Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Sep 1.
Published in final edited form as: CA Cancer J Clin. 2020 Jul 19;70(5):375–403. doi: 10.3322/caac.21626

Multidisciplinary standards of care and recent progress in pancreatic ductal adenocarcinoma

Aaron J Grossberg 1,2,3, Linda C Chu 4, Christopher R Deig 1, Eliot K Fishman 4, William L Hwang 5,6, Anirban Maitra 7, Daniel L Marks 2,8, Arnav Mehta 6,9, Nima Nabavizadeh 1, Diane M Simeone 10, Colin D Weekes 11, Charles R Thomas Jr 1
PMCID: PMC7722002  NIHMSID: NIHMS1615587  PMID: 32683683

Abstract

Despite tremendous gains in the molecular understanding of exocrine pancreatic cancer, the prognosis for this disease remains very poor, largely owing to delayed disease detection and limited effectiveness of systemic therapies. Incidence rates and mortality rates for pancreatic cancer have both increased during the past decade, in stark contrast to most other solid tumor types. Recent improvements in multimodality care have substantially improved overall survival, local control, and metastasis-free survival for patients with localized tumors amenable to surgical resection. The widening gap in prognosis between patients with resectable and unresectable or metastatic disease reinforces the importance of detecting pancreatic cancer sooner to improve outcomes. Furthermore, the developing use of therapies that target tumor-specific molecular vulnerabilities may offer improved disease control for patients with advanced disease. Finally, the substantial morbidity associated with pancreatic cancer, including wasting, fatigue, and pain, remains an underaddressed component of this disease, which powerfully impacts quality of life and limits tolerance to aggressive therapies. In this paper we review the current multidisciplinary standards of care in pancreatic cancer with a focus on emerging concepts in pancreatic cancer detection, precision therapy, and survivorship.

Background and Epidemiology

Pancreatic cancer has the poorest prognosis of any common solid malignancy, with a 5-year overall survival of approximately 10%.1 Although this represents a modest improvement in survival, the absolute number of individuals who succumb to this disease continues to rise. In 2020, it is estimated that 57,600 people will be diagnosed with and 47,050 deaths will be attributed to pancreatic cancer in the United States, recently eclipsing breast cancer as the 3rd leading cause of overall cancer death.2 The median age at diagnosis of pancreatic cancer is 70 years.1, 3 Incidence rates during 2013–2017 were higher among males than females (14.9 and 11.6 cases annually per 100,000 persons, respectively), as were mortality rates (12.7 and 9.6 deaths annually per 100,000 persons, respectively).1, 3 Incidence and mortality rates during this period were highest for blacks (15.3 cases and 13.3 deaths annually per 100,000 persons, respectively), followed by non-Hispanic whites (13.1 cases and 10.9 deaths annually per 100,000 persons, respectively), with lower rates among Hispanics, and especially among Asians/Pacific Islanders and American Indians/Alaska Natives.1 Lost earnings from person-years of life lost from pancreatic cancer in 2015 are estimated to be over $6 billion.4 Incidence and mortality rates both increased by an average of 0.3% per year during the past decade.1 Underlying these trends is a combination of an aging population, longer expected lifespan, and the public health pandemics of obesity and diabetes.

Approximately 95% of pancreatic cancers are exocrine cell tumors, most commonly ductal adenocarcinomas (PDAC). Endocrine pancreas cancers are generally more indolent tumors with a more favorable prognosis, reviewed elsewhere.5 There are four fundamental challenges that underlie the high mortality of PDAC. First, the pancreas is situated deep within the upper abdomen, seated behind the stomach and between the aorta and its major upper abdominal branches. Not only does this shield growing tumors from detection, but, because the cancer often grows around and encases these vessels, only 15–20% can undergo surgical resection, the foundation of curative treatment.6 Second, PDAC exhibits an aggressive biology characterized by early metastasis. Greater than 50% of patients have distant metastatic disease on presentation, and the majority of patients who undergo resection will develop metastases within 4 years of surgery, suggesting the de facto presence of micrometastases in patients with apparently localized tumors.1, 3, 7, 8 Third, the physiologic effects of PDAC can dramatically weaken patients, limiting their ability to withstand aggressive treatment. The wasting syndrome of cachexia is present in up to 80% of PDAC patients at diagnosis,9 and this may be further complicated by exocrine and endocrine pancreatic dysfunction.10, 11 Cachectic patients exhibit poor treatment tolerance, as evidenced by decreased survival following pancreatectomy12, 13 or chemotherapy.14, 15 Finally, PDAC exhibits resistance to many antineoplastic therapies, with rapid progression and low rates of pathologic complete response even with the most effective systemic agents and radiotherapy.16 Indeed, fewer than 3% of patients who present with metastatic disease are alive after 5 years, whereas this number jumps to over 70% in patients with localized, stage IA disease.17, 18 The impact of these challenges is reflected in the prognostic factors predicting poorer survival: advanced T-category, the presence of nodal metastasis or distant metastasis, the presence of gross or microscopic residual disease following resection, high histologic grade, invasion of major blood vessels, and poor performance status.1921

Presentation

Although most patients are symptomatic at presentation, symptoms of PDAC are often non-specific, leading to a median delay between presentation and diagnosis of over 2 months.22 The most commonly reported symptoms are fatigue (86%), weight loss (85%), anorexia (83%), jaundice (56%), nausea (51%), abdominal pain (79%), diarrhea (44%), pruritis (32%), and steatorrhea (25%).23 Clinical signs of PDAC, including jaundice (55%), hepatomegaly (29%), cachexia (13%), epigastric mass (9%), or ascites (5%), are much less common.23 This can make it difficult for primary care and front-line physicians to know when it is appropriate to escalate a workup, as there is no specified diagnostic algorithm for PDAC. The development of any of these symptoms in the context of newly diagnosed diabetes, a family history of PDAC, or a history of recurrent or chronic pancreatitis should alert the managing physician to strongly consider PDAC in the differential.24, 25 Recent evidence indicates that PDAC-associated metabolic disruptions, leading to skeletal muscle and adipose wasting, provide early evidence of PDAC growth.10, 26, 27

Risk Factors and Early Detection

Despite the poor prognosis of PDAC overall, data from the National Cancer Institute’s Surveillance, Epidemiology and End Results (SEER) program underscore the impact of earlier detection on outcomes.3 For example, analysis of SEER data between 2010–2016 shows that while only 2.9% of PDAC patients with distant metastases survived 5-years or beyond, 39.4% of patients with localized disease survived that duration. Patients with PDAC smaller than 1 cm who were diagnosed by endoscopic ultrasound (EUS) had even higher 5-year survival rates (reaching ~70% for patients with no obstructive symptoms or detectable “mass” on CT)18, while a more recent study from the United States of individuals at high risk for PDAC undergoing longitudinal surveillance has demonstrated that resecting advanced preneoplastic lesions is essentially curative.28 These findings reiterate the importance of earlier detection of PDAC, possibly even at the stage of carcinoma-in-situ (a.k.a. pancreatic intraepithelial neoplasia [PanIN]-3).

Nonetheless, the United States Screening and Preventive Task Force (USPSTF), an expert body charged with making recommendations for screening and other clinical preventive services for the nation, has recently reaffirmed its longstanding recommendation of discouraging screening for PDAC in the general population, concluding that, “there is moderate or high certainty that the service has no net benefit or that the harms outweigh the benefits” and thus service a failing “D” grade.29 On the one hand, the USPSTF guidelines sound counterintuitive in light of the aforementioned impact of earlier detection of PDAC on stage specific survival. On the other hand, there is compelling rationale against screening the general population for PDAC. At an incidence rate of ~13 cases per 100,000 adults,3 PDAC is still relatively uncommon. In contrast, the incidence rate of two cancers for which general population screening is recommended by the USPSTF - breast cancer and colorectal cancer – are ~69 cases and ~38 cases per 100,000 adults, respectively.3033 This implies that even a “perfect” PDAC biomarker with a sensitivity of 100% (i.e. not a single cancer being missed) and a specificity of 99% (one false positive of 100 abnormal tests), would only have a positive predictive value close to 1%34, 35, leading to a large number of individuals undergoing unnecessary imaging tests or potentially harmful procedures and adding greatly to healthcare costs and patient morbidity. In order to circumvent this pitfall, the USPSTF has excluded defined cohorts that are at higher than average risk for PDAC from its screening recommendation. In the following paragraphs, we will discuss some of these high-risk cohorts, and other emerging paradigms in early detection.

Approximately 10% of PDAC patients harbor a pathogenic germline mutation in a cancer predisposing gene, of which BRCA2 and ATM are the two most common candidates, followed by BRCA1, PALB2, CDKN2A/p16, LKB1/STK11, and the mismatch repair genes (hMLH1, hMSH2 and hPMS6), and other rarer variants (Table 1).36, 37 Of note, only half of the patients with a deleterious germline mutation report an overt family history of PDAC, in light of which the American Society of Clinical Oncology (ASCO) and the National Comprehensive Cancer Network (NCCN) recently updated their guidelines to recommend universal germline mutation testing for all patients diagnosed with PDAC (instead of only those with a suspicious family history).38, 39 This has the added benefit of identifying patients with BRCA1/2 mutations who might benefit from PARP inhibitor therapy (see later).40 The first-degree relatives of positive index cases can then be approached for testing done of the pathogenic mutation of interest (a process known as “cascade testing”), and the ready availability of relatively inexpensive blood and saliva-based multi-gene panels has greatly simplified the process.41 Asymptomatic germline mutation carriers represent a rich pool of high-risk individuals for cancer interception, and in fact, this subset has been explicitly excluded from the USPSTF recommendations against screening in the general population at average risk.29 There is currently no broad consensus on how to conduct longitudinal surveillance on germline mutation carriers, including the optimal imaging modalities to use in this population, although a recent international consortium has suggested some overall guidelines that can be pursued within an academic research setting.17 Nonetheless, retrospective data from two of the largest familial PDAC registries have shown the impact of longitudinal surveillance on outcomes, with the majority (75–90%) of incident cancer cases being diagnosed at a resectable stage, which in turn, translated into disease specific survival exceeding 3 years.17, 42

Table 1.

Pancreatic cancer susceptibility genes and estimated risk

Syndrome Gene Relative Risk
Familial atypical multiple mole melanoma (FAMMM) CDKN2A 13 to 39-fold37, 269, 270
Familial breast and ovarian BRCA1 and 2 2-fold and 3–9 fold37, 271
Fanconi anemia, Breast CA PALB2 Unknown272
Familial adenomatous polyposis (FAP) APC 5-fold273
Lynch syndrome MLH1, MSH6, MSH2, PMS2, EPCAM 9 to 11-fold37, 274
Peutz-Jeghers syndrome STK11/LKB1 132-fold275
Hereditary pancreatitis PRSS1 53-fold276
Li-Fraumeni syndrome p53 7-fold37
Ataxia-telangiectasia ATM ~3-fold37

A separate subgroup of patients, with pancreatic cysts, may benefit from early detection efforts as well. Pancreatic cysts are categorized as inflammatory (including pancreatic pseudocysts) and non-inflammatory (including mucinous and non-mucinous lesions).43 Mucinous cysts of the pancreas are comprised of two distinct entities – intraductal papillary mucinous neoplasms (IPMNs) and mucinous cystic neoplasms (MCNs), and both are considered bona fide precursor lesions of PDAC.44 In contrast to microscopic PanINs, which are the most common (~90%) precursor subtype associated with an invasive adenocarcinoma45, the macroscopic (cystic) precursors can be readily imaged using CT or MRI scans, and are thus amenable to longitudinal surveillance for progression to cancer. Notably, retrospective studies on imaging data have shown that as many as 2–3% of the general population might harbor asymptomatic pancreatic cysts, and this number rises to over 10-fold higher in elderly individuals.46 This “man-made” epidemic in pancreatic cysts can be attributed to the tens of millions of abdominal scans that are conducted each year in the United States for unrelated causes. Given that no more than 5–10% of PDAC annually arise in the backdrop of a cystic lesion, the vast majority of these asymptomatic pancreatic cysts are essentially benign and can be followed with conservative surveillance. The importance of early detection in cyst patients is underscored by the data that patients with non-invasive cystic lesions are usually cured upon surgical resection, while those with an invasive component can see their 5-year survival drop by 50% or greater.44 Therefore, identifying the minor subset of mucinous cysts that have either progressed to high-grade dysplasia or PDAC, or harbor an intrinsic biological potential for progression during the patient’s lifetime, is of paramount importance. However, based on SEER data, a cyst detected incidentally on MRI has a 17 in 100,000 chance of being a ductal cancer, indicating a very elevated number needed to surveil to prevent one premature death.47 Despite this, several international societies have published largely overlapping recommendations on cyst surveillance. The evidence driving these recommendations is derived from case series and retrospective reports, and graded as “very low quality”; thus it is unclear that the benefits of imaging surveillance outweigh potential harm.47 A compendium of clinical and imaging-based criteria have been suggested by various expert bodies (reviewed in48) that can support the clinician in their management decision making, but all have various shades of imperfections, resulting in some cases of both over-treatment and missed cancer diagnoses. Recently, molecular testing of endoscopically aspirated pancreatic cyst fluid for molecular biomarkers of mucinous cysts, as well as progression to cancer, have been implemented in the clinical domain, and have resulted in improved performance over the clinical/imaging criteria alone.49, 50 The application of machine learning algorithms towards designing an integrated approach to cyst classification will result in further accuracy in predicting underlying biology and improved management.51

A third high risk subset for PDAC are patients with chronic pancreatitis, most commonly secondary to chronic alcohol dependence, smoking, hypertriglyceridemia, diabetes, or renal failure.52 Approximately 5% of patients with chronic pancreatitis of 20 years duration will progress to PDAC, and concomitant smoking enhances the risk of neoplastic progression.53 Patients with sporadic chronic pancreatitis are currently not recommended to undergo PDAC surveillance under the USPSTF screening guidelines.29 However, the USPSTF recommendation against screening does not apply to a rare subset of patients with so-called “hereditary pancreatitis” secondary to germline mutations in the PRSS1 gene, which encodes for cationic trypsinogen.54 The mutation renders trypsin resistant to inactivation, resulting in recurrent episodes of acute pancreatitis beginning in childhood. These patients have a ~50-fold higher lifetime risk of PDAC55, redemonstrating the intimate link between inflammation and cancer.

While the aforementioned risk factors cumulatively impact ~15–20% of diagnosed PDAC patients annually, the majority still fall under the category of what would be considered as “sporadic” cancer (Table 2). How do we enable early detection in individuals with no apparent clinical risk factor like cysts or family history? One can certainly implement public health approaches like smoking avoidance and cessation, and maintaining a healthy body mass index (BMI), since both of these modestly (~1.5–2-fold) increase the lifetime risk of PDAC.56 The use of genome wide association studies (GWAS), where thousands of polymorphisms across the genome are compared in cases versus controls, has enabled the identification of multiple susceptibility alleles in PDAC, such as alleles within the ABO blood group genes and the gene encoding telomerase reverse transcriptase (TERT).57, 58 In contrast to deleterious germline mutations described above (BRCA1/2, ATM, etc.), these variant alleles individually only have a very modest impact on lifetime risk, but their cumulative risk could become appreciable. The incorporation of predisposing allelic information into a so-called “polygenic risk score” (PRS)59 could then identify individuals at the highest quartile of lifetime risk who could be enrolled into longitudinal surveillance programs. In the past decade, the emergence of new onset hyperglycemia or frank diabetes has been identified as the presenting symptom of an otherwise asymptomatic PDAC in up to half of all patients.60 The deregulation in glucose homeostasis is a paraneoplastic syndrome caused by the underlying PDAC, which can start to appear as early as 36 months prior to clinical diagnosis and is accompanied by changes in subcutaneous adipose tissue.26 Circulating factors, including antigen and microRNA panels, show promise in discriminating patients with early PDAC from controls or individuals with benign pancreatic conditions, offering the promise of liquid biopsy approaches toward PDAC early detection.6164 Additionally, recent data establishes that alterations in the gut and pancreatic microbiome directly promote pancreatic oncogenesis and influence survival, indicating that changes in fecal microbial composition may also help identify disease development.65, 66 In a “cancer interception paradigm of the future”, one can envision a pipeline wherein asymptomatic individuals are identified at higher than average lifetime risk for PDAC based on a combination of PRS, family history, smoking history, and BMI, and enrolled into a risk reduction program for PDAC (which includes guidance on lifestyle modification) and a surveillance program for individuals for whom benefit is anticipated to exceed harm. The onset of hyperglycemia or frank diabetes, as well as a catalog of longitudinally structured data readily available from the EMR would then “trigger” additional work up, such as imaging studies, in these high-risk individuals, eventually leading to earlier diagnosis of PDAC at a resectable stage and improved long term survival.

Table 2.

