Your activity: 2 p.v.

Epidemiology and nonfamilial risk factors for exocrine pancreatic cancer

Epidemiology and nonfamilial risk factors for exocrine pancreatic cancer
Authors:
Carlos Fernandez-del Castillo, MD
Ramon E Jimenez, MD
Section Editor:
Kenneth K Tanabe, MD
Deputy Editor:
Diane MF Savarese, MD
Literature review current through: Dec 2022. | This topic last updated: Jan 19, 2022.

INTRODUCTION — Surgical resection is the only potentially curative treatment for exocrine pancreatic cancer, but because of the late presentation of the disease, only 15 to 20 percent of patients are candidates for pancreatectomy. Furthermore, the prognosis of pancreatic cancer is poor even in those with potentially resectable disease. The five-year survival following pancreaticoduodenectomy is only approximately 25 to 30 percent for node-negative tumors and 10 percent for node-positive tumors. (See "Overview of surgery in the treatment of exocrine pancreatic cancer and prognosis".)

The nonfamilial risk factors for carcinoma of the exocrine pancreas will be reviewed here. Familial risk factors and screening for pancreatic cancer in high-risk individuals who have a family history of the disease, as well as the pathology, molecular pathogenesis, clinical manifestations, and diagnosis of this disorder are discussed separately. (See "Familial risk factors for pancreatic cancer and screening of high-risk patients" and "Pathology of exocrine pancreatic neoplasms" and "Molecular pathogenesis of exocrine pancreatic cancer" and "Clinical manifestations, diagnosis, and staging of exocrine pancreatic cancer".)

EPIDEMIOLOGY — In the United States, approximately 62,210 patients are diagnosed with cancer of the exocrine pancreas annually, and almost all are expected to die from the disease [1]. Pancreatic cancer is the fourth leading cause of cancer-related death in the United States among both men and women. The majority of these tumors (85 percent) are adenocarcinomas arising from the ductal epithelium. (See "Pathology of exocrine pancreatic neoplasms".)

Worldwide, pancreatic cancer is the seventh leading cause of cancer deaths in both men and women according to data from the World Health Organization (WHO) GLOBOCAN database and data from the 2017 Global Burden of Disease Study [2]. In general, pancreatic cancer affects more individuals inhabiting the Western/industrialized parts of the world; the highest incidence is reported in high-income North America, high-income Asia Pacific, and Western and Central Europe, while people living in South Asia and eastern and central Sub-Saharan Africa have the lowest reported incidence [2].

The disease is rare before the age of 45, but the incidence rises sharply thereafter. In data from the Global Burden of Disease Study, the number of incident cases peaked at age 65 to 69 for men and at 75 to 79 for women [2].

At least in the United States, incidence rates vary by sex and race [3]. The incidence is greater in males than in females (male-to-female ratio 1.3:1) and in Black Americans than in White Americans (16.7 per 100,000 for Black males compared with 14.8 per 100,000 for White males; the corresponding figures for women are 14.6 versus 11.5 cases per 100,000) [4].

The majority of cases are locoregionally advanced, and only 15 to 20 percent are potentially resectable at presentation, usually (but not always) because of vascular invasion. However, at least in the United States, the proportion of patients diagnosed with stage IA pancreatic cancer appears to be increasing over time. In an analysis of data from the National Cancer Institute Surveillance, Epidemiology, and End Results database, the incidence of stage IA pancreatic cancer increased significantly between 2004 and 2016 (annual percentage change 14.5), and their average age at diagnosis decreased by 3.5 years overall [5]. Perhaps more importantly, the five-year overall survival for stage IA pancreatic cancer nearly doubled from 45 percent in 2004 to 84 percent in 2012. These trends were attributed to improved early diagnosis and detection.

Notably, guidelines from the United States Preventive Services Task Force specifically recommend against screening for pancreatic cancer in asymptomatic adults not known to be at high risk because of family history or inherited genetic syndromes [6]. Screening for early detection in high-risk individuals who have familial syndromes predisposing them to pancreatic cancer is discussed elsewhere. (See "Familial risk factors for pancreatic cancer and screening of high-risk patients".)

RISK FACTORS — In the 2017 Global Burden of Disease Study, worldwide, deaths related to pancreatic cancer were primarily attributable to smoking (21 percent), high fasting plasma glucose (8.9 percent), and high body mass index (BMI; 6.2 percent) [2].

Environmental risk factors — Several environmental risk factors have been implicated in the risk of pancreatic cancer, including tobacco use, diet, alcohol consumption, and high caloric intake.

Cigarette smoking — Cigarette smoking increases the risk for pancreatic cancer [7,8], and the estimated population-attributable fraction of pancreatic cancer deaths to tobacco smoking is 11 to 32 percent [9]. In multiple cohort and case-control studies, the relative risk for developing pancreatic cancer among smokers was at least 1.5 [10-17]. The risk increases with the amount of cigarettes consumed [13,14,17], and it may be particularly high in heavy smokers who also have homozygous deletions of the gene for the carcinogen-metabolizing enzyme glutathione S-transferase theta 1 (GSTT1) [16].

Excess risk decreases with smoking cessation. In a large prospective study, the relative risk of pancreatic cancer among current smokers was 2.5 [18]. The risk fell by 48 percent by two years after discontinuing smoking and leveled off 10 to 15 years after stopping, eventually falling to the level of nonsmokers. It has been estimated that cessation of smoking could eliminate approximately 25 percent of pancreatic cancer deaths in the United States [15,18].

