer after radical pancreatectomy: a retrospective study. Onco Targets Ther. 2018;11:903–907. doi:10.2147/OTT.S153886.
  • Tajima H, Kitagawa H, Tsukada T, et al. Hepatic arterial infusion chemotherapy with gemcitabine and 5-fluorouracil or oral S-1 improves the prognosis of patients with postoperative liver metastases from pancreatic cancer. Mol Clin Oncol. 2013;1(5):869–874. doi:10.3892/mco.2013.152.
  • Sohal DPS, Kennedy EB, Cinar P, et al. Metastatic pancreatic cancer: ASCO guideline update. JCO. 2020;38(27):3217–3230. doi:10.1200/JCO.20.01364.
  • Casolino R, Paiella S, Azzolina D, et al. Homologous recombination deficiency in pancreatic cancer: a systematic review and prevalence meta-analysis. J Clin Oncol. 2021;39(23):2617–2631. doi:10.1200/JCO.20.03238.
  • Dalmasso B, Puccini A, Catalano F, et al. Beyond BRCA: the emerging significance of DNA damage response and personalized treatment in pancreatic and prostate cancer patients. IJMS. 2022;23(9):4709. doi:10.3390/ijms23094709.
  • Registry. APG. Pancreatic Cancer Genomic Distribution: Mutated Genes. https://www.aacr.org/professionals/research/aacr-project-genie/aacr-project-genie-data/11-most-common-cancer-types-in-the-aacr-project-genie-registry/pancreatic-cancer/. Published 2022.
  • Strickler JH, Satake H, Hollebecque A, 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. JCO. 2022;40(36_suppl):360490–360490. doi:10.1200/JCO.2022.40.36_suppl.360490.
  • Bekaii-Saab TS, Spira AI, Yaeger R, 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. JCO. 2022;40(4_suppl):519–519. doi:10.1200/JCO.2022.40.4_suppl.519.
  • Golan T, Khvalevsky EZ, Hubert A, et al. RNAi therapy targeting KRAS in combination with chemotherapy for locally advanced pancreatic cancer patients. Oncotarget. 2015;6(27):24560–24570. doi:10.18632/oncotarget.4183.
  • Leidner R, Sanjuan Silva N, Huang H, et al. Neoantigen T-cell receptor gene therapy in pancreatic cancer. N Engl J Med. 2022;386(22):2112–2119. doi:10.1056/NEJMoa2119662.
  • 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):1–10. doi:10.1200/JCO.19.02105.
  • Chellakkan SB, Dinesh C, Wilber Q-T, et al. Listeria delivers tetanus toxoid protein to pancreatic tumors and induces cancer cell death in mice. Sci Transl Med. 2022;14(637):eabc1600. doi:10.1126/scitranslmed.abc1600.
  • Pishvaian MJ, Garrido-Laguna I, Liu SV, Multani PS, Chow-Maneval E, Rolfo C. Entrectinib in TRK and ROS1 fusion-positive metastatic pancreatic cancer. JCO Precis Oncol. 2018;(2):1–7. doi:10.1200/PO.18.00039.