Risk factors for the development of pancreatic cancer

Factor Relative Risk
Tobacco Smoking 1.7 to 2.6-fold277279
Obesity 1.1 to 1.5-fold280, 281
Diabetes 1.5 to 2-fold11
Family History 1.7 to 2.3-fold282284
Chronic pancreatitis 13.3-fold285
*

Smoking appears to increase risk of PDAC in women more than men.286, 287

Diagnosis and Imaging

Diagnostic Workup

The diagnosis of PDAC cannot be made based on symptoms and signs alone. Patients presenting with jaundice or epigastric pain should be evaluated with complete blood count, blood chemistry panel, and liver function tests, including serum aminotransferases, alkaline phosphatase, and bilirubin. These values can help assess the extent of cholestasis (bilirubin), liver metastasis (alkaline phosphatase), hepatitis (aminotransferases), and nutritional status (albumin, prealbumin). Those with epigastric pain should also have serum lipase measured to evaluate for acute pancreatitis. The tumor marker sialylated Lewisa blood group antigen CA19–9 is frequently used in the workup for PDAC. In symptomatic patients, the sensitivity and specificity of CA19–9 range from 70–90%, but the positive predictive value of elevated CA19–9 in asymptomatic patients was only 0.9%, making it inadequate as a diagnostic in this population.35, 67, 68 Its limited utility is based on elevations in benign pancreaticobiliary diseases, cancers other than PDAC, and the fact that 5–10% of the population do not express Lewis antigens.69, 70 Emerging data suggest that the combination of serum CA19–9 with additional biomarkers, such as MUC5AC or thrombospondin-2 improves the specificity of serum testing, offering potential for a future blood-based diagnostic approach.63, 69 Serum CA19–9 levels are closely related to tumor size, and the degree of elevation in CA19–9 is associated with prognosis.68 In a study of patients with apparently localized disease, values above 130 units/mL predicted occult unresectable disease and were prognostic for survival among >1,500 patients with resectable cancers.71, 72 Although patients with apparently localized PDAC and high levels of CA19–9 are commonly recommended for staging laparoscopy and neoadjuvant therapy, ASCO guidelines do not specify a cutoff value of CA19–9 to be used in this manner.73 Because elevations in serum CA19–9 can be induced by either tumor production or cholestasis, CA19–9 should be remeasured after stent placement in patients with biliary obstruction to estimate “true” tumor burden, accounting for its 4–8 day half-life.74 Serial monitoring of CA19–9 is commonly used to track response to therapy in patients who present with elevated CA19–9. A failure in CA19–9 normalization after surgery is associated with poor survival and thought to represent occult metastatic disease.7577 Similarly, declining CA19–9 during systemic therapy correlates with improved patient survival, although it is unclear what magnitude of decline is most prognostic.78, 79 Rises in CA19–9 after a nadir can represent treatment failure and often precede imaging evidence of recurrent or progressive cancer.80 Serum CA19–9 changes are not considered to be a substitute for imaging evidence of treatment response or recurrence.81 In some tumors additional cancer-specific biomarkers, such as carcinoembryonic antigen or CA-125, are elevated and can also be used to track response to therapy and recurrence. Because these markers are elevated in only a subset of patients with PDAC, their utility in diagnosis is limited.82

Imaging Techniques

Computed tomography (CT) is the first-line imaging modality for initial evaluation of suspected PDAC and is preferred over magnetic resonance imaging (MRI) due to its lower cost and widespread availability.83 Both CT and MRI have comparable sensitivity in detection of PDAC, ranging from 76% to 96% for CT and from 83% to 94% for MRI.84 MRI is usually reserved as a second-line modality in patients with contraindications to CT (e.g. severe iodinated contrast allergy or renal insufficiency). MRI is also used as a problem-solving tool in cases with equivocal CT features and for characterization of indeterminate liver lesions.83 Position emission tomography/computed tomography (PET/CT) has been shown to detect extrapancreatic metastatic disease that was not detected based on traditional staging examination.85 Although not recommended as part of routine staging, PET/CT may be considered in patients at high-risk of extrapancreatic metastases.83 The primary role of endoscopic ultrasound (EUS) is to guide needle biopsies to confirm the diagnosis of PDAC. In select cases, EUS may be helpful in detecting a small pancreatic mass that may be difficult to visualize on CT or MRI, and is therefore the preferred imaging modality in some early detection surveillance programs.28, 86, 87

Reported accuracy in determining tumor resectability ranges from 73% to 87% for CT and from 70% to 79% for MRI.84 CT offers superior spatial resolution and is less susceptible to respiratory motion artifacts than MRI, which is essential in demonstrating the critical relationship between the tumor and adjacent vasculature. The accuracy of PDAC detection and staging critically depends on the appropriate imaging protocol, post-processing technique, and experience of radiologists.

Pancreatic cancer CT protocol, endorsed by both Society of Abdominal Radiology and American Pancreatic Association, states that CT examination should be performed with intravenous contrast (>300 mg I/mL) at an injection rate of 3–5 mL/s with scans obtained at pancreatic parenchymal phase (40–50 s) and portal venous phase (65–70 s). A neutral or low-Hounsfield unit oral agent should be administered. The dataset should be obtained with submillimeter slice thickness, reconstructed into 0.75 to 3 mm axial slices, with multiplanar reconstruction, and 3D reconstruction to allow for full assessment of vascular involvement.88 Cinematic rendering is a recently described 3D rendering technique that can provide photorealistic detail, and has the potential to improve visualization of tumor-vessel relationships (Figures 1 & 2).89

Figure 1.

Figure 1.

44-year-old woman with resectable PDAC arising from pancreatic body. A. Axial IV contrast-enhanced CT image shows a hypoenhancing pancreatic body mass (arrow) associated with pancreatic duct dilatation (white arrowhead). Tumor abuts splenic artery and splenic vein (black arrowhead). B. Coronal IV contrast-enhanced CT image with cinematic rendering improves conspicuity of pancreatic tumor (arrow) relatively to background pancreas and improves visualization of minimal tumor abutment of splenic artery (arrowhead).

Figure 2.

Figure 2.

50-year-old man with locally advanced PDAC arising from head of pancreas. A, B. Axial IV contrast-enhanced CT images show (A) tumor (arrow) with SMA encasement (arrowhead), and (B) tumor (arrow) with encasement and near occlusion of portal vein (arrowhead). C. Coronal IV contrast-enhanced CT image with cinematic rendering improves visualization of tumor encasement (arrow) of SMA (white arrowhead) and portal vein (black arrowhead).

Staging Systems

The American Joint Committee on Cancer (AJCC) stages PDAC based on a TNM staging system. The revised 8th edition of the AJCC manual addressed some of the criticisms of earlier versions with changes to T and N categories.90 T categories are mostly based on tumor size. T4 is defined as tumor with arterial involvement regardless of size. N categories are further classified in the 8th edition based on absence of lymph node involvement (N0) and number of regional lymph nodes involved (1–3 for N1, ≥4 for N2), rather than only absence (N0) or presence (N1) of lymph nodes.90 The primary goal of AJCC system is to provide prognostic information, rather than guiding management. A number of organizations have issued management guidelines.83, 9193 NCCN guidelines classify the resectability of localized PDAC based on preoperative imaging findings into resectable, borderline resectable, and locally advanced disease, and are summarized in Table 3. Arterial abutment (<180°) is considered borderline resectable, whereas arterial encasement (≥180°) is usually considered locally advanced (exception noted below) (Figure 2). Venous abutment, encasement, or thrombosis are considered borderline resectable, as long as the venous segment is reconstructable. Unreconstructable venous involvement is considered locally advanced.83 NCCN guidelines share many common features with other guidelines (Table 4) with notable exception of celiac artery encasement (>180°). A subset of patients with pancreatic body or tail tumor with celiac artery encasement, but without involvement of the aorta or gastroduodenal artery, can be considered borderline resectable with a modified Appleby procedure, a distal pancreatectomy with en bloc resection of the celiac axis (Figure 3). However, in other guidelines, the presence of celiac artery encasement is considered unresectable.

Table 3.

NCCN Criteria in Defining Resectability of PDAC at Diagnosis83

Resectability Status Arterial Involvement Venous Involvement
Celiac Artery SMA Common Hepatic Artery Portal Vein/SMV
Resectable
  • None

  • None

  • None

  • None

  • ≤180° contact without contour irregularity

Borderline Resectable
  • ≤180°

  • >180°, without involvement of aorta or GDA (body/tail)

  • ≤180°

  • Solid tumor contact without extension into CA or hepatic artery bifurcation

  • >180°

  • ≤180° with contour irregularity or thrombosis, with reconstructible PV/SMV

  • Solid tumor contact with IVC

Locally Advanced
  • >180° (head/uncinate)

  • Solid tumor contact with CA and aorta

  • >180°

  • Unreconstructible PV/SMV due to tumor involvement or occlusion

Table 4.

Comparison of Resectability Criteria among Organizations

Vessel Involvement NCCN 201983 MD Anderson91 ACTO92 AHPBA/SSAT/SSO93
CA Abutment (≤180°) Borderline Borderline Borderline Unresectable
CA Encasement (>180°) Borderline (body/tail)
Locally Advanced (head/uncinate)
Unresectable Unresectable Unresectable
SMA Abutment (≤180°) Borderline Borderline Borderline Borderline
SMA Encasement (>180°) Locally Advanced Unresectable Unresectable Unresectable
CHA Abutment or Encasement Borderline Borderline Borderline Borderline
PV/SMV Encasement (>180°) or Abutment (≤180°) with Contour Abnormality Borderline Borderline Borderline Borderline

ACT = Alliance for Clinical Trials in Oncology; AHPBA = American Hepato-Pancreato-Biliary Association; SSAT = Society for Surgery of the Alimentary Tract; SSO = Society for Surgical Oncology

Figure 3.

Figure 3.

70-year-old man with borderline resectable PDAC arising from pancreatic body. A. Axial IV contrast-enhanced CT image in maximum intensity projection shows tumor (arrow) encasement of celiac artery (arrowhead), common hepatic artery, and splenic artery. B. Sagittal IV contrast-enhanced CT image shows tumor encasement of the celiac artery (arrow) and sparing of the SMA (arrowhead), which allows for modified Appleby procedure.

Principles of Multidisciplinary Treatment

Metastatic Disease Therapy: An Evolving Treatment Paradigm

Metastatic disease represents the most common clinical presentation of PDAC.2 Historically, gemcitabine was the standard of care for the first-line treatment of metastatic disease based on a 5.65 month improvement in median overall survival (OS) in comparison to fluorouracil (5FU).94 Major studies establishing standard of care first-line therapy for PDAC are listed in Table 5. In 2011, the PRODIGE 4/ACCORD 11 phase III trial demonstrated an improved median OS and median progression-free survival (PFS) in patients treated with FOLFIRINOX (fluorouracil, leucovorin, irinotecan and oxaliplatin) in comparison to those receiving gemcitabine as first-line therapy for metastatic PDAC (median OS: 11.1 v 6.8 mo, hazard ratio (HR) 0.57, p<0.001 and median PFS: 6.4 v 3.3 mo, HR 0.47, p<0.001).95 FOLFIRINOX now embodies standard first-line therapy for fit patients. The combination of gemcitabine plus nab-paclitaxel represents another first-line therapy for the disease. The phase III MPACT trial demonstrated an improvement of 1.8 months in both median OS (HR 0.72, p<0.001) and median PFS (HR 0.69, p<0.001), respectively for patients with metastatic PDAC treated with gemcitabine plus nab-paclitaxel compared to gemcitabine.96 Collectively, these studies demonstrate that patients with metastatic PDAC both tolerate and benefit from aggressive combination chemotherapy. In the absence of a head-to-head comparison of these treatments, many providers select therapy based on patient performance status and ability to adhere to the treatment schedule, with FOLFIRINOX considered the more challenging, but perhaps more effective regimen.

Table 5.

Trials evaluating chemotherapy for metastatic pancreatic cancer.

Trial N Chemotherapy MS
Cullinan et al. (1985)288 144 5FU vs 5FU+dox vs 5FU+dox+mitomycin 5.5 vs 5.5 vs 4.5 mo. NS
Burris et al. (1997)94 126 5FU vs Gem 4.4 vs 5.6 mo. P=.0025
Tempero et al. (2003)289 92 Gem vs Gem (fixed rate) 5 vs 8 mo. P=.013
Heinemann et al. (2006)290 195 Gem ± cisplatin 6.0 vs 7.5 mo. P=.015
NCIC CTG PA.3 (2007)291 569 Gem ± erlotinib 5.9 vs 6.2 mo. P=.038
Cunningham et al. (2009)292 533 Gem ± capecitabine 6.2 vs 7.1 mo. P=.08
CALGB 80303293 602 Gem ± bevacizumab 5.9 vs 5.8 mo. P=.95
SWOG S0205 (2010)294 745 Gem ± cetuximab 5.9 vs 6.3 mo. P=.23
PRODIGE 4/ACCORD 11 (2011)95 342 Gem vs FOLFIRINOX 6.8 vs 11.1 mo. P<.0001
MPACT (2013)96 861 Gem ± nab-paclitaxel 6.7 vs 8.5 mo. P<.001

NS = not significant; dox = doxorubicin; Gem = gemcitabine.

Second-line therapy primarily consists of doublet therapy utilizing the alternative pyrimidine backbone to what was used in the first-line setting. Patients receiving FOLFIRINOX for first-line therapy are commonly transitioned to gemcitabine plus nab-paclitaxel as second line therapy based upon the finding of the MPACT study.97, 98 In contrast, patient receiving gemcitabine plus nab-paclitaxel in the first-line setting generally are treated with FOLFIRINOX, FOLFOX (5FU and oxaliplatin), or 5FU plus NALIRI (nanoliposomal irinotecan).95, 98100 These additional options are important for patients with impaired performance status or those who exhibit dose-limiting toxicities precluding FOLFIRINOX treatment. FOLFOX has been evaluated in several trials demonstrating a median PFS of ~ 3.5 months.98, 99 However, the randomized phase III PANCREOX study, which compared second-line FOLFOX against infusional 5FU/leucovorin found that the FOLFOX was associated with similar PFS, but worse OS and toxicity than the comparison arm, calling into question the advantage of adding oxaliplatin in this setting.99 The randomized phase III NAPOLI-1 trial demonstrated an improved OS of 6.1 months associated with NALIRI treatment in comparison to 4.2 months for 5FU (HR for death 0.67, p = 0.012). The use of 5FU as the control versus FOLFOX is a common criticism of this study, but perhaps unwarranted, given the PANCREOX results.100

The addition of new combinations of cytotoxic chemotherapies is being evaluated as an optimization strategy. The results of a phase IB/II study of gemcitabine, nab-paclitaxel and cisplatin as first-line therapy demonstrated the safety of the regimen, whilst providing early evidence of potential clinical benefit, with a remarkable PFS of 10.1 months.101 Although these results suggest increased efficacy with the addition of platinum, these data should be interpreted with caution in the absence of phase III data.

Maintenance Therapy

The improved outcomes associated with multimodality chemotherapy has led to prolonged exposure of patients to chemotherapy-related toxicities. The PANOPTIMOX-PRODIGE 35 phase II study was designed to evaluate the role of employing a strategy of abbreviated FOLFIRINOX plus maintenance FU or sequential gemcitabine and FOLFIRI3 versus continuous FOLFIRINOX until disease progression to mitigate oxaliplatin induced neuropathy.102 Treatment with alternating gemcitabine and FOLFIRI3 demonstrated inferior clinical outcomes compared to the FOLFIRINOX regimens. Patients treated with FOLFIRINOX were randomly assigned to continuous treatment for 6 months or 4 months followed by 5FU maintenance with reintroduction of FOLFIRINOX at disease progression. The primary endpoint of the study was 6-month PFS. Treatment with the FOLFIRINOX regimens resulted in equivalent 6-month PFS and OS. Patients treated with maintenance 5FU therapy experienced increased grade 3/4 neurotoxicity, which may be reflective of a higher cumulative oxaliplatin dose in these patients. Retrospective single-institution data out of Germany reports 11 month PFS in patients receiving maintenance FOLFIRI, delivered after 4 months of FOLFIRINOX.103 Importantly, these studies demonstrate that maintenance therapy is a viable treatment strategy that is not associated with inferior clinical outcomes in comparison to continued therapy until disease progression.

The POLO study was a phase III study that addressed the role of maintenance targeted therapy in a biomarker selected patient population. Patients with metastatic PDAC harboring deleterious germline BRCA1 or BRCA2 mutations who had not progressed on first-line platinum-based chemotherapy were randomized to receive either the poly(adenosine diphosphate-ribose) polymerase (PARP) inhibitor olaparib or placebo as maintenance therapy.40 Olaparib maintenance therapy improved the median PFS of 7.4 months versus 3.8 months in the placebo group (HR 0.53, p=0.004), but did not improve the median OS.40 Although this represents the first trial supporting molecular-guided therapy for PDAC, interpretation of this study is limited by both a low patient number and comparison against a no-treatment control arm, which is not standard of care. However, the POLO trial has led to the FDA approval of olaparib for maintenance therapy in patients with germline BRCA mutant disease.

Precision Oncologic Approaches to Pancreatic Cancer

With the increased analysis of the genomic signatures of PDAC, PDAC has been reliably classified into two distinct molecular subtypes of basal/quasi-mesenchymal or classical subtype. The classical subtype is characterized by an epitheloid phenotype; whereas, the basal/quasi-mesenchymal subtype has a mesenchymal phenotype that has a propensity to metastasize.104106 The COMPASS trial demonstrated that patients with the classical subtype demarcated by high levels of GATA6 expression by RNASeq or RNA in situ hybridization (ISH) in baseline tumor specimens have a higher response rate to FOLFIRINOX therapy, leading to an improved mPFS.107 Consistent with these findings, acquired resistance to FOLFIRINOX was associated with the development of basal phenotype upon FOLFIRINOX exposure.108, 109 Prospective studies are now required to confirm these observations. If these observations hold true, the choice of chemotherapy may be determined by the predominant molecular phenotype of the tumor.

Patients with either germline or somatic deleterious mutations resulting in homologous DNA repair deficiency (HRD) represent another important subset of patients likely to require specific therapies. HRD patients comprise upwards of 15 to 20% of patients with metastatic PDAC36, 110 As outlined previously, these patients may be specifically sensitive to PARP inhibition. The PARP inhibitor rucaparib has demonstrated clinical activity as a single agent in patients with advanced metastatic PDAC with both germline and somatic mutations in BRCA1/BRCA2.111 In addition, there are a number of small molecular inhibitors in development targeting various aspects of the homologous DNA repair pathway. Combination strategies incorporating multiple agents targeting the HRD pathway in combinations with PARP inhibition are in development.112 In addition to these effects, PARP inhibition possesses radiosensitizing properties.113 Platinum drugs induce double-strand DNA breaks resulting in increased sensitivity of HRD pancreatic tumors to this class of chemotherapy. Consistent with this observation, a randomized phase II study of gemcitabine plus cisplatin with or without veliparib as first-line therapy in HRD patients demonstrated response rates of 74% and 65% and median OS of 16 and 15 months, respectively.114

PDAC harboring microsatellite instability represents approximately 1% of patients afflicted with the disease.115 However, it is important to that all patients be tested for microsatellite instability because 40% of patients who have microsatellite instability respond to immune checkpoint inhibition and derive improved survival from this therapy.116, 117 This paradigm of tailoring therapeutic approaches to the molecular vulnerabilities of each cancer is known as precision medicine. In PDAC, where so few agents demonstrate clinically meaningful activity, precision medicine approaches hold substantial promise. In support of precision medicine in PDAC, a recent publication on the Know Your Tumor program, found that patients with actionable molecular alterations who received matched molecular therapies had significantly longer median OS than patients who received unmatched therapies (2.58 years vs 1.51 years, HR 0.42, p=0.0004).118 Furthermore, this study demonstrated the feasibility of molecular assessment on biopsy samples in real time. As capacity for molecular testing improve, such precision approaches may become the standard for the subset of patients with actionable molecular drivers.