Obesity and physical inactivity — Several studies suggest a link between high body mass, lack of physical activity, and pancreatic cancer risk [19-28]. These relationships can be illustrated by the result of a representative cohort study that was based on follow-up data from the Health Professionals Follow-up Study and the Nurses' Health Study [20]. A BMI of at least 30 kg/m2 was associated with a significantly increased risk of pancreatic cancer compared with a BMI of less than 23 kg/m2 (relative risk 1.72, 95% CI 1.19-2.48). Height was also associated with an increased risk (relative risk 1.81, 95% CI 1.31-2.52). An inverse relationship was also observed for moderate physical activity when comparing the highest versus the lowest categories (relative risk 0.45, 95% CI 0.29-0.70), particularly among those with a BMI of at least 25 kg/m2.

Others have suggested that overweight and obese individuals develop pancreatic cancer at a younger age than do patients with a normal weight, and that they also have lower rates and shorter duration of survival once pancreatic cancer is diagnosed [23,29].

Diet — Studies evaluating the relationship between diet and pancreatic cancer are inconclusive, as evidenced by the following observations:

A "Western" dietary pattern (high intake of saturated fat and/or meat, particularly smoked or processed meats) has been linked to the development of pancreatic cancer in many [14,30-32], but not all [33-35], studies.

Several case-control studies [36-38] report a protective effect from the consumption of fresh fruits and vegetables, but prospective studies have not observed such an association [39].

A case-control study from the National Institutes of Health (NIH)-American Association of Retired Persons (AARP) cohort found an inverse association between consumption of a "healthy diet" and risk of pancreatic cancer [40]. Compared with individuals meeting the fewest guidelines for a healthy diet according to the 2005 Dietary Guidelines for Americans (which included higher intake of fruits, vegetables, whole grains, milk, meat and beans, and oils [nonhydrogenated vegetable oils and those found in fish, nuts, and seeds] and lower intake of saturated fat, sodium, and calories from solid fat, alcohol, and added sugar), those who met the most dietary guidelines had a significantly reduced risk for pancreatic cancer (hazard ratio [HR] 0.85, 95% CI 0.74-0.97). Among men, the protective effect of a healthy diet was seen in men who were overweight/obese (BMI ≥25 kg/m2) but not those with a normal weight.

Lower serum levels of lycopene (a carotenoid present in fruits) and selenium have been found in subjects who subsequently developed pancreatic cancer [41]. However, whether dietary or supplemental intake of these nutrients is associated with a reduced risk of pancreatic cancer is unclear [42,43].

Coffee and alcohol consumption — Data regarding the impact of coffee and alcohol ingestion on the risk of pancreatic cancer have been conflicting. Two pooled analyses suggest that, if there is an effect of alcohol consumption, it is small and limited to heavy drinkers [44,45]. The relationship between alcohol use and pancreatic cancer is confounded by cigarette smoking.

Similarly, the relationship between coffee intake and pancreatic cancer risk has been controversial, with some studies indicating an elevated risk with higher levels of consumption, and others indicating no relationship. A meta-analysis of 37 case-control and 17 cohort studies concluded that the pooled relative risk of pancreatic cancer for highest versus lowest intake of coffee was 1.13 (95% CI 0.99-1.29), and it remained not statistically significant when the analysis was limited to those studies that adjusted for smoking [46].

Aspirin and NSAID use — Laboratory data suggest a possible inhibitory effect of aspirin and other nonsteroidal anti-inflammatory drugs (NSAIDs) on pancreatic tumorigenesis. However, the available epidemiologic data in humans are conflicting [47-55]. In fact, preliminary data from the Nurses' Health study, in which 88,378 women without cancer were followed for up to 18 years, raise the possibility that extended regular use of aspirin might increase the risk of pancreatic cancer [52]. Compared with nonusers, participants who ingested 14 or more aspirin tablets weekly (but not fewer) for four or more years had a significantly increased relative risk of pancreatic cancer (relative risk 1.86). The findings did not change appreciably when obesity and smoking were taken into account.

Despite these data, studies of other large cohorts have not found any link between aspirin use and higher pancreatic cancer risk, even among individuals using aspirin ≥30 times monthly for over 20 years [53].

Helicobacter pylori — A weak but statistically significant association between Helicobacter pylori infection and pancreatic cancer has been reported. (See "Association between Helicobacter pylori infection and gastrointestinal malignancy", section on 'Pancreatic cancer'.)

HBV and HCV infection — Several reports indicate an association between infection with hepatitis B virus (HBV) and hepatitis C virus (HCV), and pancreatic cancer [56-58]. The magnitude of risk is less than that for hepatocellular cancer. (See "Epidemiology and risk factors for hepatocellular carcinoma", section on 'Hepatitis B virus' and "Epidemiology and risk factors for hepatocellular carcinoma", section on 'Hepatitis C virus'.)

Diabetes mellitus, glucose metabolism, and insulin resistance — Numerous epidemiologic studies describe an association between diabetes mellitus and pancreatic cancer [59-67]. In a meta-analysis of 88 studies (50 cohort and 30 case-control studies, predominantly of patients with type 2 diabetes), the pooled relative risk for pancreatic cancer in diabetics compared with patients without diabetes was 2.08 (95% CI 1.87-2.32) [67]. Patients with diabetes may also have unfavorable pathologic features and poorer long-term outcomes compared with those without diabetes [68].