Investigational Therapeutic Approaches

Treatments Targeting the Tumor Microenvironment

The PDAC tumor microenvironment (TME) consists of a complex network of cells including cancer associated fibroblasts (CAFs), immune cells, endothelial cells plus a dense extracellular matrix.119122 The inhibitory immune checkpoint programmed death ligand-1 (PD-L1) is expressed on myeloid cells within the TME and on tumor cells.123 High expression of PD-L1 expression in PDAC predicts a poor prognosis, yet immune checkpoint blockade (ICB) has been largely an ineffective therapeutic strategy. Anti-CTLA4 inhibition with ipilimumab or tremelimumab either as a single agent or in combination with gemcitabine and anti-PD-L1 (BMS-936559 or durvalumab) demonstrated no evidence of clinical activity.124127 Combined CXCR4 and ICB is currently under investigation as an approach to overcome immune exclusion of effector T cells based on preclinical data linking CXCR4 blockade to tumor infiltration of effector T-cells.128, 129 CD40 agonism represents another promising approach of ICB. CD40 is a component the costimulatory cascade resulting in upregulation of MHC class I and costimulatory molecules and skewing of myeloid cells to a tumoricidal phenotype.130133 A phase I study of the CD40 agonist APX005M in combination with gemcitabine and nab-paclitaxel with or without nivolumab demonstrated early evidence of clinical benefit with a PR rate of 58% and DCR of 91%.134 Lastly, ionizing radiation can induce immune priming through the production of toxic nucleotide adducts, enhancing intratumoral antigen presentation by dendritic cells.135, 136 An initial evaluation of this hypothesis has been conducted in 25 patients with metastatic PDAC by combining radiation therapy with nivolumab and ipilimumab with overall response rate (ORR) of 14% and median PFS of 2.5 months on interim analysis.137 The value of this approach will be further tested in future studies.

The extracellular matrix (ECM), comprised of a collagenous matrix and glycosaminoglycans such as hyaluronic acid (HA), is not inert—specifically, hyaluronic acid promotes metastasis and PDAC initiation.138 Recombinant human pegylated hyaluronidase, peg-PH20, was developed as an ECM-targeted strategy. Phase II or III studies evaluating peg-PH20 in combination with both FOLFIRINOX and gemcitabine plus nab-paclitaxel in patients with metastatic PDAC expressing high HA levels by immunohistochemistry failed to demonstrate the benefit of adding peg-PH20 to either chemotherapy backbone.139, 140 In fact, the combination of peg-PH20 with FOLFIRINOX led to excessive toxicity.140 Stroma modification remains a challenging therapeutic approach, however recent work evaluating angiotensin receptor blockade using losartan in patients with locally advanced pancreatic cancer show therapeutic promise (see later).141, 142

Treatments Targeting Metabolism and Autophagy

PDAC arising from PanIN is characterized by activating mutations in KRAS.143 The ensuing mitogen activated protein kinase (MAPK) activation results in a hypermetabolic state characterized by increased glycolysis, as well as increased metabolic plasticity via altered glutamine metabolism, dependence on oxidative phosphorylation, and metabolite scavenging through the process of macropinocytosis.144147 Autophagy is a cellular process designed to allow cells to utilize cellular components as an alternative fuel source in response to cellular stress. Autophagy is integral for PDAC oncogenesis and proliferative capacity.148150 Recently, two independent research teams demonstrated that MAPK activation regulates PDAC dependency on autophagy.151, 152 Therefore, phase I clinical trials are now ongoing or in development to evaluate combining either MEK or ERK inhibitors with the autophagy inhibitor, hydroxychloroquine (NCT04132505 and NCT03825289).

Resectable Pancreatic Cancer

Surgery

Surgical resection is currently the only means to achieve long-term survival in patients with PDAC. Although only 15–20% of patients present with resectable disease, the increasing use of neoadjuvant therapies and advances in surgical techniques have broadened the pool of patients who are eligible for surgical resection.73 The goals of care for patients with resectable PDAC are increasing the likelihood of margin-negative (R0) resection, decreasing procedural morbidity and mortality, preventing metastatic spread, and improving the patient’s quality of life. As surgical quality, peri-operative care, and systemic therapies have improved, the ultimate goal is to determine the optimal timing and sequencing of high-quality surgery in the appropriate patient as part of a multi-modality treatment plan.

A step by step review of pancreatectomy is beyond the scope of this review, but key steps to emphasize include: 1) a thorough exploration of the abdomen to rule out metastatic disease, 2) biopsy and examination by frozen section of any suspicious lesions outside of the field of resection, and 3) careful management of the surgical margin to ensure adequate tumor clearance, with the uncinate or retroperitoneal margin the margin most at risk. Careful dissection along the periadvential plane of the SMA mitigates the risk of a positive margin at this location during a pancreaticoduodenectomy (Figure 4).

Figure 4.

Figure 4.

The Whipple procedure (pancreaticoduodenectomy) for resectable PDAC in the head of the gland. A. Normal anatomic relationship of the pancreas and surrounding structures. B. Diagram of a pancreatic head mass (white) to surround the pancreatic and bile ducts (shown in green). C. Standard resection for pancreaticoduodenectomy to include the head of the pancreas, duodenum, distal common bile duct, distal stomach, and gallbladder. D. Reconstruction to reconnect the pancreas, common bile duct, and stomach to the gastrointestinal tract.

While mortality rates from pancreatectomy have fallen significantly and are below 2% in numerous high-volume centers world-wide, elevated morbidity remains common and still impacts the delivery of adjuvant therapy in up to 40% of patients.153, 154 It is recommended that patients seek out high volume centers with multi-disciplinary expertise to optimize their treatment plan and increase opportunities for clinical trial participation. The use of minimally invasive pancreatic resection has been expanding in the last decade. Excellent outcomes in some studies have been reported, however others cite safety concerns, particularly for the use of minimally invasive pancreaticoduodenenctomy.155157 In all cases, it is clear that implementation of rigorous dedicated training programs and experience with open pancreatic surgical techniques are needed to ensure safe use of these techniques in clinical practice and to determine if they will improve longer term oncologic outcomes.

Pathology

Rigorous pathologic assessment is essential for accurate prognosis as well as determining the appropriate adjuvant treatment plan. Despite guidelines from the College of American Pathologists and NCCN on standard protocols for the pathologic analysis of PDAC surgical specimens, this practice remains inconsistent, owing both to vagueness in the protocols and variable degrees of adherence across institutions.158 Protocols for specimen orientation and inking should be well-established between the surgeon and pathologist to ensure clear definition of key margins, generally using different colored inks (specific recommendations found at83, 159). The definition of involved margins are variable across institutions, with most centers in the US defining a positive margin as “tumor on ink”, whereas in Europe margins are called positive if tumor cells fall within 1 mm of the inked margin.160 Such inconsistency may help to explain the broad variation in reported R0 resection rates across studies.161 This distinction is significant for determining prognosis, as resection margin distance correlates closely with locoregional failure and survival162, 163 The pathology report should also include maximal tumor diameter (for staging), histologic subtype, tumor grade, and the presence of lymphovascular or perineural invasion. For those patients who have undergone neoadjuvant therapy, there are multiple histopathologic systems to evaluate treatment response.164166 Unlike other solid tumor types, the relationship between neoadjuvant treatment effect and prognosis is less clear in PDAC, possibly owing to the lack of standardization in this practice (reviewed in167). The most important prognostic factors for patients who have undergone curative-intent resection are the presence of lymph node metastasis and the ratio of positive lymph nodes to total lymph nodes.168, 169 Because the number of negative lymph nodes and total lymph node count influence stage-based survival prediction, the College of American Pathologists recommends microscopic evaluation of at least 12 lymph nodes for pancreatectomy specimens.159, 170, 171 Some groups advocate for reporting the involvement of specific lymph node groups, such as the hepatic artery lymph nodes, however the prognostic value of this practice remains controversial in light of contradictory data from retrospective studies.172174

Adjuvant Chemotherapy

The median OS of patients with localized PDAC who are treated with surgery alone is 11–20 months.7, 8, 175 Currently both NCCN and ASCO recommend 6 months of adjuvant systemic chemotherapy for all patients who undergo pancreatectomy.83 Multiple randomized studies demonstrated consistent improvement in median OS and disease-free survival (DFS) using a variety of fluoropyrimidine-based chemotherapies. In general, the selection of chemotherapeutic agents has closely followed those found superior in the metastatic setting. The evolution of adjuvant chemotherapy regimens is outlined in Table 6. More recently the PRODIGE-24 trial demonstrated an improved DFS for modified FOLFIRINOX treatment of nearly 9 months in comparison to gemcitabine (HR 0.58, CI 0.46 to 0.73, p <0.001), establishing it as standard of care in fit patients.7 Unfortunately, the combination of gemcitabine with nab-paclitaxel failed to meet its primary endpoint of improved DFS in the APACT trial.176 Tolerance of adjuvant therapy remains a limitation with patients commonly receiving < 50% of planned dose.7, 8, 176, 177 This observation is a reflection of exposure to significant chemotherapy-related toxicity in patients experiencing substantial post-pancreatectomy morbidity. This has led many centers to move toward a total neoadjuvant approach to systemic therapy, discussed below.

Table 6.

Trials evaluating adjuvant chemotherapy for resectable pancreatic cancer

Trial Chemotherapy MS
ESPAC-18 5FU vs Obs MS 21 vs 15.5 mo. P=0.009
CONKO-001175 Gemcitabine vs Obs MS 22.8 vs 20.2 mo. P=0.01
ESPAC-3295 Gem vs 5FU/LV MS 46 vs 39 mo. (ns)
ESPAC-4177 Gem vs Gem/Cape MS 28 vs 25.5 mo. P=0.03
PRODIGE-247 mFOLFIRINOX vs Gem MS 54.4 vs 35 mo. P=0.003
APACT176* Gem/Abraxane vs Gem MS 40.5 vs 36.2 mo. P=0.045
*

Presented as abstract

Radiation

Although the role of systemic therapy in the adjuvant setting for resected PDAC has been well-validated, the value of adjuvant (chemo)radiation to improve local control, and in turn survival, is strongly debated. The initial results of the 43 patient GITSG study suggested a substantial benefit to chemoradiotherapy, but the failure of subsequent randomized studies to reproduce this result may be attributable to the inclusion of adjuvant chemotherapy in this study, which would later be established as standard of care in the ESPAC-1 trial (Table 7). Although no modern level 1 evidence supports the role of adjuvant radiation in PDAC, multiple large retrospective observational database studies from both SEER and the National Cancer Database (NCDB) demonstrate improved OS in patients who receive adjuvant radiation therapy, particularly patients with pN+ disease or microscopically positive surgical margins.178181 Thus, at many centers adjuvant chemoradiation is reserved for patients who exhibit these risk factors after upfront surgical resection and adjuvant chemotherapy. This practice is being tested again in Radiation Therapy Oncology Group (RTOG) 0848, in which patients with no evidence of disease progression following R0 or R1 resections and multiple cycles of adjuvant chemotherapy were randomized to sequential chemoradiation or systemic chemotherapy alone. This closed trial accrued patients between 2009 and 2018, and results of the primary endpoint (OS) and stratified analyses of patient subgroups (by nodal status, margin status, pre-treatment CA 19–9 levels, adjuvant systemic treatment regimens) are eagerly awaited.

Table 7.

Trials evaluating adjuvant radiotherapy for resectable pancreatic cancer

Trial Comparison RT dose Concurrent Chemo Adjuvant Chemo OS
GITSG 9173296 ChemoRT + chemo vs Obs 40 Gy (split-course) Bolus 5FU 5FU MS 20 vs 11 mo. P=0.03
EORTC 40891297 ChemoRT vs Obs 40 Gy (split-course) CI 5FU -- MS 17.1 vs 12.6 mo. P=0.099
ESPAC-18 ChemoRT vs Obs 40 Gy (split-course) Bolus 5FU +/− 5FU MS 16 vs 18 mo. (ns)
RTOG 0848298 ChemoRT vs Obs 50.4 Gy CI 5FU or capecitabine Gemcitabine +/− erlotinib Not reported

To allow for standardized adjuvant radiotherapy techniques, RTOG has developed a contouring guideline in the adjuvant setting.182 Standard high-risk regions include the anastomoses (hepatico-, pancreatico-, and gastro-jejunostomies) and regional paraaortic nodal volumes near the root of the superior mesenteric and celiac arteries. The focal targeting of microscopic residual disease following pancreatectomy is impossible; thus, the contents of adjuvant radiation volumes consist solely of normal tissue, with maximum prescription doses constrained entirely by the tolerance of radiosensitive endoluminal organs (stomach, duodenum and jejunum). An example of the dose distribution in the adjuvant setting is shown in Figure 5. As a result, standard radiation prescriptions in the adjuvant setting consist of daily treatments over the course of 5–6 weeks to a total dose of 50–54 Gy. Given this and other factors, neoadjuvant regimens consisting of targeted pancreatic tumoral delivery of radiation while maximizing normal tissue avoidance is attractive. These approaches will be discussed in more detail below.

Figure 5.

Figure 5.

76 year-old woman with a large PDAC of the pancreatic tail underwent distal pancreatectomy and splenectomy revealing an 8.5 cm invasive carcinoma with a microscopically positive proximal pancreatic margin and 3 of 14 lymph nodes involved by carcinoma. She underwent 6 months of adjuvant FOLFIRINOX followed by adjuvant chemoradiation therapy to a dose of 45-to-50 Gy in 25 fractions using simultaneous integrated boost technique with concurrent capecitabine. Images show standard adjuvant radiation fields and dose distribution using IMRT in axial (A), coronal (B) and sagittal (C) planes. Scale: blue 17 Gy -> red 50 Gy.

Total neoadjuvant therapy

Patients undergoing upfront surgical resection plus adjuvant chemotherapy experience 5-year OS rates of 25–50% due to high rates of systemic relapse.7, 8 Based on this premise, neoadjuvant approaches have been tested in resectable PDAC with the following possible advantages: upfront treatment of occult micrometastases, avoiding unnecessary resection for rapidly progressing tumors, improved likelihood of margin negative resection, and improved chemotherapy delivery compared to post-resection adjuvant therapy. In the absence of clear guidelines, current clinical criteria for considering neoadjuvant therapy in resectable PDAC are large primary tumors, high CA 19–9 levels (>1000 U/ml), peri-pancreatic lymph node involvement or equivocal radiographic features suggestive of more advanced disease.183 A number of randomized clinical trials are underway to more comprehensively define the role of neoadjuvant compared to adjuvant therapies in resectable patients, with an ultimate goal to most effectively target micrometastatic disease.

Results from the PREOPANC-1 randomized phase III trial in patients with resectable and borderline resectable PDAC suggests patients had longer survival if gemcitabine–based therapy was given in the neoadjuvant versus the adjuvant setting. In the study, 246 patients were randomized to immediate surgery, then 6 cycles of adjuvant gemcitabine (arm A) versus preoperative chemoradiotherapy, consisting of 3 courses of gemcitabine (second combined with radiotherapy to 36 Gy in 15 fractions), followed by surgery and 4 cycles of adjuvant gemcitabine (arm B). Among patients who underwent resection, this study demonstrated an OS benefit from arm B at a median of 35.2 months vs 19.8 months for arm A (p=.029), although the intention-to-treat analysis revealed only a trend toward improved survival.184 These results are supported by a propensity score-matched analysis of adult patients with resected stage 1 or 2 PDAC from the National Cancer Database (2006–2012).185 Neoadjuvant therapy followed by resection had a significant survival benefit compared with upfront resection (median survival 26 months v 21 months, p<0.01), further supporting the consideration of offering neoadjuvant therapy to resectable patients. As in the adjuvant setting, the role of radiation in the neoadjuvant approach to resectable disease remains controversial in the absence of phase III trials directly comparing neoadjuvant treatment approaches with or without radiation. A randomized clinical trial (PREOPANC-2) investigating whether neoadjuvant chemotherapy with FOLFIRINOX improves survival compared to neoadjuvant gemcitabine-based chemoradiotherapy with borderline resectable PDAC is underway.

Using a different approach of neoadjuvant systemic therapy, a randomized prospective trial (SWOG-1505) was conducted evaluating perioperative mFOLFIRINOX vs gemcitabine/nab-paclitaxel (12 weeks pre-, and 12 weeks post-surgery in both arms) in patients with resectable PDAC.186 The trial has completed accrural, and results should be forthcoming in the near future. The added value of this study will be a direct comparison of the performance of these two regimens and perhaps some interpretable data about genomic signatures that may help predict therapeutic responses.

Borderline Resectable

Treatment paradigm

Patients with borderline resectable pancreatic cancer (BRPC) have no evidence of metastatic disease but are less likely to be resected with negative margins due to close proximity to or direct involvement of venous and/or arterial structures. The goal of the treatment approach is to maximize the chance at a margin-negative resection. Although no randomized studies provide specific guidance regarding the optimal treatment paradigm, several prospective phase II studies demonstrate the feasibility and efficacy of a total neoadjuvant approach consisting of neoadjuvant chemotherapy followed by radiation or chemoradiation. Patients who do not show distant progression or local invasion precluding surgery then undergo surgical exploration and resection.

The single arm Alliance A021101 trial enrolled 23 BRPC patients who underwent neoadjuvant mFOLFIRINOX and neoadjuvant CRT (50.4Gy in 28 fractions) prior to definitive surgical resection. Fifteen of 23 patients (68%) underwent resection with an impressive 93% R0 resection rate, and neoadjuvant treatment was not found to preclude resection.187 Investigators at Massachusetts General Hospital (MGH) conducted a phase II trial evaluating the R0 resection rates in patients receiving 8-cycles of FOLFIRINOX plus short or long-course neoadjuvant chemoradiation with capecitabine. R0 resection rates for patients who underwent resection was 97% with a median PFS of 48.6 months and 2-year OS of 72%; far surpassing previously published historical outcomes.188 Further, several retrospective studies have shown high R0 resection rates (88–96.7%) with low toxicity, and one study in particular showed improvement in local control and OS with integration of neoadjuvant chemoradiation compared to neoadjuvant chemotherapy alone.189, 190 In a modern cohort of BRPC patients at MD Anderson Cancer Center who underwent either neoadjuvant chemotherapy (n=31) or neoadjuvant chemoradiation (n=227), patients who received neoadjuvant chemoradiation had significantly improved R0 resection rates (91% vs 79%), histologically node positive resection rates (3% vs 23%), and locoregional recurrence (16% vs 33%) with a non-significant OS difference (33.6 vs 26.4 months).191 Although early data suggests the importance of integrating both neoadjuvant chemotherapy and chemoradiation into the treatment paradigm for PDAC, large prospective trial data is lacking.