It has been suggested that diabetes may be a consequence rather than a cause of pancreatic cancer [69-71]. As an example, one study compared 512 patients with newly diagnosed pancreatic cancer with 933 controls of a similar age [71]. Compared with the controls, diabetes was more prevalent in pancreatic cancer cases (47 versus 7 percent) and was more likely to have been diagnosed in the preceding two years (74 versus 53 percent). After pancreaticoduodenectomy, diabetes resolved in 17 of 30 patients (57 percent) with new-onset diabetes, while its prevalence was unchanged in the 11 patients who had longstanding diabetes. One intriguing possibility derived from in vitro work is that pancreatic cancers might induce paraneoplastic beta cell dysfunction and inhibition of insulin secretion by shedding exosomes containing adrenomedullin (a vasodilator peptide hormone that regulates insulin balance and may participate in the development of diabetes [72]) into the systemic circulation [73]. An observational study is underway in Italy examining the utility of adrenomedullin measurement to see if it may identify patients with adult-onset diabetes who are at risk for pancreatic cancer.

However, increasing data support the view that abnormal glucose metabolism, insulin resistance, and hyperinsulinemia are etiologic factors for pancreatic cancer rather than the result of a subclinical cancer [19,74-78]. As examples:

In a prospective cohort study, employees of 84 Chicago-area businesses were screened for postload plasma glucose levels and followed for an average of 25 years [74]. Compared with controls with a postload plasma glucose of 119 mg/dL (6.6 mmol/L) or less, the relative risk of mortality from pancreatic cancer was 1.65, 1.6, and 2.15 for those with levels of 120 to 159 mg/dL (6.7 to 8.8 mmol/L), 160 to 199 mg/dL (8.9 to 11.1 mmol/L), and ≥200 mg/dL (11.1 mmol/L), respectively, even after adjusting for age, race, cigarette smoking, and BMI.

An association between prediagnosis serum levels of glucose and insulin, insulin resistance, and pancreatic cancer risk was suggested in a case-cohort study within the Alpha-Tocopherol, Beta-Carotene Cancer Prevention Study, a primary prevention trial enrolling 29,133 male Finnish smokers aged 50 to 69 [75]. Fasting serum concentrations of glucose and insulin, and levels of insulin resistance were assessed. The interval between serum collection and follow-up was up to 16.7 years. The study included 169 incident cases of pancreatic cancer that were diagnosed after the fifth year of follow-up, and 400 randomly selected controls.

After adjustment for age, years of smoking, and BMI, higher prediagnosis serum concentrations of glucose and insulin, as well as insulin resistance were significantly correlated with the risk of pancreatic cancer. The positive associations were stronger among the cases that occurred more than 10 years after baseline (highest versus lowest quartile for glucose, HR 2.16, 95% CI 1.05-4.42; for insulin, HR 2.90, 95% CI 1.22-6.92; for insulin resistance, HR 2.71, 95% CI 1.19-6.18). The prospective study design, with a minimum five-year follow-up prior to the detection of incident pancreatic cancer, minimizes the possibility that the identified insulin and glucose abnormalities resulted from "subclinical" pancreatic cancer.

On the other hand, an analysis derived from the Nurses' Health Study and the Health Professionals Follow-up Study examining the temporal pattern of cancer risk after a first diagnosis of type 2 diabetes mellitus determined that risk for pancreatic cancer was elevated within the first eight years after diagnosis, but decreased thereafter [78]. A central role for hyperinsulinemia rather than hyperglycemia was suggested by analysis of a subset of subjects in whom blood was available, which showed that levels of C-peptide (a marker of endogenous insulin secretion) fell beyond eight years after diagnosis, while hemoglobin A1C (a marker of glucose levels over the preceding three months) was relatively stable out to 15 years postdiagnosis.

The mechanism underlying this association is unclear. However, at least some data suggest that the increased risk of pancreatic cancer in patients with metabolic diseases, such as type 2 diabetes mellitus and other states of insulin resistance, as well as obesity may be mediated by reduced levels of plasma adiponectin, a fat-derived hormone that has insulin-sensitizing and anti-inflammatory properties [79].

The data linking type 2 diabetes with an elevated risk of pancreatic cancer have led some to suggest that the new onset of diabetes in a thin older adult should prompt consideration of screening for early diagnosis of a potentially resectable pancreatic cancer. At least three studies have addressed the utility of computed tomography (CT) screening for early detection of pancreatic cancer in adults with new-onset diabetes. Two uncovered mainly unresectable tumors, but they selected patients for screening based on the presence of cancer-related symptoms. A third series from the Mayo Clinic suggested that CT scans done at the time of newly diagnosed diabetes in otherwise asymptomatic patients were more likely to show potentially resectable tumors than scans performed six months later [80]. Whether higher resectability rates translated into higher cure rates was not addressed.

CT screening of all older subjects with new-onset diabetes in order to discover a small number of pancreatic cancers is not feasible. Identification of those features that differentiate pancreatic cancer-associated diabetes from other cases of new-onset diabetes would help direct efforts to the subset of individuals who would most benefit from screening CT, but these factors have not yet been established. At least some data suggest that the risk of pancreatic cancer is especially elevated in older adults with new-onset diabetes and a previously healthy weight with otherwise unexplained unintentional weight loss [81], but whether these patients have a high enough risk of pancreatic cancer to justify screening is not established.