Chemotherapy

The selection of chemotherapeutic agents in BRPC follows the same rationale used in the resectable and metastatic settings. Typically, patients receive ≥ 2–6 months of neoadjuvant chemotherapy before proceeding to radiation, chemoradiation, or directly to surgery, if there is no evidence of distant metastasis. If the full 6 months of chemotherapy are not delivered preoperatively, the balance is commonly offered after surgery. The goals of therapy are to improve resectability by down-staging the primary tumor, reduce micrometastasis, and avoid surgery in patients with aggressive metastatic biology. The only randomized data supporting this approach come from the PREOPANC-1 study, discussed above, which enrolled both resectable and borderline resectable patients and demonstrated improved OS and disease-free survival when gemcitabine and chemoradiation were delivered prior to surgery as compared with upfront resection.184 NCCN guidelines now recommend neoadjuvant chemotherapy for all patients with borderline resectable PDAC. The ongoing SWOG 1505 and ALLIANCE A021501 trials are evaluating 3–4 months of upfront chemotherapy, whereas a completed phase II study out of MGH utilized a total neoadjuvant approach in patients who did not progress after 4 months of systemic therapy.186, 188, 192 The overall treatment goals and approach are similar among most centers that employ neoadjuvant therapy for these patients, although, in the absence of level 1 data, there is little consensus on optimal timing and sequencing of treatment.

Radiation

The goal of neoadjuvant radiation or chemoradiation in BRPC is to increase the number of patients who can receive R0 resections and improve durable locoregional control. Most commonly, standard fractions of 1.8–2 Gy per day are delivered for 5–6 weeks to the tumor, tumor-vessel interface, and the regional lymph node basins. For patients with BRPC, targeted dose-escalation of regions of the tumor abutting the vessel has been shown to be feasible and safe193 and may improve surgical resection rates and survival.194, 195 Because there is a definable tumor volume, radiation therapy in the neoadjuvant setting may also allow for the safe delivery of hypofractionated regimens consisting of larger radiation doses per day in fewer treatments, a more convenient and less costly treatment consisting of 5 to 15 fractions. This approach was used in the aforementioned PREOPANC-1 study, which included patients with BRPC.184 The effectiveness of hypofractionation in PDAC has not been compared against conventional fractionation in a randomized fashion.

The utilization of stereotactic body radiation therapy (SBRT), in which a smaller tumor and tumor-vessel interface volume is treated in 5 or fewer fractions, is increasing nationally.196 Due to small margins and rapid dose fall off, SBRT requires added measures to ensure target delineation, motion management, and daily imaging to verify that the treatment can be delivered safely and effectively (Figure 6). Initial single institution studies of SBRT in locally advanced disease indicated survival that approximated standard of care treatment and very few grade >3 toxicities.197 Neoadjuvant SBRT was included as an arm in the ongoing prospective ALLIANCE trial A021501 that is evaluating neoadjuvant FOLFIRINOX for patients with BRPC.192 The American Society for Radiation Oncology (ASTRO) clinical practice guidelines provide a conditional recommendation to treat patients with borderline resectable pancreatic cancer with either conventional or hypofractionated radiotherapy as part of a neoadjuvant treatment regimen including chemotherapy, generally limiting the use of SBRT to patients with smaller tumors located ≥ 1 cm from a gastrointestinal mucosal margin and with no evidence of nodal involvement.198

Figure 6.

Figure 6.

48 year-old man with a localized PDAC of the uncinate process encasing the SMA. He underwent 3 cycles of gemcitabine/abraxane, then SBRT guided by fiducial markers to 33 Gy in 5 fractions. He had 2 more cycles of chemotherapy, then underwent exploration revealing retraction from SMA and subsequent Whipple procedure. Images show (A) SBRT tumor volume (red) and duodenal planning organ at risk volume (PRV, purple). Dose distribution in axial (B) and coronal (C) planes. Scale: blue 11 Gy -> red 33 Gy.

Surgery

The challenge to achieve a margin negative resection at pancreatectomy in increased when the tumor is adherent to or invading critical blood vessels. The addition of vascular resection and reconstruction adds to the technical complexity of the procedure. The most critical aspects of vascular resection and reconstruction in this setting are having high quality pre-operative imaging to develop an operative game plan, and to have a surgeon or surgical team experienced in both pancreatic and vascular surgery. Given the increased risk of perioperative complications and the complex nature of these operations, they should only be performed in high volume pancreatic centers and by surgeons with the appropriate level of expertise to deal with the added complexity and morbidity that may accompany these types of cases. The relative value of SMV/PV resection and /or reconstruction has been clearly shown. Venous resection and reconstruction due to increased surgical experience is now safe and standardly performed in high volume centers. In 2 meta-analyses comparing survival for patients who underwent pancreaticoduodenectomy with or without vein resection, no differences in survival were observed.199, 200 Further, across a number of studies, patients who undergo pancreatectomy with vein resection have increased survival compared to those who do not undergo resection or have a R2 resection performed. Generally, margin status is a predictor of survival.201

Locally advanced

Treatment paradigm

Locally advanced pancreatic cancer (LAPC) accounts for 30% of newly diagnosed cases and is considered surgically unresectable due to local involvement of adjacent critical blood vessels.202 Current guidelines for LAPC recommend enrollment in clinical trials where available, or, in the absence of clinical trials, nonoperative treatment through a multidisciplinary approach. At present, because this disease is generally considered incurable, standard of care is very similar to metastatic patients, based around at least 6 months of chemotherapy.203 At this time there are no randomized data supporting the inclusion of local therapy in these patients, but more studies are re-evaluating this question for patients who complete chemotherapy with no evidence of distant metastatic disease. We will review these approaches below.

Chemotherapy

Chemotherapy forms the backbone of the therapeutic approach to LAPC, as many patients will never convert to resectability, and the risk of distant progression is very high in this population. The goals of therapy are to control disease progression, reduce symptoms, and maintain quality of life. For a subset of patients, chemotherapy may assist in shrinking the local tumor and converting unresectable to resectable disease. First-line therapy, like in the metastatic and resectable settings, has shifted toward the use of FOLFIRINOX or gemcitabine + nab-paclitaxel, despite the absence of randomized data in the locally advanced setting. The recommendation to utilize these regimens are based on retrospective data or extrapolated from the MPACT and PRODIGE4/ACCORD11 studies discussed above. A recent meta-analysis suggested a potential survival benefit of FOLFIRINOX in patients with LAPC.204 Furthermore, results from the phase II SCALOP study and phase III LAP-07 studies, each of which investigated single agent chemotherapy, report progression in the majority of patients, reinforcing the importance of more aggressive combination chemotherapy.205, 206 All patients with LAPC should therefore be strongly considered for upfront combination chemotherapy unless precluded by performance status or toxicity. In these cases, dose adjustments or less toxic chemotherapy regimens may be considered.

Radiation

Although LAPC represents an ideal opportunity to achieve local tumor control using radiotherapy, its role in unresectable pancreatic cancer is controversial, as historically, technological limitations and organ tolerance prevented ablative dose delivery to a central abdominal organ. The LAP-07 trial randomized 450 patients initially to gemcitabine alone vs gemcitabine + erlotinib followed by randomization, if no progression, to the same chemotherapy or chemoradiotherapy. Chemoradiotherapy was delivered to 54 Gy in 30 fractions to the pancreas and peripancreatic nodal basins with concurrent capecitabine using basic 3D-conformal radiotherapy, and revealed no improvement in OS, despite decreased rates of local progression (32% vs 46%), thereby removing radiotherapy from the treatment algorithm.206 These results should perhaps not be surprising, as the delivered dose was inadequate for tumor ablation. Despite the equivocal results of LAP-07, current ASCO guidelines include a strong recommendation to follow chemotherapy with localized chemoradiation or SBRT in patients who have stable disease after 6 months of chemotherapy or those who cannot tolerate further chemotherapy due to toxicities.203

Multiple technological advancements, including 4D-motion management, improved image guidance, and the implementation of intensity modulated radiotherapy (IMRT)and proton therapy, have allowed for minimizing unwanted dose in normal tissues (i.e. liver, small bowel, stomach). Early studies of ablative (biologically effective dose [BED] > 100) SBRT had excellent tumor control but evidence of increased acute and late GI toxicities.207209 Using either high dose conventional fractionation or a hypofractionated approach (15 fractions) to a BED = 77.2–97.9 Gy, there is published 3 and 5-year OS of 35% and 18%, far exceeding historical controls.194, 209 Further, utilizing MRI-guided radiotherapy, treating to a BED > 70 Gy was found to significant improve OS in a small cohort (median OS = 8.8 months in conventional dose group versus not reached in high dose cohort).210 Fractionation should be selected based on the safe delivery of an ablative dose to maximal tumor volume; tumors that closely appose the bowel may require increased fractionation, whereas those with more clearance may be amenable to shorter courses. Ablative doses of 75 Gy in 25 fractions, 67.5 Gy in 15 fractions, and 50 Gy in 5 fractions each allow for the delivery of a BED of ~100 Gy and show evidence of efficacy in appropriately selected populations.209, 211 (Figure 8) Ongoing prospective trials, such as panCRS (ClinicalTrials.gov Identifier: NCT01926197) and a phase I/II dose escalation trial for pancreas SBRT with the radioprotector GC4419 (ClinicalTrials.gov Identifier: NCT03340974) are further evaluating the efficacy and safety of pancreas SBRT in patients with locally advanced disease. Importantly, these approaches should only be utilized at centers with technology and experience delivering ablative therapy under image guidance.

Figure 8.

Figure 8.

63 year-old woman with locally advanced PDAC arising from the head of pancreas. After 6 months of mFOLFIRINOX, tumor continued to encase SMA and celiac artery. She was treated with hypofractionated, dose-escalated radiotherapy to an ablative dose of 67.5 Gy delivered in 15 daily fractions during breath hold. A. Target delineation shows integrated tumor volume (red), duodenum (blue), and stomach (pink). A 10mm planning organ at risk volume (PRV) around luminal bowel structures shown in cyan. Planning treatment volumes for 67.5 Gy (magenta) excluded from bowel PRV, whereas low dose 37.5 Gy PTV (green) overlaps both PRV and true bowel volume. B. Dose distribution for plan based on contours shown in (A). Scale: blue 20 Gy -> red 65 Gy.

Alternatively, radiation may play an important role in converting unresectable patients into surgical candidates, as with BRPC. In a recent phase II single-arm study out of Massachusetts General Hospital, optimizing neoadjuvant treatment response with neoadjuvant FOLFIRINOX, losartan, and chemoradiotherapy (conventional fractionation with 5FU or capecitabine for the majority of patients), resulted in an unprecedented 69% R0 resection rate and median OS = 33 months in those who underwent resection (n = 42).141 A key determinant in the selection of radiation approaches will be the likelihood for resectability, as those patients with persistent extensive vascular involvement after neoadjuvant chemotherapy may be more likely to benefit from an ablative rather than neoadjuvant dose and fractionation. In the absence of high quality evidence supporting the use of radiation to downstage or definitively treat locally advanced disease, current ASTRO clinical guidelines provide conditional recommendation for each of these practices.198

Surgery

As surgical outcomes of pancreatectomy performed in combination with vascular reconstruction have improved, and as neoadjuvant combination regimens have become more effective, there has been an increased interest in defining patients with LAPC who may benefit from a more aggressive surgical approach. Following neoadjuvant therapy, it may be difficult to accurately assess treatment response, with studies showing resection rates ranging from 12–60%.212214 While some of these patients (about 10–25%) may demonstrate downstaging to a borderline resectable or resectable category following neoadjuvant treatment, it has been increasingly appreciated that imaging may not be reflective of surgical candidacy due to the presence of fibrotic changes that persist after effective treatment. As such, there has been considerable development and use of more advanced surgical approaches to increase the number of patients with LAPC who may be considered surgical candidates. In general, these operations are longer in duration, and are associated with increased blood loss and higher morbidity and mortality, in particular when resection of the celiac artery or superior mesenteric artery, either alone or in combination with venous reconstruction, is performed.215, 216 (Figure 7) In several recent single center series, it has been shown that some of these patients can successfully undergo R0 resection, and patients in this subgroup may experience improved survival compared to non-surgical treatment.141, 212, 214

Figure 7.

Figure 7.

Examples of borderline resectable and unresectable PDAC. A. CT scan image (3D vascular reconstruction) of a borderline resectable PDAC with narrowing of the superior mesenteric vein (SMV, bright yellow arrows). Proximal and distal targets for reconstruction shown at orange arrows. B. Following neoadjuvant chemotherapy, the patient was taken to the operating room and the tumor was excised to include the involved SMV (left panel). The SMV was reconstructed using an interposition internal jugular vein graft (B, right panel, and C). D. Cross-sectional CT images of a locally advanced PDAC (middle panel, orange arrow). There is a proximal portal vein target (left panel, orange arrow), but only collaterals distally (right panel, blue arrows). With no distal target, reconstruction is not possible, making the tumor unresectable.

The definition of resectability for LAPC varies among surgeons and at different centers, depending on the level of expertise and willingness to undertake these complex procedures. Given the morbidity and mortality associated with pancreaticoduodenectomy and complex vascular reconstruction, careful patient selection is warranted, and these cases should be done in the setting of significant clinical expertise. In the absence of clear guidelines, administration of 4–6 months of neoadjuvant therapy, and documentation of absence of disease progression, stable or improving Ca 19–9, and good performance status (ECOG ≤1) should occur before considering surgical resection.

Post-treatment Surveillance

Per NCCN guidelines, after completing both local and systemic therapy patients are followed up every 3–6 months for the first two years, then every 6–12 months thereafter. Patients should be evaluated with a history and physical examination focused on symptoms, specifically weight loss, anorexia, fatigue, and pain. Labs may be ordered as clinically indicated, although most physicians will follow complete blood count, blood chemistries, and liver function tests as these can indicate long term sequelae of therapy or new metastatic disease. Imaging surveillance includes chest CT and CT or MRI of the abdomen and pelvis with contrast to identify recurrence or metastasis. As described above, in appropriate patients, CA19–9 can be used to track therapy response and recurrence, although this is considered a category 2B recommendation.83 Patients exhibiting recurrence with good performance status should be considered for clinical trials, if available, or the next appropriate line of systemic therapy. If not already engaged, supportive (or palliative) care should be introduced at this time, given the poor prognosis and high disease- and treatment-related morbidities associated with recurrence.

Survivorship and Patient Resilience

Although cure remains the top priority for research and practice, the high mortality of PDAC calls for increased use of supportive care in the management of this disease. As one example, venous thromboembolism (VTE), including deep vein thrombosis and pulmonary embolism, is highly prevalent in patients with PDAC and contributes to both morbidity and mortality.217, 218 Two large randomized studies, the FRAGEM trial and CONKO-004, randomized unselected patients with unresectable or metastatic PDAC to chemotherapy with or without the addition of a low molecular weight heparin anticoagulant at higher-than-standard prophylactic dosing--FRAGEM used dalteparin 200 IU/kg daily for 4 weeks before reducing to 150 IU/kg daily for a further 8 weeks, whereas CONKO-004 used enoxaparin at a dose of 1 mg/kg daily. Both studies reported concomittant reductions in VTE risk and mortality without a significant increase in bleeding events.219, 220 ASCO guidelines recommend VTE prophylaxis for with low molecular weight heparin or, based on recent reports demonstrating effectiveness and safety with factor Xa inhibitors,221, 222 apixaban or rivaroxaban for patients with a Khorana score ≥ 2, which includes all patients with PDAC.223

Regardless of cancer stage and patient prognosis, early introduction to expert supportive care improves the social, psychological, and physical wellbeing of patients, decreases the intensity of medical intervention at the end of life, and ultimately improves survival224226. Indeed, while systemic therapies have a modest impact on final survival outcomes, modern chemotherapy combinations do significantly improve symptoms such as pain, sleep disturbance, appetite, gastrointestinal distress, and emotional functioning94, 227. Collectively, the many dimensions of the patient experience and perception of health during PDAC treatment is captured in patient-related outcome measures (PROM) within the broader concept of Health-Related Quality of Life (HRQoL), and measurement of this index is an important part of research and clinical care in this patient population228230.

As described in previous sections, the initial presenting symptoms of PDAC are vague and inconsistent231. Some symptoms are primarily due to local invasion and distortion of normal anatomy (e.g. abdominal pain, jaundice), but many are due to a conserved systemic response to illness. This sickness response consists of a number of stereotypical behaviors and metabolic adaptations that serve to protect the host from acute survival challenges such as infection and trauma232, 233. Lethargy, anorexia, fever, and catabolism of muscle are beneficial to the host in the short term, but in the setting of chronic disease become maladaptive. In particular, a constellation of weight loss, muscle wasting, adipose tissue reprogramming, and anorexia comprise a debilitating condition known as cachexia that is found in ~80% of PDAC patients234. Though tumor burden alone can be debilitating, even small PDAC tumors are capable of inducing cachexia, and up to 30% of cancer deaths are attributable to this condition alone235. Unfortunately, there are currently no effective therapies proven to prevent or treat these problems in PDAC patients, although recent preclinical data and new clinical trials are providing some hope236238. The current standard therapeutic for cachexia, megestrol acetate, does produce modest improvements in appetite and quality of life, but has no impact on functionality or survival239. Amongst the other therapeutic options, clinical trials provide strong support for exercise programs both prior to and during cancer therapy (see240 for recent extensive review), although direct evidence of benefit in PDAC patients is currently lacking. Finally, comprehensive prehabilitation programs aimed at improving patient resiliency prior to treatment intervention (e.g. surgical resection) show promise in other cancer cohorts, but this modality also remains unproven in the PDAC patient population241.

Nutritional compromise in PDAC is common but may be driven by a variety of underlying pathologies. Nausea due to both the primary disease process and its associated chemotherapy is most effectively treated with serotonin type 3 receptor antagonists and atypical antipsychotics (e.g. olanzapine), with some emerging evidence suggesting efficacy with cannabinoids as well242, 243. Even in the absence of overt nausea, patients frequently report loss of appetite and this symptom is driven by central pathways that are largely distinct from those that produce nausea244, 245. Nutrition status also declines due to malabsorption which is most often secondary to pancreatic exocrine deficiency. Pancreatic enzyme replacement therapy helps to stabilize weight loss, and also improves quality of life by decreasing gastrointestinal symptoms246248. Malabsorption also occurs due to biliary obstruction, a complication found in up to 90% of PDAC patients249, 250. Similar to replacement of pancreatic enzymes, treatment of biliary obstruction improves symptoms beyond its effects on digestion, including anorexia, pruritus, and fatigue251, 252. Collectively, careful attention to nutritional status of patients with PDAC improves both their survival and quality of life, and early involvement of nutrition experts in their care is recommended247, 253.