Thus, screening for pancreatic cancer is not warranted in older, otherwise asymptomatic adults with new-onset atypical diabetes. This recommendation is consistent with guidelines from the United States Preventive Services Task Force, which specifically recommend against screening for pancreatic cancer in asymptomatic adults not known to be at high risk because of family history or inherited genetic syndromes, including those with preexisting or new-onset diabetes [6]. Screening for high-risk individuals who have familial syndromes predisposing them to pancreatic cancer is discussed elsewhere. (See "Familial risk factors for pancreatic cancer and screening of high-risk patients", section on 'Pancreatic cancer screening'.)

Hereditary risk factors

Family history and genetic predisposition syndromes — Pancreatic cancer aggregates in some families; approximately 5 to 10 percent of individuals with pancreatic cancer have a family history of the disease [82,83]. There are two broad categories of hereditary risk for pancreatic cancer: defined syndromes in which patients are at risk for a pancreatic cancer (and frequently for other malignancies), and familial pancreatic cancer, for which a specific molecular basis has not been identified. These syndromes and the approximate risk for pancreatic cancer are outlined in the table (table 1). A separate topic is available that discusses these syndromes and screening for pancreatic cancer in high-risk individuals. (See "Familial risk factors for pancreatic cancer and screening of high-risk patients".)

At least some data suggest that germline mutations in pancreatic cancer susceptibility genes are not rare, even in the absence of a significant family history of pancreatic cancer [84-86]. In the largest analysis, of 854 patients undergoing pancreatic cancer resection at a single institution over a 15-year period, 33 were identified as having a potentially deleterious germline mutation, 31 of which affected a known pancreatic cancer susceptibility gene (including BRCA, ATM, PALB2, CDKN2A, and MLH1) [84].

Routine genetic testing of patients with newly diagnosed, apparently sporadic pancreatic cancer may yield some clinical benefits (eg, identifying mutation carriers for whom specific forms of enhanced cancer screening are recommended to reduce the risk of associated cancers, identifying family members of the index case who might benefit from screening for the cancer-predisposing mutation, identifying cancer susceptibility mutations that might be therapeutically targetable). As an example, the POLO trial demonstrated benefit from maintenance therapy with the poly(ADP-ribose) polymerase (PARP) inhibitor olaparib in patients with advanced BRCA-mutated pancreatic cancer. (See "Initial systemic chemotherapy for metastatic exocrine pancreatic cancer", section on 'Patients with homologous recombination repair deficiency'.)

A year 2019 Provisional Clinical Opinion from the American Society of Clinical Oncology (ASCO) states that germline genetic testing may be offered to patients with pancreatic cancer and an unremarkable family history if an informative result could directly benefit the patient or his or her family members [87]. This practice is also supported by updated year 2019 consensus-based guidelines from the National Comprehensive Cancer Network (NCCN) [88]. (See "Familial risk factors for pancreatic cancer and screening of high-risk patients", section on 'Genetic predisposition syndromes'.)

Other inherited risk factors

ABO blood group — ABO blood type is an inherited characteristic that has been associated with the risk of several gastrointestinal malignancies, including pancreatic cancer [89-92]. Blood type is determined by the presence or absence of human blood group antigens, glycoproteins that are expressed on the surface of red blood cells and several other types of cells, including pancreatic cancer cells.

The relationship between blood type and pancreatic cancer risk was examined in two large independent prospective cohorts (the Nurses' Health Study and the Health Professionals Follow-up Study) [89]. Compared with blood group O, individuals with non-O blood group (type A, AB, or B) were significantly more likely to develop pancreatic cancer (adjusted HR for incident pancreatic cancer 1.32, 1.51, and 1.72, respectively), and the association was nearly identical in the two cohorts. Overall, 17 percent of the 316 pancreatic cancer cases were attributable to inheriting a non-O blood type.

These findings are supported by the results of a genome-wide association study that identified variants in the ABO locus that were associated with susceptibility to pancreatic cancer [93].

While these data suggest that ABO blood group status may represent a common inherited susceptibility for pancreatic cancer, additional studies are needed to confirm these findings and establish the potential mechanisms by which ABO blood group influences the risk of pancreatic cancer. An interaction between non-O blood type and colonization with H. pylori has been suggested [91]. (See 'Helicobacter pylori' above.)

Cystic fibrosis — Patients with cystic fibrosis (most of whom have inherited the predisposing gene in an autosomal recessive pattern) have an increased risk of pancreatic cancer. In a meta-analysis, the standardized incidence ratio was 6.18, 95% CI 1.31-29.27, and it was two- to fivefold higher in patients who had undergone lung transplantation [94]. (See "Cystic fibrosis: Overview of gastrointestinal disease", section on 'Gastrointestinal cancer'.)

Nonhereditary chronic pancreatitis — Chronic inflammation of the pancreas is a risk factor for pancreatic cancer [95-98]. In a report from the International Pancreatitis Study Group, for example, 2015 patients with chronic pancreatitis were followed for a mean of 7.4 years, during which a total of 56 pancreatic cancers were identified [95]. The expected number of cases of cancer calculated from country-specific incidence data and adjusted for age and sex was 2.13, yielding a standardized incidence ratio (the ratio of observed to expected cases) of 26.3. The cumulative risk reached 1.8 percent at 10 years and 4 percent at 20 years, independent of the type of pancreatitis. A less pronounced relationship has been noted by others [96]. In any event, the population-attributable risk has been estimated to be 1.3 percent, suggesting that a relatively small proportion of pancreatic cancers might be avoided if pancreatitis could be prevented [97]. (See "Chronic pancreatitis: Clinical manifestations and diagnosis in adults".)

The signaling mechanisms that underlie the transition from chronic pancreatitis to invasive cancer are largely undefined, but they are beginning to be elucidated [99].

Pancreatic cysts — Patients with intraductal papillary mucinous neoplasm of the pancreas (IPMN), which is the most common type of neoplastic pancreatic cyst, are at risk for malignant degeneration and are commonly managed with surveillance. When an IPMN develops invasive malignancy, it is usually referred to as IPMN-associated adenocarcinoma. These patients, however, are also at risk of developing conventional pancreatic cancer (concurrent or distinct ductal adenocarcinoma), which arises away from the cyst, suggesting the existence of a pancreatic field defect. This phenomenon has been described in 2 to 9 percent of patients who are being followed for IPMN [100,101]. An in-depth discussion of IPMNs, including a recommended surveillance strategy, is discussed in detail elsewhere. (See "Intraductal papillary mucinous neoplasm of the pancreas (IPMN): Pathophysiology and clinical manifestations" and "Intraductal papillary mucinous neoplasm of the pancreas (IPMN): Evaluation and management".)

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Pancreatic cancer".)

SUMMARY — The major risk factors for pancreatic cancer are the following:

Cigarette smoking. (See 'Cigarette smoking' above.)

High body mass and lack of physical activity. (See 'Obesity and physical inactivity' above.)

Numerous epidemiologic studies describe an association between diabetes mellitus and pancreatic cancer, although it is not clear whether abnormal glucose metabolism is a consequence or cause of pancreatic cancer. (See 'Diabetes mellitus, glucose metabolism, and insulin resistance' above.)

Hereditary risk factors, such as hereditary pancreatitis, other highly penetrant conditions caused by germline mutations in known cancer-causing genes, and familial pancreatic cancer, for which a specific genetic abnormality has not yet been identified (table 1). (See "Familial risk factors for pancreatic cancer and screening of high-risk patients".)

Nonhereditary chronic pancreatitis. (See 'Nonhereditary chronic pancreatitis' above.)

Pancreatic cysts. (See 'Pancreatic cysts' above.)