Fatigue is a common symptom in cancer patients and numerous etiologies are proposed (for a recent review, see254). However, specific data in the PDAC population is rare, with most studies reporting high levels of fatigue both at presentation and throughout treatment255. A variety of interventions for cancer-related fatigue have been proposed, ranging from counseling-based therapy to pharmacotherapy (e.g. corticosteroids, methylphenidate), but none have produced definitive benefit in this patient population. Some evidence suggests that mindfulness techniques and exercise may benefit patients, but available data indicate that benefits are modest and inconsistent256.

Although the association of mood disorders, fatigue, and cognitive decline with PDAC is extensively documented, it was often assumed that these were secondary to both the psychological impact of the diagnosis itself, and to the overall toxicity of PDAC treatment. However, it is now apparent that these are often presenting complaints with this diagnosis, demonstrating that the cancer has independent detrimental effects on the brain257259. Furthermore, these symptoms are collectively the most significant drivers of declines in HRQoL, and are independently predictive of survival in PDAC patients260, 261. Although the true prevalence of mood disorders in PDAC patients is controversial, it is likely far higher than in the healthy population with rates ranging from a third to a half of patients being reported in some studies262, 263. Clinicians should be sensitive to these comorbidities and routinely implement screening for and, if present, treatment for depression.264

Emerging technologies such as electronic patient-reported outcomes hold promise as more efficient and standardized ways to assess HRQoL and deliver supportive care. Future clinical care and research will also benefit from the advent of modern methods for recording patient reported symptoms in real time, particularly when combined with more objective measures of daily activity (e.g. actigraphy devices). Several studies demonstrate efficacy of these electronic methods to improve patient symptoms, reduce health care costs, and improve OS in other cancer types, suggesting that these technologies will also benefit the PDAC patient population265267.

Conclusions

As we enter the 3rd decade of the 21st century, the story of PDAC is one of two competing narratives. On the one hand, progress in PDAC outcomes continues to lag far behind the survival gains made in other solid tumors. Despite being relatively uncommon, PDAC is expected to become the second leading cause of cancer death by the end of the decade.268 The vast majority of patients diagnosed with PDAC in 2020 will succumb to the disease. On the other hand, 5-year survival among all patients has eclipsed double digits for the first time. Led by improvements in the effectiveness of systemic therapy, an increase in the proportion of patients with early stage disease, and stage-specific treatment paradigms, a true separation in expected survival is widening between those with resectable cancer and patients with locally advanced or metastatic disease. Moving forward, efforts are focusing in on surveillance approaches and imaging innovations to improve early detection of PDAC, thereby increasing the proportion of patients diagnosed with curable localized disease. Simultaneously, advances in systemic therapy led by the implementation of precision oncology and an increasing focus on quality of life outcomes promise to improve both lifespan and healthspan in patients with both localized and metastatic disease. Together, fueled by these innovations, we may be on the cusp on meaningfully changing outcomes in PDAC.

Acknowledgements:

The authors would like to thank Dr. Theodore Braun for his assistance with proofreading and editing the final manuscript.

Funding: Aaron J. Grossberg receives research funding support from the National Cancer Institute (K08 CA245188), Brenden-Colson Center for Pancreatic Care and the Cancer Early Detection Advanced Research Center at OHSU. Linda C. Chu receives research funding support from The Lustgarten Foundation and The Emerson Collective. Elliot K. Fishman receives research funding support from The Lustgarten Foundation. Anirban Maitra receives funding from the National Cancer Institute (U01 CA196403, U01 CA200468, P50 CA221707). Daniel L. Marks receives funding from the National Cancer Institute (R01 CA217989, R01 CA234006) and from the Brenden-Colson Center for Pancreatic Care at OHSU. Diane M. Simeone receives research funding from the National Cancer Institute (R01 CA131045, R01 CA174836), Tyme, Repare Therapeutics, and Tempus. Charles R. Thomas, Jr. receives funding support from the American Medical Association.

Footnotes

Conflicts of Interest: Anirban Maitra receives royalties for a pancreatic cancer biomarker test from Cosmos Wisdom Biotechnology, and this financial relationship is managed and monitored by the UTMDACC Conflict of Interest Committee. In addition, technology he was involved in creating while a faculty at Johns Hopkins University has been licensed to Thrive Earlier Detection. Daniel L. Marks is a paid consultant for Pfizer, Inc. and this potential conflict of interest is managed by the integrity office at OHSU. Diane M. Simeone serves on the advisory board of Interpace and Tyme and has been a paid consultant for Merck. Charles R. Thomas, Jr. is deputy editor for JAMA Oncology. Aaron J. Grossberg, Christopher R. Deig, William L. Hwang, Arnav Mehta, Nima Nabavizadeh, and Colin D. Weekes declare no conflicts.