  1. Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2022. CA Cancer J Clin 2022; 72:7.
  2. GBD 2017 Pancreatic Cancer Collaborators. The global, regional, and national burden of pancreatic cancer and its attributable risk factors in 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol 2019; 4:934.
  3. Zhang J, Dhakal I, Ning B, Kesteloot H. Patterns and trends of pancreatic cancer mortality rates in Arkansas, 1969-2002: a comparison with the US population. Eur J Cancer Prev 2008; 17:18.
  4. Howlader N, Noone AM, Krapcho M, Miller D, Brest A, Yu M, Ruhl J, Tatalovich Z, Mariotto A, Lewis DR, Chen HS, Feuer EJ, Cronin KA (eds). SEER Cancer Statistics Review, 1975-2016, National Cancer Institute. Bethesda, MD, https://seer.cancer.gov/csr/1975_2016/, based on November 2018 SEER data submission, posted to the SEER web site, April 2019. https://seer.cancer.gov/csr/1975_2017/ (Accessed on May 01, 2020).
  5. Blackford AL, Canto MI, Klein AP, et al. Recent Trends in the Incidence and Survival of Stage 1A Pancreatic Cancer: A Surveillance, Epidemiology, and End Results Analysis. J Natl Cancer Inst 2020; 112:1162.
  6. 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.
  7. Rebours V, Lévy P, Ruszniewski P. An overview of hereditary pancreatitis. Dig Liver Dis 2012; 44:8.
  8. 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.
  9. Maisonneuve P, Lowenfels AB. Risk factors for pancreatic cancer: a summary review of meta-analytical studies. Int J Epidemiol 2015; 44:186.
  10. Mack TM, Yu MC, Hanisch R, Henderson BE. Pancreas cancer and smoking, beverage consumption, and past medical history. J Natl Cancer Inst 1986; 76:49.
  11. Farrow DC, Davis S. Risk of pancreatic cancer in relation to medical history and the use of tobacco, alcohol and coffee. Int J Cancer 1990; 45:816.
  12. Ghadirian P, Simard A, Baillargeon J. Tobacco, alcohol, and coffee and cancer of the pancreas. A population-based, case-control study in Quebec, Canada. Cancer 1991; 67:2664.
  13. 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.
  14. Nöthlings U, Wilkens LR, Murphy SP, et al. Meat and fat intake as risk factors for pancreatic cancer: the multiethnic cohort study. J Natl Cancer Inst 2005; 97:1458.
  15. Silverman DT, Dunn JA, Hoover RN, et al. Cigarette smoking and pancreas cancer: a case-control study based on direct interviews. J Natl Cancer Inst 1994; 86:1510.
  16. Duell EJ, Holly EA, Bracci PM, et al. A population-based, case-control study of polymorphisms in carcinogen-metabolizing genes, smoking, and pancreatic adenocarcinoma risk. J Natl Cancer Inst 2002; 94:297.
  17. Bosetti C, Lucenteforte E, Silverman DT, et al. Cigarette smoking and pancreatic cancer: an analysis from the International Pancreatic Cancer Case-Control Consortium (Panc4). Ann Oncol 2012; 23:1880.
  18. Fuchs CS, Colditz GA, Stampfer MJ, et al. A prospective study of cigarette smoking and the risk of pancreatic cancer. Arch Intern Med 1996; 156:2255.
  19. Carreras-Torres R, Johansson M, Gaborieau V, et al. The Role of Obesity, Type 2 Diabetes, and Metabolic Factors in Pancreatic Cancer: A Mendelian Randomization Study. J Natl Cancer Inst 2017; 109.
  20. Michaud DS, Giovannucci E, Willett WC, et al. Physical activity, obesity, height, and the risk of pancreatic cancer. JAMA 2001; 286:921.
  21. Stolzenberg-Solomon RZ, Adams K, Leitzmann M, et al. Adiposity, physical activity, and pancreatic cancer in the National Institutes of Health-AARP Diet and Health Cohort. Am J Epidemiol 2008; 167:586.
  22. Luo J, Margolis KL, Adami HO, et al. Obesity and risk of pancreatic cancer among postmenopausal women: the Women's Health Initiative (United States). Br J Cancer 2008; 99:527.
  23. Li D, Morris JS, Liu J, et al. Body mass index and risk, age of onset, and survival in patients with pancreatic cancer. JAMA 2009; 301:2553.
  24. O'Rorke MA, Cantwell MM, Cardwell CR, et al. Can physical activity modulate pancreatic cancer risk? a systematic review and meta-analysis. Int J Cancer 2010; 126:2957.
  25. 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.
  26. Aune D, Greenwood DC, Chan DS, et al. Body mass index, abdominal fatness and pancreatic cancer risk: a systematic review and non-linear dose-response meta-analysis of prospective studies. Ann Oncol 2012; 23:843.
  27. Lauby-Secretan B, Scoccianti C, Loomis D, et al. Body Fatness and Cancer--Viewpoint of the IARC Working Group. N Engl J Med 2016; 375:794.
  28. Jiao L, Berrington de Gonzalez A, Hartge P, et al. Body mass index, effect modifiers, and risk of pancreatic cancer: a pooled study of seven prospective cohorts. Cancer Causes Control 2010; 21:1305.
  29. Yuan C, Bao Y, Wu C, et al. Prediagnostic body mass index and pancreatic cancer survival. J Clin Oncol 2013; 31:4229.
  30. Silverman DT, Swanson CA, Gridley G, et al. Dietary and nutritional factors and pancreatic cancer: a case-control study based on direct interviews. J Natl Cancer Inst 1998; 90:1710.
  31. Thiébaut AC, Jiao L, Silverman DT, et al. Dietary fatty acids and pancreatic cancer in the NIH-AARP diet and health study. J Natl Cancer Inst 2009; 101:1001.
  32. Larsson SC, Wolk A. Red and processed meat consumption and risk of pancreatic cancer: meta-analysis of prospective studies. Br J Cancer 2012; 106:603.
  33. Michaud DS, Giovannucci E, Willett WC, et al. Dietary meat, dairy products, fat, and cholesterol and pancreatic cancer risk in a prospective study. Am J Epidemiol 2003; 157:1115.
  34. Michaud DS, Skinner HG, Wu K, et al. Dietary patterns and pancreatic cancer risk in men and women. J Natl Cancer Inst 2005; 97:518.
  35. Rohrmann S, Linseisen J, Nöthlings U, et al. Meat and fish consumption and risk of pancreatic cancer: results from the European Prospective Investigation into Cancer and Nutrition. Int J Cancer 2013; 132:617.
  36. Norell SE, Ahlbom A, Erwald R, et al. Diet and pancreatic cancer: a case-control study. Am J Epidemiol 1986; 124:894.
  37. Howe GR, Jain M, Miller AB. Dietary factors and risk of pancreatic cancer: results of a Canadian population-based case-control study. Int J Cancer 1990; 45:604.
  38. Gold EB, Gordis L, Diener MD, et al. Diet and other risk factors for cancer of the pancreas. Cancer 1985; 55:460.
  39. Coughlin SS, Calle EE, Patel AV, Thun MJ. Predictors of pancreatic cancer mortality among a large cohort of United States adults. Cancer Causes Control 2000; 11:915.
  40. Arem H, Reedy J, Sampson J, et al. The Healthy Eating Index 2005 and risk for pancreatic cancer in the NIH-AARP study. J Natl Cancer Inst 2013; 105:1298.
  41. Burney PG, Comstock GW, Morris JS. Serologic precursors of cancer: serum micronutrients and the subsequent risk of pancreatic cancer. Am J Clin Nutr 1989; 49:895.
  42. Han X, Li J, Brasky TM, et al. Antioxidant intake and pancreatic cancer risk: the Vitamins and Lifestyle (VITAL) Study. Cancer 2013; 119:1314.
  43. Nkondjock A, Ghadirian P, Johnson KC, et al. Dietary intake of lycopene is associated with reduced pancreatic cancer risk. J Nutr 2005; 135:592.
  44. Michaud DS, Vrieling A, Jiao L, et al. Alcohol intake and pancreatic cancer: a pooled analysis from the pancreatic cancer cohort consortium (PanScan). Cancer Causes Control 2010; 21:1213.
  45. Lucenteforte E, La Vecchia C, Silverman D, et al. Alcohol consumption and pancreatic cancer: a pooled analysis in the International Pancreatic Cancer Case-Control Consortium (PanC4). Ann Oncol 2012; 23:374.
  46. Turati F, Galeone C, Edefonti V, et al. A meta-analysis of coffee consumption and pancreatic cancer. Ann Oncol 2012; 23:311.
  47. Anderson KE, Johnson TW, Lazovich D, Folsom AR. Association between nonsteroidal anti-inflammatory drug use and the incidence of pancreatic cancer. J Natl Cancer Inst 2002; 94:1168.
  48. Bradley MC, Hughes CM, Cantwell MM, et al. Non-steroidal anti-inflammatory drugs and pancreatic cancer risk: a nested case-control study. Br J Cancer 2010; 102:1415.
  49. Langman MJ, Cheng KK, Gilman EA, Lancashire RJ. Effect of anti-inflammatory drugs on overall risk of common cancer: case-control study in general practice research database. BMJ 2000; 320:1642.
  50. Schreinemachers DM, Everson RB. Aspirin use and lung, colon, and breast cancer incidence in a prospective study. Epidemiology 1994; 5:138.
  51. Gridley G, McLaughlin JK, Ekbom A, et al. Incidence of cancer among patients with rheumatoid arthritis. J Natl Cancer Inst 1993; 85:307.
  52. Schernhammer ES, Kang JH, Chan AT, et al. A prospective study of aspirin use and the risk of pancreatic cancer in women. J Natl Cancer Inst 2004; 96:22.
  53. Jacobs EJ, Connell CJ, Rodriguez C, et al. Aspirin use and pancreatic cancer mortality in a large United States cohort. J Natl Cancer Inst 2004; 96:524.
  54. Cook NR, Lee IM, Gaziano JM, et al. Low-dose aspirin in the primary prevention of cancer: the Women's Health Study: a randomized controlled trial. JAMA 2005; 294:47.
  55. Streicher SA, Yu H, Lu L, et al. Case-control study of aspirin use and risk of pancreatic cancer. Cancer Epidemiol Biomarkers Prev 2014; 23:1254.
  56. Hassan MM, Li D, El-Deeb AS, et al. Association between hepatitis B virus and pancreatic cancer. J Clin Oncol 2008; 26:4557.
  57. Mahale P, Torres HA, Kramer JR, et al. Hepatitis C virus infection and the risk of cancer among elderly US adults: A registry-based case-control study. Cancer 2017; 123:1202.
  58. Huang J, Magnusson M, Törner A, et al. Risk of pancreatic cancer among individuals with hepatitis C or hepatitis B virus infection: a nationwide study in Sweden. Br J Cancer 2013; 109:2917.
  59. Huxley R, Ansary-Moghaddam A, Berrington de González A, et al. Type-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer 2005; 92:2076.
  60. Inoue M, Iwasaki M, Otani T, et al. Diabetes mellitus and the risk of cancer: results from a large-scale population-based cohort study in Japan. Arch Intern Med 2006; 166:1871.
  61. Chow WH, Gridley G, Nyrén O, et al. Risk of pancreatic cancer following diabetes mellitus: a nationwide cohort study in Sweden. J Natl Cancer Inst 1995; 87:930.
  62. Calle EE, Murphy TK, Rodriguez C, et al. Diabetes mellitus and pancreatic cancer mortality in a prospective cohort of United States adults. Cancer Causes Control 1998; 9:403.
  63. Wideroff L, Gridley G, Mellemkjaer L, et al. Cancer incidence in a population-based cohort of patients hospitalized with diabetes mellitus in Denmark. J Natl Cancer Inst 1997; 89:1360.
  64. Jee SH, Ohrr H, Sull JW, et al. Fasting serum glucose level and cancer risk in Korean men and women. JAMA 2005; 293:194.
  65. Aggarwal G, Kamada P, Chari ST. Prevalence of diabetes mellitus in pancreatic cancer compared to common cancers. Pancreas 2013; 42:198.
  66. Bosetti C, Rosato V, Li D, et al. Diabetes, antidiabetic medications, and pancreatic cancer risk: an analysis from the International Pancreatic Cancer Case-Control Consortium. Ann Oncol 2014; 25:2065.
  67. Batabyal P, Vander Hoorn S, Christophi C, Nikfarjam M. Association of diabetes mellitus and pancreatic adenocarcinoma: a meta-analysis of 88 studies. Ann Surg Oncol 2014; 21:2453.
  68. Hank T, Sandini M, Qadan M, et al. Diabetes mellitus is associated with unfavorable pathologic features, increased postoperative mortality, and worse long-term survival in resected pancreatic cancer. Pancreatology 2020; 20:125.
  69. Gullo L, Pezzilli R, Morselli-Labate AM, Italian Pancreatic Cancer Study Group. Diabetes and the risk of pancreatic cancer. N Engl J Med 1994; 331:81.
  70. Chari ST, Leibson CL, Rabe KG, et al. Pancreatic cancer-associated diabetes mellitus: prevalence and temporal association with diagnosis of cancer. Gastroenterology 2008; 134:95.
  71. Pannala R, Leirness JB, Bamlet WR, et al. Prevalence and clinical profile of pancreatic cancer-associated diabetes mellitus. Gastroenterology 2008; 134:981.
  72. Wong HK, Tang F, Cheung TT, Cheung BM. Adrenomedullin and diabetes. World J Diabetes 2014; 5:364.
  73. Javeed N, Sagar G, Dutta SK, et al. Pancreatic Cancer-Derived Exosomes Cause Paraneoplastic β-cell Dysfunction. Clin Cancer Res 2015; 21:1722.
  74. Gapstur SM, Gann PH, Lowe W, et al. Abnormal glucose metabolism and pancreatic cancer mortality. JAMA 2000; 283:2552.
  75. Stolzenberg-Solomon RZ, Graubard BI, Chari S, et al. Insulin, glucose, insulin resistance, and pancreatic cancer in male smokers. JAMA 2005; 294:2872.
  76. Michaud DS, Liu S, Giovannucci E, et al. Dietary sugar, glycemic load, and pancreatic cancer risk in a prospective study. J Natl Cancer Inst 2002; 94:1293.
  77. Wolpin BM, Bao Y, Qian ZR, et al. Hyperglycemia, insulin resistance, impaired pancreatic β-cell function, and risk of pancreatic cancer. J Natl Cancer Inst 2013; 105:1027.
  78. Hu Y, Zhang X, Ma Y, et al. Incident type 2 diabetes duration and cancer risk: A prospective study in two US cohorts. J Natl Cancer Inst 2020.
  79. Bao Y, Giovannucci EL, Kraft P, et al. A prospective study of plasma adiponectin and pancreatic cancer risk in five US cohorts. J Natl Cancer Inst 2013; 105:95.
  80. Pelaez-Luna M, Takahashi N, Fletcher JG, Chari ST. Resectability of presymptomatic pancreatic cancer and its relationship to onset of diabetes: a retrospective review of CT scans and fasting glucose values prior to diagnosis. Am J Gastroenterol 2007; 102:2157.
  81. Yuan C, Babic A, Khalaf N, et al. Diabetes, Weight Change, and Pancreatic Cancer Risk. JAMA Oncol 2020; 6:e202948.
  82. Klein AP, Hruban RH, Brune KA, et al. Familial pancreatic cancer. Cancer J 2001; 7:266.
  83. Olson SH, Kurtz RC. Epidemiology of pancreatic cancer and the role of family history. J Surg Oncol 2013; 107:1.
  84. Shindo K, Yu J, Suenaga M, et al. Deleterious Germline Mutations in Patients With Apparently Sporadic Pancreatic Adenocarcinoma. J Clin Oncol 2017; 35:3382.
  85. Grant RC, Selander I, Connor AA, et al. Prevalence of germline mutations in cancer predisposition genes in patients with pancreatic cancer. Gastroenterology 2015; 148:556.
  86. Peters MLB, Brand R, Borazanci EH, et al. Germline genetic testing in unselected pancreatic ductal adenocarcinoma (PDAC) patients. J Clin Oncol 1017; 35S:ASCO #1501.
  87. Stoffel EM, McKernin SE, Brand R, et al. Evaluating Susceptibility to Pancreatic Cancer: ASCO Provisional Clinical Opinion. J Clin Oncol 2019; 37:153.
  88. National Comprehensive Cancer Network (NCCN) guidelines available online at https://www.nccn.org/professionals/physician_gls/default.aspx (Accessed on May 24, 2021).
  89. Wolpin BM, Chan AT, Hartge P, et al. ABO blood group and the risk of pancreatic cancer. J Natl Cancer Inst 2009; 101:424.
  90. Annese V, Minervini M, Gabbrielli A, et al. ABO blood groups and cancer of the pancreas. Int J Pancreatol 1990; 6:81.
  91. Risch HA, Yu H, Lu L, Kidd MS. ABO blood group, Helicobacter pylori seropositivity, and risk of pancreatic cancer: a case-control study. J Natl Cancer Inst 2010; 102:502.
  92. 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.
  93. Amundadottir L, Kraft P, Stolzenberg-Solomon RZ, et al. Genome-wide association study identifies variants in the ABO locus associated with susceptibility to pancreatic cancer. Nat Genet 2009; 41:986.
  94. Yamada A, Komaki Y, Komaki F, et al. Risk of gastrointestinal cancers in patients with cystic fibrosis: a systematic review and meta-analysis. Lancet Oncol 2018; 19:758.
  95. 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.
  96. Ekbom A, McLaughlin JK, Karlsson BM, et al. Pancreatitis and pancreatic cancer: a population-based study. J Natl Cancer Inst 1994; 86:625.
  97. Duell EJ, Lucenteforte E, Olson SH, et al. Pancreatitis and pancreatic cancer risk: a pooled analysis in the International Pancreatic Cancer Case-Control Consortium (PanC4). Ann Oncol 2012; 23:2964.
  98. Bang UC, Benfield T, Hyldstrup L, et al. Mortality, cancer, and comorbidities associated with chronic pancreatitis: a Danish nationwide matched-cohort study. Gastroenterology 2014; 146:989.
  99. Liou GY, Döppler H, Necela B, et al. Macrophage-secreted cytokines drive pancreatic acinar-to-ductal metaplasia through NF-κB and MMPs. J Cell Biol 2013; 202:563.
  100. Tanaka M. Controversies in the management of pancreatic IPMN. Nat Rev Gastroenterol Hepatol 2011; 8:56.
  101. Pergolini I, Sahora K, Ferrone CR, et al. Long-term Risk of Pancreatic Malignancy in Patients With Branch Duct Intraductal Papillary Mucinous Neoplasm in a Referral Center. Gastroenterology 2017; 153:1284.
Topic 2480 Version 66.0

References