References

  • 1.Howlader N, Noone A, Krapcho M, et al. SEER Cancer Statistics Review, 1975–2017. Available from URL: https://seer.cancer.gov/csr/1975_2017/ based on November 2019 SEER data submission, posted to the SEER web site, April, 2020].
  • 2.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70: 7–30. [DOI] [PubMed] [Google Scholar]
  • 3.SEER Cancer Stat Facts: Pancreatic Cancer. Available from URL: https://seer.cancer.gov/statfacts/html/pancreas.html [accessed May 4, 2020.
  • 4.Islami F, Miller KD, Siegel RL, et al. National and State Estimates of Lost Earnings From Cancer Deaths in the United States. JAMA Oncol. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Cives M, Strosberg JR. Gastroenteropancreatic Neuroendocrine Tumors. CA: A Cancer Journal for Clinicians. 2018;68: 471–487. [DOI] [PubMed] [Google Scholar]
  • 6.Li D, Xie K, Wolff R, Abbruzzese JL. Pancreatic cancer. Lancet. 2004;363: 1049–1057. [DOI] [PubMed] [Google Scholar]
  • 7.Conroy T, Hammel P, Hebbar M, et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. N Engl J Med. 2018;379: 2395–2406. [DOI] [PubMed] [Google Scholar]
  • 8.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: 1200–1210. [DOI] [PubMed] [Google Scholar]
  • 9.Laviano A, Meguid MM, Inui A, Muscaritoli M, Rossi-Fanelli F. Therapy insight: Cancer anorexia-cachexia syndrome--when all you can eat is yourself. Nat Clin Pract Oncol. 2005;2: 158–165. [DOI] [PubMed] [Google Scholar]
  • 10.Danai LV, Babic A, Rosenthal MH, et al. Altered exocrine function can drive adipose wasting in early pancreatic cancer. Nature. 2018;558: 600–604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Andersen DK, Korc M, Petersen GM, et al. Diabetes, Pancreatogenic Diabetes, and Pancreatic Cancer. Diabetes. 2017;66: 1103–1110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Hendifar A, Osipov A, Khanuja J, et al. Influence of Body Mass Index and Albumin on Perioperative Morbidity and Clinical Outcomes in Resected Pancreatic Adenocarcinoma. PLoS One. 2016;11: e0152172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Bachmann J, Heiligensetzer M, Krakowski-Roosen H, Buchler MW, Friess H, Martignoni ME. Cachexia worsens prognosis in patients with resectable pancreatic cancer. J Gastrointest Surg. 2008;12: 1193–1201. [DOI] [PubMed] [Google Scholar]
  • 14.Kays JK, Shahda S, Stanley M, et al. Three cachexia phenotypes and the impact of fat-only loss on survival in FOLFIRINOX therapy for pancreatic cancer. J Cachexia Sarcopenia Muscle. 2018;9: 673–684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Babic A, Rosenthal MH, Bamlet WR, et al. Postdiagnosis Loss of Skeletal Muscle, but Not Adipose Tissue, Is Associated with Shorter Survival of Patients with Advanced Pancreatic Cancer. Cancer Epidemiol Biomarkers Prev. 2019;28: 2062–2069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.He J, Blair AB, Groot VP, et al. Is a Pathological Complete Response Following Neoadjuvant Chemoradiation Associated With Prolonged Survival in Patients With Pancreatic Cancer? Ann Surg. 2018;268: 1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Goggins M, Overbeek KA, Brand R, et al. Management of patients with increased risk for familial pancreatic cancer: updated recommendations from the International Cancer of the Pancreas Screening (CAPS) Consortium. Gut. 2020;69: 7–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Ishikawa O, Ohigashi H, Imaoka S, et al. Minute carcinoma of the pancreas measuring 1 cm or less in diameter--collective review of Japanese case reports. Hepatogastroenterology. 1999;46: 8–15. [PubMed] [Google Scholar]
  • 19.Tummers WS, Groen JV, Sibinga Mulder BG, et al. Impact of resection margin status on recurrence and survival in pancreatic cancer surgery. Br J Surg. 2019;106: 1055–1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Winter JM, Cameron JL, Campbell KA, et al. 1423 pancreaticoduodenectomies for pancreatic cancer: A single-institution experience. J Gastrointest Surg. 2006;10: 1199–1210; discussion 1210–1191. [DOI] [PubMed] [Google Scholar]
  • 21.Yamaguchi K, Enjoji M, Tsuneyoshi M. Pancreatoduodenal carcinoma: a clinicopathologic study of 304 patients and immunohistochemical observation for CEA and CA19–9. J Surg Oncol. 1991;47: 148–154. [DOI] [PubMed] [Google Scholar]
  • 22.Raptis DA, Fessas C, Belasyse-Smith P, Kurzawinski TR. Clinical presentation and waiting time targets do not affect prognosis in patients with pancreatic cancer. Surgeon. 2010;8: 239–246. [DOI] [PubMed] [Google Scholar]
  • 23.Porta M, Fabregat X, Malats N, et al. Exocrine pancreatic cancer: symptoms at presentation and their relation to tumour site and stage. Clin Transl Oncol. 2005;7: 189–197. [DOI] [PubMed] [Google Scholar]
  • 24.Chari ST, Leibson CL, Rabe KG, Ransom J, de Andrade M, Petersen GM. Probability of pancreatic cancer following diabetes: a population-based study. Gastroenterology. 2005;129: 504–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Whitcomb DC, Pogue-Geile K. Pancreatitis as a risk for pancreatic cancer. Gastroenterol Clin North Am. 2002;31: 663–678. [DOI] [PubMed] [Google Scholar]
  • 26.Sah RP, Sharma A, Nagpal S, et al. Phases of Metabolic and Soft Tissue Changes in Months Preceding a Diagnosis of Pancreatic Ductal Adenocarcinoma. Gastroenterology. 2019;156: 1742–1752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Mayers JR, Wu C, Clish CB, et al. Elevation of circulating branched-chain amino acids is an early event in human pancreatic adenocarcinoma development. Nat Med. 2014;20: 1193–1198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Canto MI, Almario JA, Schulick RD, et al. Risk of Neoplastic Progression in Individuals at High Risk for Pancreatic Cancer Undergoing Long-term Surveillance. Gastroenterology. 2018;155: 740–751 e742. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.US Preventive Services Task Force, Owens DK, Davidson KW, et al. Screening for Pancreatic Cancer: US Preventive Services Task Force Reaffirmation Recommendation Statement. JAMA. 2019;322: 438–444. [DOI] [PubMed] [Google Scholar]
  • 30.SEER Cancer Stat Facts: Colorectal Cancer. Available from URL: https://seer.cancer.gov/statfacts/html/colorect.html [accessed May 4, 2020.
  • 31.SEER Cancer Stat Facts: Female Breast Cancer. Available from URL: https://seer.cancer.gov/statfacts/html/breast.html [accessed May 4, 2020.
  • 32.Force UPST. Screening for Colorectal Cancer: US Preventive Services Task Force Recommendation Statement. JAMA. 2016;315: 2564–2575. [DOI] [PubMed] [Google Scholar]
  • 33.Siu AL. Screening for Breast Cancer: U.S. Preventive Services Task Force Recommendation Statement. Ann Intern Med. 2016;164: 279–296. [DOI] [PubMed] [Google Scholar]
  • 34.Chang CY, Huang SP, Chiu HM, Lee YC, Chen MF, Lin JT. Low efficacy of serum levels of CA 19–9 in prediction of malignant diseases in asymptomatic population in Taiwan. Hepatogastroenterology. 2006;53: 1–4. [PubMed] [Google Scholar]
  • 35.Kim JE, Lee KT, Lee JK, Paik SW, Rhee JC, Choi KW. Clinical usefulness of carbohydrate antigen 19–9 as a screening test for pancreatic cancer in an asymptomatic population. J Gastroenterol Hepatol. 2004;19: 182–186. [DOI] [PubMed] [Google Scholar]
  • 36.Yurgelun MB, Chittenden AB, Morales-Oyarvide V, et al. Germline cancer susceptibility gene variants, somatic second hits, and survival outcomes in patients with resected pancreatic cancer. Genet Med. 2019;21: 213–223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hu C, Hart SN, Polley EC, et al. Association Between Inherited Germline Mutations in Cancer Predisposition Genes and Risk of Pancreatic Cancer. JAMA. 2018;319: 2401–2409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Tempero MA. NCCN Guidelines Updates: Pancreatic Cancer. J Natl Compr Canc Netw. 2019;17: 603–605. [DOI] [PubMed] [Google Scholar]
  • 39.Stoffel EM, McKernin SE, Brand R, et al. Evaluating Susceptibility to Pancreatic Cancer: ASCO Provisional Clinical Opinion. Journal of Clinical Oncology. 2019;37: 153–164. [DOI] [PubMed] [Google Scholar]
  • 40.Golan T, Hammel P, Reni M, et al. Maintenance Olaparib for Germline BRCA-Mutated Metastatic Pancreatic Cancer. N Engl J Med. 2019;381: 317–327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Stoll J, Kupfer SS. Risk Assessment and Genetic Testing for Inherited Gastrointestinal Syndromes. Gastroenterol Hepatol (N Y). 2019;15: 462–470. [PMC free article] [PubMed] [Google Scholar]
  • 42.Vasen H, Ibrahim I, Ponce CG, et al. Benefit of Surveillance for Pancreatic Cancer in High-Risk Individuals: Outcome of Long-Term Prospective Follow-Up Studies From Three European Expert Centers. Journal of Clinical Oncology. 2016;34: 2010–2019. [DOI] [PubMed] [Google Scholar]
  • 43.Chen JC, Beal EW, Pawlik TM, Cloyd J, Dillhoff ME. Molecular Diagnosis of Cystic Neoplasms of the Pancreas: a Review. J Gastrointest Surg. 2020. [DOI] [PubMed] [Google Scholar]
  • 44.Matthaei H, Schulick RD, Hruban RH, Maitra A. Cystic precursors to invasive pancreatic cancer. Nat Rev Gastroenterol Hepatol. 2011;8: 141–150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Basturk O, Hong SM, Wood LD, et al. A Revised Classification System and Recommendations From the Baltimore Consensus Meeting for Neoplastic Precursor Lesions in the Pancreas. Am J Surg Pathol. 2015;39: 1730–1741. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Singhi AD, Koay EJ, Chari ST, Maitra A. Early Detection of Pancreatic Cancer: Opportunities and Challenges. Gastroenterology. 2019;156: 2024–2040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Vege SS, Ziring B, Jain R, et al. American Gastroenterological Association Institute Guideline on the Diagnosis and Management of Asymptomatic Neoplastic Pancreatic Cysts. Gastroenterology. 2015;148: 819–822. [DOI] [PubMed] [Google Scholar]
  • 48.van Huijgevoort NCM, Del Chiaro M, Wolfgang CL, van Hooft JE, Besselink MG. Diagnosis and management of pancreatic cystic neoplasms: current evidence and guidelines. Nat Rev Gastroenterol Hepatol. 2019;16: 676–689. [DOI] [PubMed] [Google Scholar]
  • 49.Singhi AD, McGrath K, Brand RE, et al. Preoperative next-generation sequencing of pancreatic cyst fluid is highly accurate in cyst classification and detection of advanced neoplasia. Gut. 2018;67: 2131–2141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Springer S, Wang Y, Dal Molin M, et al. A combination of molecular markers and clinical features improve the classification of pancreatic cysts. Gastroenterology. 2015;149: 1501–1510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Springer S, Masica DL, Dal Molin M, et al. A multimodality test to guide the management of patients with a pancreatic cyst. Sci Transl Med. 2019;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Lowenfels AB, Maisonneuve P, Cavallini G, et al. Pancreatitis and the risk of pancreatic cancer. International Pancreatitis Study Group. N Engl J Med. 1993;328: 1433–1437. [DOI] [PubMed] [Google Scholar]
  • 53.Yadav D, Lowenfels AB. The epidemiology of pancreatitis and pancreatic cancer. Gastroenterology. 2013;144: 1252–1261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Whitcomb DC. Genetic aspects of pancreatitis. Annu Rev Med. 2010;61: 413–424. [DOI] [PubMed] [Google Scholar]
  • 55.Lowenfels AB, Maisonneuve P, Whitcomb DC. Risk factors for cancer in hereditary pancreatitis. International Hereditary Pancreatitis Study Group. Med Clin North Am. 2000;84: 565–573. [DOI] [PubMed] [Google Scholar]
  • 56.Naudin S, Viallon V, Hashim D, et al. Healthy lifestyle and the risk of pancreatic cancer in the EPIC study. Eur J Epidemiol. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wolpin BM, Kraft P, Gross M, et al. Pancreatic cancer risk and ABO blood group alleles: results from the pancreatic cancer cohort consortium. Cancer Res. 2010;70: 1015–1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Wolpin BM, Rizzato C, Kraft P, et al. Genome-wide association study identifies multiple susceptibility loci for pancreatic cancer. Nat Genet. 2014;46: 994–1000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Lambert SA, Abraham G, Inouye M. Towards clinical utility of polygenic risk scores. Hum Mol Genet. 2019;28: R133–R142. [DOI] [PubMed] [Google Scholar]
  • 60.Sah RP, Nagpal SJ, Mukhopadhyay D, Chari ST. New insights into pancreatic cancer-induced paraneoplastic diabetes. Nat Rev Gastroenterol Hepatol. 2013;10: 423–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Schultz NA, Dehlendorff C, Jensen BV, et al. MicroRNA biomarkers in whole blood for detection of pancreatic cancer. JAMA. 2014;311: 392–404. [DOI] [PubMed] [Google Scholar]
  • 62.Li A, Yu J, Kim H, et al. MicroRNA array analysis finds elevated serum miR-1290 accurately distinguishes patients with low-stage pancreatic cancer from healthy and disease controls. Clin Cancer Res. 2013;19: 3600–3610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kim J, Bamlet WR, Oberg AL, et al. Detection of early pancreatic ductal adenocarcinoma with thrombospondin-2 and CA19–9 blood markers. Science Translational Medicine. 2017;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Mellby LD, Nyberg AP, Johansen JS, et al. Serum Biomarker Signature-Based Liquid Biopsy for Diagnosis of Early-Stage Pancreatic Cancer. J Clin Oncol. 2018;36: 2887–2894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Riquelme E, Zhang Y, Zhang L, et al. Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes. Cell. 2019;178: 795–806.e712. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Aykut B, Pushalkar S, Chen R, et al. The fungal mycobiome promotes pancreatic oncogenesis via activation of MBL. Nature. 2019;574: 264–267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Goonetilleke KS, Siriwardena AK. Systematic review of carbohydrate antigen (CA 19–9) as a biochemical marker in the diagnosis of pancreatic cancer. Eur J Surg Oncol. 2007;33: 266–270. [DOI] [PubMed] [Google Scholar]
  • 68.Steinberg W The clinical utility of the CA 19–9 tumor-associated antigen. Am J Gastroenterol. 1990;85: 350–355. [PubMed] [Google Scholar]
  • 69.Kaur S, Smith LM, Patel A, et al. A Combination of MUC5AC and CA19–9 Improves the Diagnosis of Pancreatic Cancer: A Multicenter Study. Am J Gastroenterol. 2017;112: 172–183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.DiMagno EP, Reber HA, Tempero MA. AGA technical review on the epidemiology, diagnosis, and treatment of pancreatic ductal adenocarcinoma. American Gastroenterological Association. Gastroenterology. 1999;117: 1464–1484. [DOI] [PubMed] [Google Scholar]
  • 71.Hartwig W, Strobel O, Hinz U, et al. CA19–9 in potentially resectable pancreatic cancer: perspective to adjust surgical and perioperative therapy. Ann Surg Oncol. 2013;20: 2188–2196. [DOI] [PubMed] [Google Scholar]
  • 72.Maithel SK, Maloney S, Winston C, et al. Preoperative CA 19–9 and the yield of staging laparoscopy in patients with radiographically resectable pancreatic adenocarcinoma. Ann Surg Oncol. 2008;15: 3512–3520. [DOI] [PubMed] [Google Scholar]
  • 73.Khorana AA, Mangu PB, Berlin J, et al. Potentially Curable Pancreatic Cancer: American Society of Clinical Oncology Clinical Practice Guideline. Journal of Clinical Oncology. 2016;34: 2541–2556. [DOI] [PubMed] [Google Scholar]
  • 74.Marrelli D, Caruso S, Pedrazzani C, et al. CA19–9 serum levels in obstructive jaundice: clinical value in benign and malignant conditions. Am J Surg. 2009;198: 333–339. [DOI] [PubMed] [Google Scholar]
  • 75.Berger AC, Garcia M Jr., Hoffman JP, et al. Postresection CA 19–9 predicts overall survival in patients with pancreatic cancer treated with adjuvant chemoradiation: a prospective validation by RTOG 9704. J Clin Oncol. 2008;26: 5918–5922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Abdel-Misih SR, Hatzaras I, Schmidt C, et al. Failure of normalization of CA19–9 following resection for pancreatic cancer is tantamount to metastatic disease. Ann Surg Oncol. 2011;18: 1116–1121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Kondo N, Murakami Y, Uemura K, et al. Prognostic impact of perioperative serum CA 19–9 levels in patients with resectable pancreatic cancer. Ann Surg Oncol. 2010;17: 2321–2329. [DOI] [PubMed] [Google Scholar]
  • 78.Ishii H, Okada S, Sato T, et al. CA 19–9 in evaluating the response to chemotherapy in advanced pancreatic cancer. Hepatogastroenterology. 1997;44: 279–283. [PubMed] [Google Scholar]
  • 79.Maisey NR, Norman AR, Hill A, Massey A, Oates J, Cunningham D. CA19–9 as a prognostic factor in inoperable pancreatic cancer: the implication for clinical trials. Br J Cancer. 2005;93: 740–743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Azizian A, Rühlmann F, Krause T, et al. CA19–9 for detecting recurrence of pancreatic cancer. Sci Rep. 2020;10: 1332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Locker GY, Hamilton S, Harris J, et al. ASCO 2006 update of recommendations for the use of tumor markers in gastrointestinal cancer. J Clin Oncol. 2006;24: 5313–5327. [DOI] [PubMed] [Google Scholar]
  • 82.Rückert F, Pilarsky C, Grützmann R. Serum tumor markers in pancreatic cancer-recent discoveries. Cancers (Basel). 2010;2: 1107–1124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Tempero MA, Malafa MP, Al-Hawary M, et al. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) - Pancreatic Adenocarcinoma. Available from URL: https://www.nccn.org/professionals/physician_gls/pdf/pancreatic.pdf [accessed 1/22/2020, 2020].
  • 84.Chu LC, Goggins MG, Fishman EK. Diagnosis and Detection of Pancreatic Cancer. Cancer J. 2017;23: 333–342. [DOI] [PubMed] [Google Scholar]
  • 85.Gambhir SS, Czernin J, Schwimmer J, Silverman DH, Coleman RE, Phelps ME. A tabulated summary of the FDG PET literature. J Nucl Med. 2001;42: 1S–93S. [PubMed] [Google Scholar]
  • 86.Krishna SG, Rao BB, Ugbarugba E, et al. Diagnostic performance of endoscopic ultrasound for detection of pancreatic malignancy following an indeterminate multidetector CT scan: a systemic review and meta-analysis. Surg Endosc. 2017. [DOI] [PubMed] [Google Scholar]
  • 87.Harinck F, Konings IC, Kluijt I, et al. A multicentre comparative prospective blinded analysis of EUS and MRI for screening of pancreatic cancer in high-risk individuals. Gut. 2016;65: 1505–1513. [DOI] [PubMed] [Google Scholar]
  • 88.Al-Hawary MM, Francis IR, Chari ST, et al. Pancreatic ductal adenocarcinoma radiology reporting template: consensus statement of the Society of Abdominal Radiology and the American Pancreatic Association. Radiology. 2014;270: 248–260. [DOI] [PubMed] [Google Scholar]
  • 89.Chu LC, Johnson PT, Fishman EK. Cinematic rendering of pancreatic neoplasms: preliminary observations and opportunities. Abdom Radiol (NY). 2018;43: 3009–3015. [DOI] [PubMed] [Google Scholar]
  • 90.Amin MB. AJCC Cancer Staging Manual. Springer, 2017. [Google Scholar]
  • 91.Varadhachary GR, Tamm EP, Abbruzzese JL, et al. Borderline resectable pancreatic cancer: definitions, management, and role of preoperative therapy. Ann Surg Oncol. 2006;13: 1035–1046. [DOI] [PubMed] [Google Scholar]
  • 92.Katz MH, 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: 2787–2795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Callery MP, Chang KJ, Fishman EK, Talamonti MS, William Traverso L, Linehan DC. Pretreatment assessment of resectable and borderline resectable pancreatic cancer: expert consensus statement. Ann Surg Oncol. 2009;16: 1727–1733. [DOI] [PubMed] [Google Scholar]
  • 94.Burris HA 3rd, Moore MJ, Andersen J, 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. [DOI] [PubMed] [Google Scholar]
  • 95.Conroy T, Desseigne F, Ychou M, et al. FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med. 2011;364: 1817–1825. [DOI] [PubMed] [Google Scholar]
  • 96.Von Hoff DD, Ervin T, Arena FP, et al. Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med. 2013;369: 1691–1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Portal A, Pernot S, Tougeron D, et al. Nab-paclitaxel plus gemcitabine for metastatic pancreatic adenocarcinoma after Folfirinox failure: an AGEO prospective multicentre cohort. Br J Cancer. 2015;113: 989–995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Zaanan A, Trouilloud I, Markoutsaki T, et al. FOLFOX as second-line chemotherapy in patients with pretreated metastatic pancreatic cancer from the FIRGEM study. BMC Cancer. 2014;14: 441. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Gill S, Ko YJ, Cripps C, 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. [DOI] [PubMed] [Google Scholar]
  • 100.Wang-Gillam A, Li CP, Bodoky G, et al. Nanoliposomal irinotecan with fluorouracil and folinic acid in metastatic pancreatic cancer after previous gemcitabine-based therapy (NAPOLI-1): a global, randomised, open-label, phase 3 trial. Lancet. 2016;387: 545–557. [DOI] [PubMed] [Google Scholar]
  • 101.Jameson GS, Borazanci E, Babiker HM, et al. Response Rate Following Albumin-Bound Paclitaxel Plus Gemcitabine Plus Cisplatin Treatment Among Patients With Advanced Pancreatic Cancer: A Phase 1b/2 Pilot Clinical Trial. JAMA Oncol. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Dahan L, Phelip JM, Malicot KL, et al. FOLFIRINOX until progression, FOLFIRINOX with maintenance treatment, or sequential treatment with gemcitabine and FOLFIRI.3 for first-line treatment of metastatic pancreatic cancer: A randomized phase II trial (PRODIGE 35-PANOPTIMOX). Journal of Clinical Oncology. 2018;36: 4000–4000. [Google Scholar]
  • 103.Franck C, Canbay A, Malfertheiner P, Venerito M. Maintenance Therapy with FOLFIRI after FOLFIRINOX for Advanced Pancreatic Ductal Adenocarcinoma: A Retrospective Single-Center Analysis. J Oncol. 2019;2019: 5832309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Bailey P, Chang DK, Nones K, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531: 47–52. [DOI] [PubMed] [Google Scholar]
  • 105.Collisson EA, Sadanandam A, Olson P, et al. Subtypes of pancreatic ductal adenocarcinoma and their differing responses to therapy. Nat Med. 2011;17: 500–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Moffitt RA, Marayati R, Flate EL, et al. Virtual microdissection identifies distinct tumor- and stroma-specific subtypes of pancreatic ductal adenocarcinoma. Nat Genet. 2015;47: 1168–1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Aung KL, Fischer SE, Denroche RE, et al. Genomics-Driven Precision Medicine for Advanced Pancreatic Cancer: Early Results from the COMPASS Trial. Clin Cancer Res. 2018;24: 1344–1354. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Porter RL, Magnus NKC, Thapar V, et al. Epithelial to mesenchymal plasticity and differential response to therapies in pancreatic ductal adenocarcinoma. Proc Natl Acad Sci U S A. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Ligorio M, Sil S, Malagon-Lopez J, et al. Stromal Microenvironment Shapes the Intratumoral Architecture of Pancreatic Cancer. Cell. 2019;178: 160–175.e127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Aguirre AJ, Nowak JA, Camarda ND, et al. Real-time Genomic Characterization of Advanced Pancreatic Cancer to Enable Precision Medicine. Cancer Discov. 2018;8: 1096–1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Shroff RT, Hendifar A, McWilliams RR, et al. Rucaparib Monotherapy in Patients With Pancreatic Cancer and a Known Deleterious BRCA Mutation. JCO Precis Oncol. 2018;2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Fok JHL, Ramos-Montoya A, Vazquez-Chantada M, et al. AZD7648 is a potent and selective DNA-PK inhibitor that enhances radiation, chemotherapy and olaparib activity. Nat Commun. 2019;10: 5065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Lohse I, Kumareswaran R, Cao P, et al. Effects of Combined Treatment with Ionizing Radiation and the PARP Inhibitor Olaparib in BRCA Mutant and Wild Type Patient-Derived Pancreatic Cancer Xenografts. PLoS One. 2016;11: e0167272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.O’Reilly EM, Lee JW, Zalupski M, et al. Randomized, Multicenter, Phase II Trial of Gemcitabine and Cisplatin With or Without Veliparib in Patients With Pancreas Adenocarcinoma and a Germline BRCA/PALB2 Mutation. J Clin Oncol. 2020: Jco1902931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Hu ZI, Shia J, Stadler ZK, et al. Evaluating Mismatch Repair Deficiency in Pancreatic Adenocarcinoma: Challenges and Recommendations. Clin Cancer Res. 2018;24: 1326–1336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Le DT, Durham JN, Smith KN, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017;357: 409–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Le DT, Uram JN, Wang H, et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med. 2015;372: 2509–2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Pishvaian MJ, Blais EM, Brody JR, et al. Overall survival in patients with pancreatic cancer receiving matched therapies following molecular profiling: a retrospective analysis of the Know Your Tumor registry trial. Lancet Oncol. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Amedei A, Niccolai E, Prisco D. Pancreatic cancer: role of the immune system in cancer progression and vaccine-based immunotherapy. Hum Vaccin Immunother. 2014;10: 3354–3368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Apte MV, Wilson JS, Lugea A, Pandol SJ. A starring role for stellate cells in the pancreatic cancer microenvironment. Gastroenterology. 2013;144: 1210–1219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Clark CE, Hingorani SR, Mick R, Combs C, Tuveson DA, Vonderheide RH. Dynamics of the immune reaction to pancreatic cancer from inception to invasion. Cancer Res. 2007;67: 9518–9527. [DOI] [PubMed] [Google Scholar]
  • 122.Erkan M, Hausmann S, Michalski CW, et al. The role of stroma in pancreatic cancer: diagnostic and therapeutic implications. Nat Rev Gastroenterol Hepatol. 2012;9: 454–467. [DOI] [PubMed] [Google Scholar]
  • 123.Yamaki S, Yanagimoto H, Tsuta K, Ryota H, Kon M. PD-L1 expression in pancreatic ductal adenocarcinoma is a poor prognostic factor in patients with high CD8(+) tumor-infiltrating lymphocytes: highly sensitive detection using phosphor-integrated dot staining. Int J Clin Oncol. 2017;22: 726–733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Aglietta M, Barone C, Sawyer MB, et al. A phase I dose escalation trial of tremelimumab (CP-675,206) in combination with gemcitabine in chemotherapy-naive patients with metastatic pancreatic cancer. Ann Oncol. 2014;25: 1750–1755. [DOI] [PubMed] [Google Scholar]
  • 125.Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366: 2455–2465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Royal RE, Levy C, Turner K, et al. Phase 2 trial of single agent Ipilimumab (anti-CTLA-4) for locally advanced or metastatic pancreatic adenocarcinoma. J Immunother. 2010;33: 828–833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.O’Reilly EM, Oh DY, Dhani N, et al. Durvalumab With or Without Tremelimumab for Patients With Metastatic Pancreatic Ductal Adenocarcinoma: A Phase 2 Randomized Clinical Trial. JAMA Oncol. 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Feig C, Jones JO, Kraman M, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci U S A. 2013;110: 20212–20217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Seo YD, Jiang X, Sullivan KM, et al. Mobilization of CD8(+) T Cells via CXCR4 Blockade Facilitates PD-1 Checkpoint Therapy in Human Pancreatic Cancer. Clin Cancer Res. 2019;25: 3934–3945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Bennett SR, Carbone FR, Karamalis F, Flavell RA, Miller JF, Heath WR. Help for cytotoxic-T-cell responses is mediated by CD40 signalling. Nature. 1998;393: 478–480. [DOI] [PubMed] [Google Scholar]
  • 131.Ridge JP, Di Rosa F, Matzinger P. A conditioned dendritic cell can be a temporal bridge between a CD4+ T-helper and a T-killer cell. Nature. 1998;393: 474–478. [DOI] [PubMed] [Google Scholar]
  • 132.Schoenberger SP, Toes RE, van der Voort EI, Offringa R, Melief CJ. T-cell help for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions. Nature. 1998;393: 480–483. [DOI] [PubMed] [Google Scholar]
  • 133.Beatty GL, Chiorean EG, Fishman MP, et al. CD40 Agonists Alter Tumor Stroma and Show Efficacy Against Pancreatic Carcinoma in Mice and Humans. Science. 2011;331: 1612–1616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.O’Hara MH, O’Reilly EM, Rosemarie M, et al. Abstract CT004: A Phase Ib study of CD40 agonistic monoclonal antibody APX005M together with gemcitabine (Gem) and nab-paclitaxel (NP) with or without nivolumab (Nivo) in untreated metastatic ductal pancreatic adenocarcinoma (PDAC) patients. Cancer Research. 2019;79: CT004–CT004. [Google Scholar]
  • 135.Rodriguez-Ruiz ME, Vanpouille-Box C, Melero I, Formenti SC, Demaria S. Immunological Mechanisms Responsible for Radiation-Induced Abscopal Effect. Trends Immunol. 2018;39: 644–655. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Twyman-Saint Victor C, Rech AJ, Maity A, et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature. 2015;520: 373–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Parikh A, Wo JY-L, Ryan DP, et al. A phase II study of ipilimumab and nivolumab with radiation in metastatic pancreatic adenocarcinoma. Journal of Clinical Oncology. 2019;37: 391–391. [Google Scholar]
  • 138.Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21: 418–429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Hingorani SR, Zheng L, Bullock AJ, et al. HALO 202: Randomized Phase II Study of PEGPH20 Plus Nab-Paclitaxel/Gemcitabine Versus Nab-Paclitaxel/Gemcitabine in Patients With Untreated, Metastatic Pancreatic Ductal Adenocarcinoma. J Clin Oncol. 2018;36: 359–366. [DOI] [PubMed] [Google Scholar]
  • 140.Ramanathan RK, McDonough SL, Philip PA, et al. Phase IB/II Randomized Study of FOLFIRINOX Plus Pegylated Recombinant Human Hyaluronidase Versus FOLFIRINOX Alone in Patients With Metastatic Pancreatic Adenocarcinoma: SWOG S1313. J Clin Oncol. 2019;37: 1062–1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Murphy JE, Wo JY, Ryan DP, et al. Total Neoadjuvant Therapy With FOLFIRINOX in Combination With Losartan Followed by Chemoradiotherapy for Locally Advanced Pancreatic Cancer: A Phase 2 Clinical Trial. JAMA Oncol. 2019;5: 1020–1027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Liu H, Naxerova K, Pinter M, et al. Use of Angiotensin System Inhibitors Is Associated with Immune Activation and Longer Survival in Nonmetastatic Pancreatic Ductal Adenocarcinoma. Clinical Cancer Research. 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Wilentz RE, Iacobuzio-Donahue CA, Argani P, et al. Loss of expression of Dpc4 in pancreatic intraepithelial neoplasia: evidence that DPC4 inactivation occurs late in neoplastic progression. Cancer Res. 2000;60: 2002–2006. [PubMed] [Google Scholar]
  • 144.Santana-Codina N, Roeth AA, Zhang Y, et al. Oncogenic KRAS supports pancreatic cancer through regulation of nucleotide synthesis. Nat Commun. 2018;9: 4945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Ying H, Kimmelman AC, Lyssiotis CA, et al. Oncogenic Kras maintains pancreatic tumors through regulation of anabolic glucose metabolism. Cell. 2012;149: 656–670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Commisso C, Davidson SM, Soydaner-Azeloglu RG, et al. Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature. 2013;497: 633–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Viale A, Pettazzoni P, Lyssiotis CA, et al. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature. 2014;514: 628–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Bryant KL, Mancias JD, Kimmelman AC, Der CJ. KRAS: feeding pancreatic cancer proliferation. Trends Biochem Sci. 2014;39: 91–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Yang S, Wang X, Contino G, et al. Pancreatic cancers require autophagy for tumor growth. Genes Dev. 2011;25: 717–729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Yang A, Herter-Sprie G, Zhang H, et al. Autophagy Sustains Pancreatic Cancer Growth through Both Cell-Autonomous and Nonautonomous Mechanisms. Cancer Discov. 2018;8: 276–287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Bryant KL, Stalnecker CA, Zeitouni D, et al. Combination of ERK and autophagy inhibition as a treatment approach for pancreatic cancer. Nat Med. 2019;25: 628–640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Lee CS, Lee LC, Yuan TL, et al. MAP kinase and autophagy pathways cooperate to maintain RAS mutant cancer cell survival. Proc Natl Acad Sci U S A. 2019;116: 4508–4517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Kagedan DJ, Goyert N, Li Q, et al. The Impact of Increasing Hospital Volume on 90-Day Postoperative Outcomes Following Pancreaticoduodenectomy. J Gastrointest Surg. 2017;21: 506–515. [DOI] [PubMed] [Google Scholar]
  • 154.Pugalenthi A, Protic M, Gonen M, et al. Postoperative complications and overall survival after pancreaticoduodenectomy for pancreatic ductal adenocarcinoma. J Surg Oncol. 2016;113: 188–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.de Rooij T, van Hilst J, van Santvoort H, et al. Minimally Invasive Versus Open Distal Pancreatectomy (LEOPARD): A Multicenter Patient-blinded Randomized Controlled Trial. Ann Surg. 2019;269: 2–9. [DOI] [PubMed] [Google Scholar]
  • 156.Kutlu OC, Lee JE, Katz MH, et al. Open Pancreaticoduodenectomy Case Volume Predicts Outcome of Laparoscopic Approach: A Population-based Analysis. Ann Surg. 2018;267: 552–560. [DOI] [PubMed] [Google Scholar]
  • 157.van Hilst J, de Rooij T, Bosscha K, et al. Laparoscopic versus open pancreatoduodenectomy for pancreatic or periampullary tumours (LEOPARD-2): a multicentre, patient-blinded, randomised controlled phase 2/3 trial. Lancet Gastroenterol Hepatol. 2019;4: 199–207. [DOI] [PubMed] [Google Scholar]
  • 158.Katz MHG, Merchant NB, Brower S, et al. Standardization of Surgical and Pathologic Variables is Needed in Multicenter Trials of Adjuvant Therapy for Pancreatic Cancer: Results from the ACOSOG Z5031 Trial. Annals of Surgical Oncology. 2011;18: 337–344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.College of American Pathologists. Protocol for the Examination of Specimens From Patients With Carcinoma of the Pancreas. Available from URL: https://documents.cap.org/protocols/cp-gihepatobiliary-pancreasexocrine-20-4100.pdf [accessed May 21, 2020.
  • 160.Raut CP, Tseng JF, Sun CC, et al. Impact of Resection Status on Pattern of Failure and Survival After Pancreaticoduodenectomy for Pancreatic Adenocarcinoma. Annals of Surgery. 2007;246: 52–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Verbeke CS. Resection margins and R1 rates in pancreatic cancer – are we there yet? Histopathology. 2008;52: 787–796. [DOI] [PubMed] [Google Scholar]
  • 162.Osipov A, Nissen N, Rutgers J, et al. Redefining the Positive Margin in Pancreatic Cancer: Impact on Patterns of Failure, Long-Term Survival and Adjuvant Therapy. Ann Surg Oncol. 2017;24: 3674–3682. [DOI] [PubMed] [Google Scholar]
  • 163.Kim KS, Kwon J, Kim K, Chie EK. Impact of Resection Margin Distance on Survival of Pancreatic Cancer: A Systematic Review and Meta-Analysis. Cancer Res Treat. 2017;49: 824–833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.White RR, Xie HB, Gottfried MR, et al. Significance of histological response to preoperative chemoradiotherapy for pancreatic cancer. Ann Surg Oncol. 2005;12: 214–221. [DOI] [PubMed] [Google Scholar]
  • 165.Evans DB, Rich TA, Byrd DR, et al. Preoperative Chemoradiation and Pancreaticoduodenectomy for Adenocarcinoma of the Pancreas. Archives of Surgery. 1992;127: 1335–1339. [DOI] [PubMed] [Google Scholar]
  • 166.Ishikawa O, Ohhigashi H, Teshima T, et al. Clinical and histopathological appraisal of preoperative irradiation for adenocarcinoma of the pancreatoduodenal region. Journal of Surgical Oncology. 1989;40: 143–151. [DOI] [PubMed] [Google Scholar]
  • 167.Kang CM, Hwang HK, Choi SH, Lee WJ. Controversial issues of neoadjuvant treatment in borderline resectable pancreatic cancer. Surg Oncol. 2013;22: 123–131. [DOI] [PubMed] [Google Scholar]
  • 168.Brennan MF, Kattan MW, Klimstra D, Conlon K. Prognostic nomogram for patients undergoing resection for adenocarcinoma of the pancreas. Ann Surg. 2004;240: 293–298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Lee SR, Kim HO, Son BH, Yoo CH, Shin JH. Prognostic factors associated with long-term survival and recurrence in pancreatic adenocarcinoma. Hepatogastroenterology. 2013;60: 358–362. [DOI] [PubMed] [Google Scholar]
  • 170.Schwarz RE, Smith DD. Extent of lymph node retrieval and pancreatic cancer survival: information from a large US population database. Ann Surg Oncol. 2006;13: 1189–1200. [DOI] [PubMed] [Google Scholar]
  • 171.Tomlinson JS, Jain S, Bentrem DJ, et al. Accuracy of staging node-negative pancreas cancer: a potential quality measure. Arch Surg. 2007;142: 767–723; discussion 773–764. [DOI] [PubMed] [Google Scholar]
  • 172.Philips P, Dunki-Jacobs E, Agle SC, Scoggins C, McMasters KM, Martin RC. The role of hepatic artery lymph node in pancreatic adenocarcinoma: prognostic factor or a selection criterion for surgery. HPB (Oxford). 2014;16: 1051–1055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Cordera F, Arciero CA, Li T, Watson JC, Hoffman JP. Significance of common hepatic artery lymph node metastases during pancreaticoduodenectomy for pancreatic head adenocarcinoma. Ann Surg Oncol. 2007;14: 2330–2336. [DOI] [PubMed] [Google Scholar]
  • 174.Connor S, Bosonnet L, Ghaneh P, et al. Survival of patients with periampullary carcinoma is predicted by lymph node 8a but not by lymph node 16b1 status. Br J Surg. 2004;91: 1592–1599. [DOI] [PubMed] [Google Scholar]
  • 175.Oettle H, Neuhaus P, Hochhaus A, et al. Adjuvant chemotherapy with gemcitabine and long-term outcomes among patients with resected pancreatic cancer: the CONKO-001 randomized trial. JAMA. 2013;310: 1473–1481. [DOI] [PubMed] [Google Scholar]
  • 176.Tempero MA, Reni M, Riess H, et al. APACT: phase III, multicenter, international, open-label, randomized trial of adjuvant nab-paclitaxel plus gemcitabine (nab-P/G) vs gemcitabine (G) for surgically resected pancreatic adenocarcinoma. Journal of Clinical Oncology. 2019;37: 4000–4000. [Google Scholar]
  • 177.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: 1011–1024. [DOI] [PubMed] [Google Scholar]
  • 178.Hazard L, Tward JD, Szabo A, Shrieve DC. Radiation therapy is associated with improved survival in patients with pancreatic adenocarcinoma: results of a study from the Surveillance, Epidemiology, and End Results (SEER) registry data. Cancer. 2007;110: 2191–2201. [DOI] [PubMed] [Google Scholar]
  • 179.Kooby DA, Gillespie TW, Liu Y, et al. Impact of adjuvant radiotherapy on survival after pancreatic cancer resection: an appraisal of data from the national cancer data base. Ann Surg Oncol. 2013;20: 3634–3642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Rutter CE, Park HS, Corso CD, et al. Addition of radiotherapy to adjuvant chemotherapy is associated with improved overall survival in resected pancreatic adenocarcinoma: An analysis of the National Cancer Data Base. Cancer. 2015;121: 4141–4149. [DOI] [PubMed] [Google Scholar]
  • 181.Torgeson A, Tao R, Garrido-Laguna I, Willen B, Dursteler A, Lloyd S. Large database utilization in health outcomes research in pancreatic cancer: an update. J Gastrointest Oncol. 2018;9: 996–1004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Goodman KA, Regine WF, Dawson LA, et al. Radiation Therapy Oncology Group consensus panel guidelines for the delineation of the clinical target volume in the postoperative treatment of pancreatic head cancer. Int J Radiat Oncol Biol Phys. 2012;83: 901–908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Heestand GM, Murphy JD, Lowy AM. Approach to patients with pancreatic cancer without detectable metastases. J Clin Oncol. 2015;33: 1770–1778. [DOI] [PubMed] [Google Scholar]
  • 184.Versteijne E, Suker M, Groothuis K, et al. Preoperative Chemoradiotherapy Versus Immediate Surgery for Resectable and Borderline Resectable Pancreatic Cancer: Results of the Dutch Randomized Phase III PREOPANC Trial. J Clin Oncol. 2020: Jco1902274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Mokdad AA, Minter RM, Zhu H, et al. Neoadjuvant Therapy Followed by Resection Versus Upfront Resection for Resectable Pancreatic Cancer: A Propensity Score Matched Analysis. J Clin Oncol. 2017;35: 515–522. [DOI] [PubMed] [Google Scholar]
  • 186.Sohal D, McDonough S, Ahmad SA, et al. SWOG S1505: Initial findings on eligibility and neoadjuvant chemotherapy experience with mfolfirinox versus gemcitabine/nab-paclitaxel for resectable pancreatic adenocarcinoma. Journal of Clinical Oncology. 2019;37: 414–414. [Google Scholar]
  • 187.Katz MH, 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: e161137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.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: 963–969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Cho IR, Chung MJ, Bang S, et al. Gemcitabine based neoadjuvant chemoradiotherapy therapy in patients with borderline resectable pancreatic cancer. Pancreatology. 2013;13: 539–543. [DOI] [PubMed] [Google Scholar]
  • 190.Stokes JB, Nolan NJ, Stelow EB, et al. Preoperative capecitabine and concurrent radiation for borderline resectable pancreatic cancer. Ann Surg Oncol. 2011;18: 619–627. [DOI] [PubMed] [Google Scholar]
  • 191.Cloyd JM, Chen HC, Wang X, et al. Chemotherapy Versus Chemoradiation as Preoperative Therapy for Resectable Pancreatic Ductal Adenocarcinoma: A Propensity Score Adjusted Analysis. Pancreas. 2019;48: 216–222. [DOI] [PubMed] [Google Scholar]
  • 192.Katz MHG, Ou FS, Herman JM, et al. Alliance for clinical trials in oncology (ALLIANCE) trial A021501: preoperative extended chemotherapy vs. chemotherapy plus hypofractionated radiation therapy for borderline resectable adenocarcinoma of the head of the pancreas. BMC Cancer. 2017;17: 505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Wang LS, Shaikh T, Handorf EA, Hoffman JP, Cohen SJ, Meyer JE. Dose escalation with a vessel boost in pancreatic adenocarcinoma treated with neoadjuvant chemoradiation. Pract Radiat Oncol. 2015;5: e457–e463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Krishnan S, Chadha AS, Suh Y, et al. Focal Radiation Therapy Dose Escalation Improves Overall Survival in Locally Advanced Pancreatic Cancer Patients Receiving Induction Chemotherapy and Consolidative Chemoradiation. Int J Radiat Oncol Biol Phys. 2016;94: 755–765. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Morganti AG, Cellini F, Buwenge M, et al. Adjuvant chemoradiation in pancreatic cancer: impact of radiotherapy dose on survival. BMC Cancer. 2019;19: 569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Bernard V, Herman JM. Pancreas SBRT: Who, What, When, Where, and How. Pract Radiat Oncol. 2019. [DOI] [PubMed] [Google Scholar]
  • 197.Herman JM, Chang DT, Goodman KA, et al. Phase 2 multi-institutional trial evaluating gemcitabine and stereotactic body radiotherapy for patients with locally advanced unresectable pancreatic adenocarcinoma. Cancer. 2015;121: 1128–1137. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.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: 322–332. [DOI] [PubMed] [Google Scholar]
  • 199.Yu XZ, Li J, Fu DL, et al. Benefit from synchronous portal-superior mesenteric vein resection during pancreaticoduodenectomy for cancer: a meta-analysis. Eur J Surg Oncol. 2014;40: 371–378. [DOI] [PubMed] [Google Scholar]
  • 200.Zhou Y, Zhang Z, Liu Y, Li B, Xu D. Pancreatectomy combined with superior mesenteric vein-portal vein resection for pancreatic cancer: a meta-analysis. World J Surg. 2012;36: 884–891. [DOI] [PubMed] [Google Scholar]
  • 201.Ghaneh P, Kleeff J, Halloran CM, et al. The Impact of Positive Resection Margins on Survival and Recurrence Following Resection and Adjuvant Chemotherapy for Pancreatic Ductal Adenocarcinoma. Ann Surg. 2019;269: 520–529. [DOI] [PubMed] [Google Scholar]
  • 202.Tucker ON, Rela M. Controversies in the management of borderline resectable proximal pancreatic adenocarcinoma with vascular involvement. HPB Surg. 2008;2008: 839503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Balaban EP, Mangu PB, Khorana AA, et al. Locally Advanced, Unresectable Pancreatic Cancer: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2016;34: 2654–2668. [DOI] [PubMed] [Google Scholar]
  • 204.Suker M, Beumer BR, Sadot E, et al. FOLFIRINOX for locally advanced pancreatic cancer: a systematic review and patient-level meta-analysis. Lancet Oncol. 2016;17: 801–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Mukherjee S, Hurt CN, Bridgewater J, et al. Gemcitabine-based or capecitabine-based chemoradiotherapy for locally advanced pancreatic cancer (SCALOP): a multicentre, randomised, phase 2 trial. Lancet Oncol. 2013;14: 317–326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Hammel P, Huguet F, van Laethem JL, et al. Effect of Chemoradiotherapy vs Chemotherapy on Survival in Patients With Locally Advanced Pancreatic Cancer Controlled After 4 Months of Gemcitabine With or Without Erlotinib: The LAP07 Randomized Clinical Trial. JAMA. 2016;315: 1844–1853. [DOI] [PubMed] [Google Scholar]
  • 207.Hoyer M, Roed H, Sengelov L, et al. Phase-II study on stereotactic radiotherapy of locally advanced pancreatic carcinoma. Radiother Oncol. 2005;76: 48–53. [DOI] [PubMed] [Google Scholar]
  • 208.Schellenberg D, Goodman KA, Lee F, et al. Gemcitabine chemotherapy and single-fraction stereotactic body radiotherapy for locally advanced pancreatic cancer. Int J Radiat Oncol Biol Phys. 2008;72: 678–686. [DOI] [PubMed] [Google Scholar]
  • 209.Reyngold M, O’Reilly E, Zinovoy M, et al. Ablative RT Results in Excellent Local Control and Survival in Localized Pancreatic Cancer. International Journal of Radiation Oncology • Biology • Physics. 2019;105: S206. [Google Scholar]
  • 210.Rudra S, Jiang N, Rosenberg SA, et al. High Dose Adaptive MRI Guided Radiation Therapy Improves Overall Survival of Inoperable Pancreatic Cancer. International Journal of Radiation Oncology • Biology • Physics. 2017;99: E184. [Google Scholar]
  • 211.Koay EJ, Hanania AN, Hall WA, et al. Dose-Escalated Radiation Therapy for Pancreatic Cancer: A Simultaneous Integrated Boost Approach. Pract Radiat Oncol. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.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: 12–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Gemenetzis G, Groot VP, Blair AB, et al. Survival in Locally Advanced Pancreatic Cancer After Neoadjuvant Therapy and Surgical Resection. Ann Surg. 2019;270: 340–347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Truty MJ, Kendrick ML, Nagorney DM, et al. Factors Predicting Response, Perioperative Outcomes, and Survival Following Total Neoadjuvant Therapy for Borderline/Locally Advanced Pancreatic Cancer. Ann Surg. 2019. [DOI] [PubMed] [Google Scholar]
  • 215.Gong H, Ma R, Gong J, Cai C, Song Z, Xu B. Distal Pancreatectomy With En Bloc Celiac Axis Resection for Locally Advanced Pancreatic Cancer: A Systematic Review and Meta-Analysis. Medicine (Baltimore). 2016;95: e3061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Mollberg N, Rahbari NN, Koch M, et al. Arterial resection during pancreatectomy for pancreatic cancer: a systematic review and meta-analysis. Ann Surg. 2011;254: 882–893. [DOI] [PubMed] [Google Scholar]
  • 217.Khorana AA, Francis CW, Culakova E, Lyman GH. Risk factors for chemotherapy-associated venous thromboembolism in a prospective observational study. Cancer. 2005;104: 2822–2829. [DOI] [PubMed] [Google Scholar]
  • 218.Chew HK, Wun T, Harvey D, Zhou H, White RH. Incidence of venous thromboembolism and its effect on survival among patients with common cancers. Arch Intern Med. 2006;166: 458–464. [DOI] [PubMed] [Google Scholar]
  • 219.Maraveyas A, Waters J, Roy R, et al. Gemcitabine versus gemcitabine plus dalteparin thromboprophylaxis in pancreatic cancer. Eur J Cancer. 2012;48: 1283–1292. [DOI] [PubMed] [Google Scholar]
  • 220.Pelzer U, Opitz B, Deutschinoff G, et al. Efficacy of Prophylactic Low–Molecular Weight Heparin for Ambulatory Patients With Advanced Pancreatic Cancer: Outcomes From the CONKO-004 Trial. Journal of Clinical Oncology. 2015;33: 2028–2034. [DOI] [PubMed] [Google Scholar]
  • 221.Khorana AA, Soff GA, Kakkar AK, et al. Rivaroxaban for Thromboprophylaxis in High-Risk Ambulatory Patients with Cancer. N Engl J Med. 2019;380: 720–728. [DOI] [PubMed] [Google Scholar]
  • 222.Carrier M, Abou-Nassar K, Mallick R, et al. Apixaban to Prevent Venous Thromboembolism in Patients with Cancer. N Engl J Med. 2019;380: 711–719. [DOI] [PubMed] [Google Scholar]
  • 223.Key NS, Khorana AA, Kuderer NM, et al. Venous Thromboembolism Prophylaxis and Treatment in Patients With Cancer: ASCO Clinical Practice Guideline Update. Journal of Clinical Oncology. 2020;38: 496–520. [DOI] [PubMed] [Google Scholar]
  • 224.Laitinen I, Sand J, Peromaa P, Nordback I, Laukkarinen J. Quality of life in patients with pancreatic ductal adenocarcinoma undergoing pancreaticoduodenectomy. Pancreatology. 2017;17: 445–450. [DOI] [PubMed] [Google Scholar]
  • 225.Laquente B, Calsina-Berna A, Carmona-Bayonas A, Jimenez-Fonseca P, Peiro I, Carrato A. Supportive care in pancreatic ductal adenocarcinoma. Clin Transl Oncol. 2017;19: 1293–1302. [DOI] [PubMed] [Google Scholar]
  • 226.Moffat GT, Epstein AS, O’Reilly EM. Pancreatic cancer-A disease in need: Optimizing and integrating supportive care. Cancer. 2019;125: 3927–3935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Gourgou-Bourgade S, Bascoul-Mollevi C, Desseigne F, et al. Impact of FOLFIRINOX compared with gemcitabine on quality of life in patients with metastatic pancreatic cancer: results from the PRODIGE 4/ACCORD 11 randomized trial. J Clin Oncol. 2013;31: 23–29. [DOI] [PubMed] [Google Scholar]
  • 228.Fitzsimmons D, Johnson CD, George S, et al. Development of a disease specific quality of life (QoL) questionnaire module to supplement the EORTC core cancer QoL questionnaire, the QLQ-C30 in patients with pancreatic cancer. EORTC Study Group on Quality of Life. Eur J Cancer. 1999;35: 939–941. [DOI] [PubMed] [Google Scholar]
  • 229.Karimi M, Brazier J. Health, Health-Related Quality of Life, and Quality of Life: What is the Difference? Pharmacoeconomics. 2016;34: 645–649. [DOI] [PubMed] [Google Scholar]
  • 230.Macarulla T, Hendifar AE, Li CP, et al. Landscape of Health-Related Quality of Life in Patients With Early-Stage Pancreatic Cancer Receiving Adjuvant or Neoadjuvant Chemotherapy: A Systematic Literature Review. Pancreas. 2020;49: 393–407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Walter FM, Mills K, Mendonca SC, et al. Symptoms and patient factors associated with diagnostic intervals for pancreatic cancer (SYMPTOM pancreatic study): a prospective cohort study. Lancet Gastroenterol Hepatol. 2016;1: 298–306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Dantzer R, O’Connor JC, Freund GG, Johnson RW, Kelley KW. From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci. 2008;9: 46–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.McCusker RH, Kelley KW. Immune-neural connections: how the immune system’s response to infectious agents influences behavior. J Exp Biol. 2013;216: 84–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Argiles JM, Busquets S, Stemmler B, Lopez-Soriano FJ. Cancer cachexia: understanding the molecular basis. Nat Rev Cancer. 2014;14: 754–762. [DOI] [PubMed] [Google Scholar]
  • 235.von Haehling S, Anker SD. Cachexia as a major underestimated and unmet medical need: facts and numbers. J Cachexia Sarcopenia Muscle. 2010;1: 1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Baracos VE, Martin L, Korc M, Guttridge DC, Fearon KCH. Cancer-associated cachexia. Nat Rev Dis Primers. 2018;4: 17105. [DOI] [PubMed] [Google Scholar]
  • 237.Takayama K, Katakami N, Yokoyama T, et al. Anamorelin (ONO-7643) in Japanese patients with non-small cell lung cancer and cachexia: results of a randomized phase 2 trial. Support Care Cancer. 2016;24: 3495–3505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Temel JS, Abernethy AP, Currow DC, et al. Anamorelin in patients with non-small-cell lung cancer and cachexia (ROMANA 1 and ROMANA 2): results from two randomised, double-blind, phase 3 trials. Lancet Oncol. 2016;17: 519–531. [DOI] [PubMed] [Google Scholar]
  • 239.Ruiz Garcia V, Lopez-Briz E, Carbonell Sanchis R, Gonzalvez Perales JL, Bort-Marti S. Megestrol acetate for treatment of anorexia-cachexia syndrome. Cochrane Database Syst Rev. 2013: CD004310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Christensen JF, Simonsen C, Hojman P. Exercise Training in Cancer Control and Treatment. Compr Physiol. 2018;9: 165–205. [DOI] [PubMed] [Google Scholar]
  • 241.Kamarajah SK, Bundred J, Weblin J, Tan BHL. Critical appraisal on the impact of preoperative rehabilitation and outcomes after major abdominal and cardiothoracic surgery: A systematic review and meta-analysis. Surgery. 2020;167: 540–549. [DOI] [PubMed] [Google Scholar]
  • 242.Cangemi DJ, Kuo B. Practical Perspectives in the Treatment of Nausea and Vomiting. J Clin Gastroenterol. 2019;53: 170–178. [DOI] [PubMed] [Google Scholar]
  • 243.Mortimer TL, Mabin T, Engelbrecht AM. Cannabinoids: the lows and the highs of chemotherapy-induced nausea and vomiting. Future Oncol. 2019;15: 1035–1049. [DOI] [PubMed] [Google Scholar]
  • 244.Mak RH, Cheung W, Cone RD, Marks DL. Mechanisms of disease: Cytokine and adipokine signaling in uremic cachexia. Nat Clin Pract Nephrol. 2006;2: 527–534. [DOI] [PubMed] [Google Scholar]
  • 245.Marks DL, Cone RD. Central melanocortins and the regulation of weight during acute and chronic disease. Recent Prog Horm Res. 2001;56: 359–375. [DOI] [PubMed] [Google Scholar]
  • 246.Brennan GT, Saif MW. Pancreatic Enzyme Replacement Therapy: A Concise Review. JOP. 2019;20: 121–125. [PMC free article] [PubMed] [Google Scholar]
  • 247.Davidson W, Ash S, Capra S, Bauer J, Cancer Cachexia Study G. Weight stabilisation is associated with improved survival duration and quality of life in unresectable pancreatic cancer. Clin Nutr. 2004;23: 239–247. [DOI] [PubMed] [Google Scholar]
  • 248.Gooden HM, White KJ. Pancreatic cancer and supportive care--pancreatic exocrine insufficiency negatively impacts on quality of life. Support Care Cancer. 2013;21: 1835–1841. [DOI] [PubMed] [Google Scholar]
  • 249.Aadam AA, Liu K. Endoscopic palliation of biliary obstruction. J Surg Oncol. 2019;120: 57–64. [DOI] [PubMed] [Google Scholar]
  • 250.van den Bosch RP, van der Schelling GP, Klinkenbijl JH, Mulder PG, van Blankenstein M, Jeekel J. Guidelines for the application of surgery and endoprostheses in the palliation of obstructive jaundice in advanced cancer of the pancreas. Ann Surg. 1994;219: 18–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Barkay O, Mosler P, Schmitt CM, et al. Effect of endoscopic stenting of malignant bile duct obstruction on quality of life. J Clin Gastroenterol. 2013;47: 526–531. [DOI] [PubMed] [Google Scholar]
  • 252.Salgado SM, Gaidhane M, Kahaleh M. Endoscopic palliation of malignant biliary strictures. World J Gastrointest Oncol. 2016;8: 240–247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Mitchell T, Clarke L, Goldberg A, Bishop KS. Pancreatic Cancer Cachexia: The Role of Nutritional Interventions. Healthcare (Basel). 2019;7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Thong MSY, van Noorden CJF, Steindorf K, Arndt V. Cancer-Related Fatigue: Causes and Current Treatment Options. Curr Treat Options Oncol. 2020;21: 17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Diouf M, Filleron T, Pointet AL, et al. Prognostic value of health-related quality of life in patients with metastatic pancreatic adenocarcinoma: a random forest methodology. Qual Life Res. 2016;25: 1713–1723. [DOI] [PubMed] [Google Scholar]
  • 256.Campbell KL, Zadravec K, Bland KA, Chesley E, Wolf F, Janelsins MC. The Effect of Exercise on Cancer-Related Cognitive Impairment and Applications for Physical Therapy: Systematic Review of Randomized Controlled Trials. Phys Ther. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Fras I, Litin EM. Comparison of psychiatric manifestations in carcinoma of the pancreas, retroperitoneal malignant lymphoma, and lymphoma in other locations. Psychosomatics. 1967;8: 275–277. [DOI] [PubMed] [Google Scholar]
  • 258.Olson B, Marks DL. Pretreatment Cancer-Related Cognitive Impairment-Mechanisms and Outlook. Cancers (Basel). 2019;11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Tang CC, Von Ah D, Fulton JS. The Symptom Experience of Patients With Advanced Pancreatic Cancer: An Integrative Review. Cancer Nurs. 2018;41: 33–44. [DOI] [PubMed] [Google Scholar]
  • 260.Lewis AR, Pihlak R, McNamara MG. The importance of quality-of-life management in patients with advanced pancreatic ductal adenocarcinoma. Curr Probl Cancer. 2018;42: 26–39. [DOI] [PubMed] [Google Scholar]
  • 261.Robinson DW Jr., Eisenberg DF, Cella D, Zhao N, de Boer C, DeWitte M. The prognostic significance of patient-reported outcomes in pancreatic cancer cachexia. J Support Oncol. 2008;6: 283–290. [PubMed] [Google Scholar]
  • 262.Massie MJ. Prevalence of depression in patients with cancer. J Natl Cancer Inst Monogr. 2004: 57–71. [DOI] [PubMed] [Google Scholar]
  • 263.Parker G, Brotchie H. Pancreatic Cancer and Depression: A Narrative Review. J Nerv Ment Dis. 2017;205: 487–490. [DOI] [PubMed] [Google Scholar]
  • 264.Mayr M, Schmid RM. Pancreatic cancer and depression: myth and truth. BMC Cancer. 2010;10: 569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Basch E, Deal AM, Dueck AC, et al. Overall Survival Results of a Trial Assessing Patient-Reported Outcomes for Symptom Monitoring During Routine Cancer Treatment. JAMA. 2017;318: 197–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Basch E, Deal AM, Kris MG, et al. Symptom Monitoring With Patient-Reported Outcomes During Routine Cancer Treatment: A Randomized Controlled Trial. J Clin Oncol. 2016;34: 557–565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 267.Currow DC, Allingham S, Yates P, Johnson C, Clark K, Eagar K. Improving national hospice/palliative care service symptom outcomes systematically through point-of-care data collection, structured feedback and benchmarking. Support Care Cancer. 2015;23: 307–315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting Cancer Incidence and Deaths to 2030: The Unexpected Burden of Thyroid, Liver, and Pancreas Cancers in the United States. Cancer Research. 2014. [DOI] [PubMed] [Google Scholar]
  • 269.Potjer TP, Kranenburg HE, Bergman W, et al. Prospective risk of cancer and the influence of tobacco use in carriers of the p16-Leiden germline variant. Eur J Hum Genet. 2015;23: 711–714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Goldstein AM, Struewing JP, Fraser MC, Smith MW, Tucker MA. Prospective risk of cancer in CDKN2A germline mutation carriers. J Med Genet. 2004;41: 421–424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Iqbal J, Ragone A, Lubinski J, et al. The incidence of pancreatic cancer in BRCA1 and BRCA2 mutation carriers. Br J Cancer. 2012;107: 2005–2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Petersen GM. Familial pancreatic cancer. Semin Oncol. 2016;43: 548–553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Giardiello FM, Offerhaus GJ, Lee DH, et al. Increased risk of thyroid and pancreatic carcinoma in familial adenomatous polyposis. Gut. 1993;34: 1394–1396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Kastrinos F, Mukherjee B, Tayob N, et al. Risk of pancreatic cancer in families with Lynch syndrome. JAMA. 2009;302: 1790–1795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Giardiello FM, Brensinger JD, Tersmette AC, et al. Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology. 2000;119: 1447–1453. [DOI] [PubMed] [Google Scholar]
  • 276.Lowenfels AB, Maisonneuve P, DiMagno EP, et al. Hereditary pancreatitis and the risk of pancreatic cancer. International Hereditary Pancreatitis Study Group. J Natl Cancer Inst. 1997;89: 442–446. [DOI] [PubMed] [Google Scholar]
  • 277.Iodice S, Gandini S, Maisonneuve P, Lowenfels AB. Tobacco and the risk of pancreatic cancer: a review and meta-analysis. Langenbecks Arch Surg. 2008;393: 535–545. [DOI] [PubMed] [Google Scholar]
  • 278.Lynch SM, Vrieling A, Lubin JH, et al. Cigarette smoking and pancreatic cancer: a pooled analysis from the pancreatic cancer cohort consortium. Am J Epidemiol. 2009;170: 403–413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Whittemore AS, Paffenbarger RS Jr., Anderson K, Halpern J. Early precursors of pancreatic cancer in college men. J Chronic Dis. 1983;36: 251–256. [DOI] [PubMed] [Google Scholar]
  • 280.Arslan AA, Helzlsouer KJ, Kooperberg C, et al. Anthropometric measures, body mass index, and pancreatic cancer: a pooled analysis from the Pancreatic Cancer Cohort Consortium (PanScan). Arch Intern Med. 2010;170: 791–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 281.Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet. 2008;371: 569–578. [DOI] [PubMed] [Google Scholar]
  • 282.Amundadottir LT, Thorvaldsson S, Gudbjartsson DF, et al. Cancer as a complex phenotype: pattern of cancer distribution within and beyond the nuclear family. PLoS Med. 2004;1: e65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Hemminki K, Li X. Familial and second primary pancreatic cancers: a nationwide epidemiologic study from Sweden. Int J Cancer. 2003;103: 525–530. [DOI] [PubMed] [Google Scholar]
  • 284.Jacobs EJ, Rodriguez C, Newton CC, et al. Family history of various cancers and pancreatic cancer mortality in a large cohort. Cancer Causes Control. 2009;20: 1261–1269. [DOI] [PubMed] [Google Scholar]
  • 285.Raimondi S, Lowenfels AB, Morselli-Labate AM, Maisonneuve P, Pezzilli R. Pancreatic cancer in chronic pancreatitis; aetiology, incidence, and early detection. Best Pract Res Clin Gastroenterol. 2010;24: 349–358. [DOI] [PubMed] [Google Scholar]
  • 286.Andersson G, Wennersten C, Borgquist S, Jirstrom K. Pancreatic cancer risk in relation to sex, lifestyle factors, and pre-diagnostic anthropometry in the Malmo Diet and Cancer Study. Biol Sex Differ. 2016;7: 66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 287.Muscat JE, Stellman SD, Hoffmann D, Wynder EL. Smoking and pancreatic cancer in men and women. Cancer Epidemiol Biomarkers Prev. 1997;6: 15–19. [PubMed] [Google Scholar]
  • 288.Cullinan SA, Moertel CG, Fleming TR, et al. A comparison of three chemotherapeutic regimens in the treatment of advanced pancreatic and gastric carcinoma. Fluorouracil vs fluorouracil and doxorubicin vs fluorouracil, doxorubicin, and mitomycin. JAMA. 1985;253: 2061–2067. [PubMed] [Google Scholar]
  • 289.Tempero M, Plunkett W, Ruiz Van Haperen V, et al. Randomized phase II comparison of dose-intense gemcitabine: thirty-minute infusion and fixed dose rate infusion in patients with pancreatic adenocarcinoma. J Clin Oncol. 2003;21: 3402–3408. [DOI] [PubMed] [Google Scholar]
  • 290.Heinemann V, Quietzsch D, Gieseler F, et al. Randomized Phase III Trial of Gemcitabine Plus Cisplatin Compared With Gemcitabine Alone in Advanced Pancreatic Cancer. Journal of Clinical Oncology. 2006;24: 3946–3952. [DOI] [PubMed] [Google Scholar]
  • 291.Moore MJ, Goldstein D, Hamm J, 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. Journal of Clinical Oncology. 2007;25: 1960–1966. [DOI] [PubMed] [Google Scholar]
  • 292.Cunningham D, Chau I, Stocken DD, et al. Phase III randomized comparison of gemcitabine versus gemcitabine plus capecitabine in patients with advanced pancreatic cancer. J Clin Oncol. 2009;27: 5513–5518. [DOI] [PubMed] [Google Scholar]
  • 293.Kindler HL, Niedzwiecki D, Hollis D, et al. Gemcitabine Plus Bevacizumab Compared With Gemcitabine Plus Placebo in Patients With Advanced Pancreatic Cancer: Phase III Trial of the Cancer and Leukemia Group B (CALGB 80303). Journal of Clinical Oncology. 2010;28: 3617–3622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 294.Philip PA, Benedetti J, Corless CL, et al. Phase III Study Comparing Gemcitabine Plus Cetuximab Versus Gemcitabine in Patients With Advanced Pancreatic Adenocarcinoma: Southwest Oncology Group–Directed Intergroup Trial S0205. Journal of Clinical Oncology. 2010;28: 3605–3610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 295.Neoptolemos JP, Moore MJ, Cox TF, et al. Effect of adjuvant chemotherapy with fluorouracil plus folinic acid or gemcitabine vs observation on survival in patients with resected periampullary adenocarcinoma: the ESPAC-3 periampullary cancer randomized trial. JAMA. 2012;308: 147–156. [DOI] [PubMed] [Google Scholar]
  • 296.Kalser MH, Ellenberg SS. Pancreatic cancer. Adjuvant combined radiation and chemotherapy following curative resection. Arch Surg. 1985;120: 899–903. [DOI] [PubMed] [Google Scholar]
  • 297.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: 776–782; discussion 782–774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Abrams RA, Winter KA, Safran H, et al. Results of the NRG Oncology/RTOG 0848 Adjuvant Chemotherapy Question-Erlotinib+Gemcitabine for Resected Cancer of the Pancreatic Head: A Phase II Randomized Clinical Trial. Am J Clin Oncol. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES