Next Article in Journal
Myeloid Cells in Myocardial Ischemic Injury: The Role of the Macrophage Migration Inhibitory Factor
Previous Article in Journal
Developing Theoretical Models for Atherosclerotic Lesions: A Methodological Approach Using Interdisciplinary Insights
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pancreatic Cancer: A Review of Risk Factors

by
Raluca Roxana Grigorescu
1,*,
Ioana Alexandra Husar-Sburlan
2,* and
Cristian Gheorghe
3
1
Gastroenterology Department, “Sfanta Maria” Hospital, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
2
Gastroenterology Department, “Sfanta Maria” Hospital, 011172 Bucharest, Romania
3
Center for Digestive Disease and Liver Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
*
Authors to whom correspondence should be addressed.
Life 2024, 14(8), 980; https://doi.org/10.3390/life14080980
Submission received: 13 June 2024 / Revised: 28 July 2024 / Accepted: 1 August 2024 / Published: 5 August 2024
(This article belongs to the Special Issue Pancreatic Cancer Long Survivals)

Abstract

:
Pancreatic adenocarcinoma is one of the most lethal types of gastrointestinal cancer despite the latest medical advances. Its incidence has continuously increased in recent years in developed countries. The location of the pancreas can result in the initial symptoms of neoplasia being overlooked, which can lead to a delayed diagnosis and a subsequent reduction in the spectrum of available therapeutic options. The role of modifiable risk factors in pancreatic cancer has been extensively studied in recent years, with smoking and alcohol consumption identified as key contributors. However, the few screening programs that have been developed focus exclusively on genetic factors, without considering the potential impact of modifiable factors on disease occurrence. Thus, fully understanding and detecting the risk factors for pancreatic cancer represents an important step in the prevention and early diagnosis of this type of neoplasia. This review reports the available evidence on different risk factors and identifies the areas that could benefit the most from additional studies.

1. Introduction

The vast majority of pancreatic cancers arise from the exocrine component and are classified as pancreatic adenocarcinoma, referred to in the text as pancreatic cancer (PC). It should be noted that there are other, less common histological subtypes, including invasive intraductal papillary mucinous neoplasm (IPMN), adenosquamous carcinoma, invasive mucinous cystic neoplasm (MCN), acinar cell and squamous cell carcinoma, and invasive solid pseudopapillary tumor. In less than 10% of cases, the condition can develop from endocrine cells and form neuroendocrine tumors, which have a more favorable prognosis [1]. PC has the worst prognosis and a very high mortality [2], with almost as many deaths as new cases per year. The delay in diagnosis is often due to its non-specific and insidious symptoms, such as middle epigastric pain that sometimes radiates to the back, weight loss, nausea, fatigue, and, if the location of the tumor is in the pancreatic head, jaundice. The lifetime risk of developing this neoplasia is 0.91–1.6% [2,3,4]. The incidence has doubled in the last two decades and is up to four times higher in countries with a higher Human Development Index [5,6]. Less than 20% of patients have a surgically resectable tumor at diagnosis, with a median survival of 12.6 months. In contrast, for those for whom surgery is no longer an option, the median survival rate is 3.5 months [7]. Incidentally discovered PC is associated with longer survival than the one diagnosed due to signs and symptoms [8].
In the last years, according to data indicated by the Surveillance, Epidemiology, and End Results (SEER) program, the incidence of pancreatic cancer represented 3.2% of all cancer cases, with an estimated mortality of 8.2% of all deaths determined by any cancer [3]. It was estimated that by the year 2030, PC incidence will increase to 15.1 per 100,000 [9] and become the second leading cause of cancer-related death [10], and the incidence will further increase by an average of 1.1% per year, up to 18.6% in 2050 [9].
It has been suggested that the sequence in sporadic pancreatic carcinoma is characterized by the change in the normal duct epithelium to ductal hyperplasia and invasive ductal adenocarcinoma. During this period, a sequence of genetic alterations may occur: first in KRAS (Kirsten-rat sarcoma) and Her-2, followed by p-16 alteration, and finally, in p53, DPC4 and BRCA2, and other tumor suppressor genes [11,12]. The presence of KRAS mutation in pancreatic juice has been frequently observed in patients with pancreatic cancer (80%) and also in patients with chronic pancreatitis (CP) before the development of PC [13,14,15,16], suggesting that it may be useful in identifying CP patients who are at a higher risk of developing cancer. On the contrary, studies on the presence of KRAS mutation performed on stools and serum samples found that it can also be present in cases of benign pancreatic diseases, thus compromising its utility as a screening tool [15,17].
The aim of this study is to review the current literature on the role of several modifiable and non-modifiable risk factors in the pathogenesis and progression of prostate cancer (PC) and to identify potential strategies for preventing the onset of this disease. Exposure to modifiable and non-modifiable risk factors may independently or jointly contribute to the occurrence of pancreatic cancer. The detection of risk factors and precursor lesions will help identify the individuals at a higher risk for this neoplasia, improve prevention efforts to reduce exposure to certain factors, reduce the increasing incidence, and help in the early detection of this neoplasia.

2. Methods

The study included original research papers encompassing a range of methodologies, including retrospective and prospective cohort studies, clinical case-control studies, and systematic reviews. Publications such as technical reports, editor responses, narrative reviews, in silico studies, applied scientific posters, research proposals, and conference abstracts were excluded. Additionally, articles not in English and those unrelated to adenocarcinoma pancreatic risk factors were excluded.
The research was gathered through PubMed and the Google Academic Database. The searches covered a period of 10 years prior to September 2023. Articles were identified through the use of keywords such as “risk factors”, “pancreatic cancer”, and “pancreatic adenocarcinoma”. Subsequently, each risk factor, both modifiable and non-modifiable, was searched individually.
Two independent reviewers (RRG, IHD) conducted a comprehensive text and abstract screening and selected the relevant studies.

3. Risk Factors

Risk factors may be divided into non-modifiable risk factors, like age, sex, blood type, and genetic susceptivity, and modifiable risk factors, like diet, obesity, infections, and exposure to different chemicals and drugs (Figure 1).

3.1. Non-Modifiable Risk Factors

3.1.1. Age

Pancreatic cancer can develop at any age but frequently occurs in patients aged between 60 and 80 years [10,18,19,20]. In the last decades, life expectancy has increased steadily at a rate of 2.5 years per decade, reaching over 84 years in Japan [21]. Globally, over 11% of the world’s population is over 60 years of age, and a doubling of this percentage is projected by 2050 [22]. Given the peak incidence of pancreatic cancer and the aging rate of the population, this will lead to an increase in the number of cases, thus burdening the health system.
The link between aging and an increased incidence of neoplasia may be explained by the gradual aging-associated dysfunction of the mitochondrial electron transport chain, resulting in the increased production of superoxides with an accumulation of oxygen radicals and increased oxidative stress over the years, leading to damage to multiple cellular elements and determining phenotype changes [22].
Early onset pancreatic cancer (EOPC) is a term used to define patients diagnosed with pancreatic tumors before the age of 50 [18,23]. The relative frequency of EOPC ranges between 5 and 18% in different studies [23,24,25]. The diagnosis is frequently made at more advanced stages than for late-onset pancreatic cancer (LOPC). However, despite the stage, there is a tendency to administer significantly more treatment or to perform more surgical interventions for those with EOPC. The prognosis remains reserved, especially in the case of operated patients. In the case of unoperated ones, although survival remained dismal, no significant difference was observed compared to that of patients with LOPC [18].

3.1.2. Sex

The rates for PC, including EOPC, are consistently higher in men than in women [26,27]. The reason why there is a difference between the sexes is not entirely known, but it could be explained by a lower exposure of women to the modifiable risk factors.

3.1.3. Height

Some studies stipulated that increased height may be associated with an increased risk of pancreatic cancer [28], with an RR of 1.81 (95% Cl, 1.31–2.52) [29], but others failed to find such an association [30,31]. The correlation between adult height and neoplasia, which was also observed in other cancers [32], could be explained by exposure to higher levels of growth factors during childhood and early adolescence.

3.1.4. ABO Group

The ABO blood group antigens are encoded by one genetic locus on chromosome 9q34, which encodes three glycosyltransferases with different substrate specificities. Several studies have indicated that patients with blood type O appear to have a relatively lower risk of developing PC than other blood groups, particularly group A, which is significantly more common in affected patients [33,34,35]. It would seem that some glycosyltransferase enzymes may play a role in malignant cell immunosurveillance and intercellular adhesion during tumor genesis, but the mechanism for this association is not fully understood [36]. In the absence of a clear explanation, potentially attributable to augmented clearance and/or diminished secretion, it has been observed that antigen A is associated with diminished sICAM-1 concentrations [37]. This may lead to an increase in inflammation, which could further contribute to tumor growth and development. An association between PC and CagA-negative Helicobacter pylori (H. pylori) positivity has been demonstrated in one study among non-O blood type individuals but not for those with O blood type [38]. Conversely, a recent study confirmed an increased risk for those with non-0 blood type but failed to find a correlation with H. pylori infection status or CagA virulence status [39].

3.1.5. Genetic Factors

A hereditary component of PC has been observed in less than 10% of cases. The risk of developing pancreatic cancer varies depending on the specific genetic mutation present (Figure 2).
BRCA1 and BRCA2 are tumor suppressor genes that encode the proteins involved in repairing proteins that restore DNA double-stranded breaks via homologous recombination [40]. Mutations of these genes promote genomic instability and increase predisposition to malignant transformation and progression [41]. BRCA1 and 2 mutations have a general population occurrence rate between 1/300 and 1/800 [42], with a higher rate for the Ashkenazi Jewish ethnic group [43]. The penetrance is higher for female breast and ovarian cancer than for male breast cancer or PC [44]. Multiple studies have identified a risk up to threefold higher for BRCA1 mutation carriers in both sexes [43,45,46]. Mutations in BRCA2 have been found in up to 19% of familial PC and 7.3% of sporadic PC cases [47]. Identifying patients with BRCA mutation may represent an essential step in PC management, favoring early diagnosis and an increased surgical resection rate of PC. It was also found that these mutations could have implications for treatment options, leading to an increased response to platinum-based chemotherapy [48]. FANCC and FANCG mutations have been associated with EOPC [49,50].
Lynch syndrome (HNPCC) is the most common inherited colorectal cancer type. It is frequently associated with other types of cancer affecting various sites, such as the brain, pancreas, stomach, small intestine, urinary tract, endometrium, and ovary [51,52]. These patients have a germline mutation of genes encoding proteins involved in DNA mismatch repair: MSH2, MSH6, MLH1, PMS 1 and 2, and/or EPCAM [52], and they have an 8.6-fold increased risk of PC [53], with a lifetime risk of 1.3–4% [54]. The PC occurring in HNPCC patients frequently has a distinctive medullary appearance, which is otherwise a rare type of pancreatic adenocarcinoma and is associated with a better prognosis than conventional ductal adenocarcinoma [55,56,57]. Conversely, patients with the medullary PC phenotype are more likely to have a family history of cancer in first-degree relatives [56], suggesting a probable inherited susceptibility to HNPCC. It is essential to identify patients with PC and a pathogenic variant of the Lynch syndrome genes, as they may have tumors deficient in mismatched DNA repair and respond better to immunotherapy, as also suggested by Le et al. [58].
Familial adenomatous polyposis (FAP) is determined by mutations in the adenomatous polyposis coli (APC) tumor suppressor gene. It is characterized by the development of hundreds of colonic adenomatous polyps that progress to malignancy. It can also be associated with cancer in other sites such as the brain, thyroid, duodenum, pancreas, or hepatoblastoma [59]. Some studies stipulate that the risk of pancreatic cancer in this condition can be up to four times higher than in the general population (HR, 6.45; 95%CI, 2.02–20.64; p = 0.002) [60,61], thus raising awareness for updating active surveillance programs, even after prophylactic colectomy. However, these data may be misleading because some studies may have included patients with ampullary cancer due to misclassification, because the diagnosis of PC was not always histological, and a diagnosis based solely on imaging techniques may have confounded the two.
Peutz–Jeghers syndrome (PJS) is an autosomal dominant disease caused by mutations of the serine-threonine kinase 11 (STK11/LKB1) gene, a tumor suppressor gene that determines hamartomatous gastrointestinal polyposis and typical mucocutaneous pigmentation [62,63]. It is also associated with an increased risk of cancer of the esophagus, stomach, small intestine, colon, pancreas, pulmonary, breast, ovary, and uterus [64]. The relative risk for PC is increased to 76 (95%CI 36 to 160; p < 0.001) [65]. It is plausible that the risk of pancreatic cancer may be elevated in individuals with Peutz–Jeghers syndrome, akin to the observations made in the familial adenomatous polyposis population. This is potentially due to the difficulties in differentiating between pancreatic, distal bile duct, and ampullary cancers in some studies. Familial atypical mole melanoma pancreatic carcinoma syndrome (FAMMM-PC) is characterized by multiple malignant melanomas in first- and second-degree relatives and a 13 to 67-fold increased risk of pancreatic cancer, frequently with early onset (median age 55 years) [66,67,68,69]. It is caused by a germline mutation of p16INK4A (also known as CDKN2A or MTS1) [70,71]. CDKN2A is a gene implicated in cell proliferation. Parker et al. described that in cases of FAMMM, PC develops from normal duct epithelium to ductal hyperplasia to invasive ductal adenocarcinoma and suggested that patients with melanoma and CDKN2A mutation should be screened for pancreatic adenocarcinoma [72]. The identification of the CDKN2A mutation places patients at a high risk for pancreatic cancer, and their inclusion in screening programs improves the survival rate, as demonstrated by Vasen et al., who incorporated 19 patients with a CDKN2A mutation in such programs and found 13 patients with PC, 9 of them having a 5-year survival rate of 75% [73], higher than previously reported for patients with PC.
The SPINK1 gene encodes a serine protease inhibitor that inhibits active trypsin [74], and the PRSS1 gene encodes the prodigestive enzyme trypsinogen [75]. Mutation to PRSS1 can increase trypsin stability or trypsinogen autoactivation, favoring the development of chronic pancreatitis [76,77]. Mutation to SPINK1 and PRSS1 genes determine hereditary pancreatitis (HP) with a penetrance rate of 80% [78]. Recurrent episodes of pancreatitis determine chronic inflammation that decreases immunosurveillance and facilitates cell proliferation through Il-6 and 11, increasing the risk of PC [79]. Patients with hereditary pancreatitis have a 53-fold increase in the risk for PC (95%CI: 23–105) compared to the general population [80]. Smokers with hereditary pancreatitis develop the disease an average of 20 years before non-smokers [81].
Cystic fibrosis is determined by the mutation of the CFTR gene that regulates the flow of chloride ions across cell membranes. The mutation of this gene impairs ion transport and subsequently determines a chronic inflammation state in both respiratory and digestive systems [82]. Cystic fibrosis is associated with a 5.3-fold increased risk of PC, frequently with early onset [83]. This may be due to the increased viscosity of pancreatic juice, which may determine clogs, inflammation, and cell damage.
Ataxia-telangiectasia is caused by a homozygous mutation in the ATM gene that encodes a serine/threonine kinase involved in DNA repair [84]. It is characterized by progressive cerebellar ataxia, oculomotor apraxia, telangiectasias, increased sensitivity to ionizing radiation, and an increased risk of malignancies, such as breast and pancreatic neoplasia [84,85,86].
Based on the presence of a family history of PC and one of the above germline mutations, the International Cancer of the Pancreas Screening (CAPS) Consortium has developed a guideline to assist clinicians in the early diagnosis of this disease to improve survival. Screening should be made by MRI/MRCP or EUS, fasting serum glucose, and/or HbA1c at baseline and by adding CA 19–9 for a follow-up [87]. Considering that patients with at least one first-degree relative (FDR) and one second-degree relative (SDR) with PC are at an increased risk for this type of neoplasia, they should be included in screening programs at the age of 45–55 or at ten years younger than the youngest relative with PC [87]. The screening recommendations for those patients who are carriers of germline mutations are summarized in the table below (Table 1). The follow-up should be made yearly by MRI or EUS if no pancreatic abnormalities or low-risk features are detected. Although it was seen that smoking is a risk factor for progression, the interval is not shorter for this particular group. The role of surveillance of circulating tumor DNA in high-risk individuals still needs to be established.

3.1.6. Preneoplasic Pancreatic Lesions

Cystic lesions of the pancreas are tumor formations with varying degrees of malignant potential that can be classified as high risk or low risk (Figure 3). The risk of progression to pancreatic cancer (PC) varies depending on the relationship with the pancreatic duct. For instance, the risk of progression to PC in branch duct IPMN (BD-IPMN) is 19–30% [88], while in main duct IPMN (MD-IPMN), it is 40–60% [89].
There are clinical, radiological, and pathological features that may help in differentiating between various pancreatic lesions [90,91,92,93,94,95,96]. Although the MRI/MRCP is excellent for evaluating the location of pancreatic nodules, there is a high inter-observer variability regarding the risk stratification for IPMN [97]. Thus, because of the lack of reliable imaging and biomarkers that can help predict the development of some pancreatic cystic lesions, and also because there are even inconsistencies between various guidelines (e.g., Fukuoka criteria, AGA, ACR), the screening recommendations remain in the hands of the clinicians.
In recent years, novel techniques have been under study to help differentiate between pancreatic lesions, such as EUS-guided SF6 pancreatography (it can more accurately determine the communication between pancreatic cystic lesions and the pancreatic duct, with a sensitivity of 96.6% and specificity of 88.9% [98]) or PET using the 68Ga-labeled fibroblast activation protein inhibitor—68Ga-FAPI PET (it showed promising results in differentiating between high- and low-risk IPMN) [99]. In pancreatic cystic lesions, such as IMPN and MCN, the risk of PC development is higher for those with a male sex, increased age, smoking, obesity, and diabetes, including non-obese diabetes [100,101,102,103]. Regarding the link between IPMN and diabetes, it was found that patients with diabetes, especially those using insulin, have a higher incidence of BD-IPMN [100,104]. Surprisingly non-obese diabetes was not associated with an increased risk of IPMN, suggesting that these lesions do not secrete diabetogenic substances, like PC.
A nomogram taking into account the serum CEA and CA 19–9, the dimensions of the cyst, the presence of a thickened enhanced wall and an enhancing mural nodule, the main pancreatic duct (MPD) diameter, the presence of jaundice, and the prognostic nutrition index was found to have a high, over 80%, sensitivity and specificity for detecting high-risk IPMNs [105], but validation in a prospective study is needed. Another retrospective study also found that male sex, smoking, multifocality, diabetes, and recent weight loss were associated with developing worrisome features and high-grade dysplasia or cancer [106]. Worrisome features on imaging at presentation, the smoker status, and the presence of symptoms at diagnosis were associated with IPMN progression in a recent retrospective study [107]. Thus, patients diagnosed with pancreatic cysts have to be encouraged to cease smoking and maintain a normal BMI in order to reduce the risk of progression to neoplasia.
The value of CA 19–9 in screening programs is inconsistent, without a clear cut-off value. Although 37 kU/L increases the sensitivity, a higher value of 133 kU/L increases the specificity, avoiding the performance of multiple unnecessary screening tests or surgery [108]. Multiple studies found that patients with BD-IPMN develop worrisome futures or high-risk stigmata beyond the standard period for surveillance of 5 years and advise continuing surveillance after this period [109,110]. Studies on the psychological impact of PCN surveillance found higher anxiety levels and stress, with an overall reduced quality of life. All of the above should be considered when proposing a conservative approach that presumes regular clinic visits and individualized case-by-case visits [111].

3.2. Modifiable Risk Factors

3.2.1. Smoking

Smoking can induce PC through the cumulative effect of tobacco-related carcinogens (heterocyclic amines and polycyclic aromatic hydrocarbons) that can produce mutation to both proto-oncogenes and tumor suppressor genes and through smoke-induced chronic inflammation [112]. Animal studies have suggested that nicotine itself also acts by inducing the secretion of interleukin 8 (IL-8) and the upregulation of the IL-8 receptor, leading to increased tumor mass, increased tumor-free weight loss, and decreased muscle mass [113].
Smoking is thought to be responsible for up to 1/3 of PC cases [114,115]. The risk of PC for current smokers increases in a dose-dependent manner up to 2.7-fold for heavy smokers of more than 25 cigarettes/per day [115,116,117]. Although low (relative risk 1.48), it appears to persist even ten years after quitting [114,115] and becomes similar to that of non-smokers as late as 15–20 years after quitting [115]. Smoking cessation can reduce the frequency of PC at all ages, especially in EOPC [26].
Studying the epidemic trends of smoking, it was observed that there is a difference according to country and gender. The incidence of smoking among men in the Anglo-Saxon and North-West European countries has a wave pattern, with a peak around 1986 and a subsequent decline. In women in the same area, compared to men, the incidence started to increase after 24.2 years, on average, and peaked in 2014, reduced afterward by approximately 22.9%, but in some countries, it is still increasing. In low-income countries, the incidence peaked approximately ten years later (1993) [118]. Projection models estimate a continued decline in the smoking-associated mortality fraction (SAMF) for European men from an average of 25% in 2014 to an average of 11% in 2040, 7% in 2065, and 6% in 2100, while for women, there will initially be an increase followed by a steady decrease of 10% in 2040, 5% in 2065, and 4% in 2100, thus remaining below the average SAMF for men [119].
The incidence of EOPC was significantly increased in men in central and eastern Europe [26]. This overlaps with the gender-specific smoking prevalence rate [120]. Thus, it can be considered that individuals more susceptible to pancreatic lesions caused by the carcinogens contained in tobacco products could lower the onset of pancreatic cancer. Different studies found that EOPC patients were more frequently smokers, with a significantly younger age at smoking initiation than those with typical onset PC [23]. Multiple studies have shown that passive smoking is not associated with PC [121,122].
Although the rate of conventional cigarette smoking appears to be declining, the use of alternative nicotine-delivering products is increasing [123,124,125,126].
Regarding the use of alternative forms of nicotine products,
  • Snus, a smokeless oral tobacco product primarily used in Sweden and Norway, was not linked to the development of PC, and health risks are considered to be lower than those associated with cigarette smoking [127,128]
  • Electronic cigarettes are battery-operated devices that heat a liquid containing nicotine, a solvent, and one or more flavors. Studies on substances contained in e-cigarette aerosols have shown that they may contain carcinogens such as formaldehyde and acetaldehyde, but in much lower amounts than in the conventional cigarette [129,130], or supersaturated 1,2-propanediol vapor, which causes an increased production of nitric oxide, promoting inflammation [131,132], but there are still no studies evaluating the possible effect on the pancreatic tissue [133]
  • Heated tobacco products generate aerosols from heating tobacco at temperatures lower than combustion temperatures [134]. It has been shown that, similar to e-cigarettes, they may contain carcinogenic substances, but at a lower level than conventional cigarettes [135]
Smoking, in addition to the fact that it supports tumor genesis and its growth, also causes a decrease in the patient’s muscle mass and weight, thus accelerating the clinical deterioration. Considering that chemotherapy can be administered depending on the Eastern Cooperative Oncology Group (ECOG) performance status, patients should be advised to quit smoking even after diagnosis, to improve nutritional status, and to decrease tumor progression. Increasing anti-smoking campaigns to reduce the number of smokers from the beginning, as well as the promotion of programs to quit smoking, could reduce the risk of cancer and other diseases that are closely related to this vice. Although, at the moment, there are different alternative options to smoking, they also do not seem safe. The long-term effects on general health and carcinogenesis are not yet fully known, as long-term studies have not yet been conducted, and their use should also be discouraged.

3.2.2. Diabetes Mellitus (DM)

According to WHO, the number of people with diabetes increased from 108 million in 1980 to 422 million in 2014, and the prevalence increased faster in low- and middle-income countries than in high-income countries [136]. This is most likely due both to the fact that screening has been intensified and to the change in the definition of diabetes, with the lowering of the blood glucose level at which you are considered to have the disease.
Multiple studies demonstrated that hyperglycemia, abnormal glucose levels, and insulin resistance are associated with an increased risk of PC and also that PC has a diabetogenic effect, with half of PC patients having diabetes [137,138]. In contrast, approximately 1% of patients over 50 years old who are newly diagnosed with a metabolic disorder have PC as a trigger for their diabetes [139].
The diagnosis of type 2 diabetes (DM2) precedes that of cancer by up to 3 years in 25–85% of cases [138,140,141,142,143], thus suggesting that it may actually represent an early manifestation. Compared to the non-diabetic population, in the first year after the diagnosis of type 2 diabetes, there is a 14–15-fold higher risk of also being diagnosed with PC. The risk decreases in the second year to 3.5–5.4-fold and stabilizes at around 3-fold [144]. Diabetes could be considered secondary to the tumor-induced destruction of pancreatic acini or canalicular obstruction. However, more recent studies have shown that insulin and C-peptide secretion are increased in patients with PC and DM2, thus suggesting a more distant secondary effect on glucose metabolism, which returns to normal after cancer resection [138,145,146].
Sharma et al. developed a model called Enriching New-Onset Diabetes for Pancreatic Cancer (END-PAC) based on three factors—weight changes, blood glucose, and age of onset of diabetes—to distinguish new-onset DM (NOD) from pancreatic cancer-related diabetes (PCRD) [147]. This model has been validated in other retrospective studies [148,149,150], but not in a prospective study. A recent systematic review suggested that NOD associated with older age, a family history of pancreatic cancer, a personal history of gallstones/pancreatitis, weight loss, and a rapid increase in blood glucose is strongly related to PC and may be targeted for further screening strategies [151]. In another study, NOD associated with chronic obstructive pulmonary disease and increasing age was associated with PC [143]. High uric acid levels in diabetic women have also been associated with an increased risk of PC [152].
In multiple prospective cohort studies, researchers have observed a 2.2-fold increased risk of pancreatic cancer among subjects with high post-load plasma glucose levels [153,154,155]. At the same time, patients with long-standing DM2 have an increased risk of up to 2.4 times (Figure 4) [156]. Regarding the risk of PC after 15–20 years of diabetes evolution, there is no consensus, as the results of the studies are inconsistent [146,156]. The mechanism that could underline the association between DM2 and PC is that insulin resistance causes compensatory hyperinsulinemia and increases insulin-like growth hormone (IGF), which stimulates pancreatic ductal carcinogenesis. Insulin promotes cell proliferation and increases glucose use [157], while IGF has mitogenic, angiogenic, and antiapoptotic activities [158,159]. A recent study found that the gene conceding the insulin receptor can also be dysregulated, and the isoform A of the insulin receptor, which also has an oncogenic function, is upregulated [160].
Regarding DM2 therapy, metformin significantly decreases the risk of PC, but interestingly, this effect was not observed in other anti-diabetics such as insulin or sulfonylurea [161,162,163,164]. Metformin acts mainly by reducing the hepatic glucose output [165], thus lowering plasma insulin concentrations and indirectly inhibiting the effects of insulin and IGF, thus having an antitumor effect, inhibiting proliferation, and stimulating apoptosis [166].
With regard to type 1 diabetes (DM1), its incidence is considerably lower than that of DM2, and there is a paucity of available data. The studies examining the association between PC and DM1 exhibit considerable variation in their definition of type 1 diabetes. Some consider a diagnosis prior to the age of 40, while others include only those who have received insulin treatment. Although the risk of PC appears to be increased up to twice that of the general population in different studies [167,168,169], the inconsistency in the definition can easily lead to confounding with type 2. Consequently, further studies are required to gain a more accurate understanding of this association.

3.2.3. Obesity, Diet, and Lifestyle

The prevalence of obesity worldwide has nearly tripled between 1975 and 2016 [170]. Being overweight, with a body mass index greater than 25, increases the risk of PC, especially in women [171,172,173]. The risk appears to be increased by 20% for every 5-unit increase in BMI in early adulthood (age of 18–21 years) [31]. Obesity is linked with type 2 diabetes [174] and promotes inflammation [79,175]. Studies have shown that predominantly central abdominal adiposity distribution is more frequently associated with insulin resistance than peripheral adiposity [176]. An analysis of the pancreas in obese and diabetic individuals revealed an increased replication of the pancreatic duct by up to 10- and 4-fold, respectively, probably favoring PC development [177]. Some studies have found that a high waist-to-hip ratio may be associated with a 20–30% higher risk [28,31,178,179].
Hypercaloric diets and sedentary lifestyles lead to obesity, which causes ectopic pancreatic fat deposition and pancreatic steatosis [180]. The presence of intrapancreatic fat deposition was found to be associated with both pancreatic cancer (PC) and premalignant lesions in 52% [180], as well as with the progression of low-risk branch duct IPMN to PC [181].
Physical activity, even in the absence of weight loss, improves glucose tolerance [182]. Considering that insulin resistance increases IGF levels and stimulates pancreatic ductal carcinogenesis, having regular physical activity decreases the risk of PC. A reduced caloric intake and physical exercise have been shown to reduce the risk of PC [29].
A high-fat, high-energy diet promotes the absorption of bacterial lipopolysaccharides into circulation [183], and more lipopolysaccharide-producing bacteria have been found in the intestines of patients with PC [184]. A higher intake of red or processed meat, saturated fat, and fried foods may increase the occurrence of pancreatic cancer [185,186]. The presence of fat in the duodenum increases lipase secretion mediated by cholecystokinin-induced lipase secretion [187], thus leading to pancreatic hypertrophy and hyperplasia, increasing the risk of carcinogenesis [188].
An increased consumption of grains, nuts, vegetables, and fruits has a negative correlation with PC [185,188,189,190] and has been stated in multiple studies to even reduce the risk of PC by 38% [189], but others failed to observe such a link [191]. These foods are high in phytochemicals, such as carotenoids, phenolics, alkaloids, nitrogen-containing compounds, and organosulfur compounds [192,193], which have antioxidant and anti-inflammatory action, inhibit cell proliferation, and induce tumor suppressor gene expression and apoptosis [194]. Indeed, it was found that eating about 30 g of nuts two to three times a week and larger amounts of orange fruits and vegetables containing higher levels of beta carotene or green and red vegetables containing alpha-tocopherol is associated with a lower incidence of PC [195,196]. Isothiocyanates found in cruciferous vegetables have been shown to inhibit pancreatic cancer cells in in vitro and animal studies [197,198]. Fruits contain a large amount of vitamin C and other antioxidants that can trap free radicals, thus protecting against oxidative damage.
Regarding vitamin D levels, the data are inconsistent [199,200,201]. Vitamin D is involved in autocrine and paracrine cell differentiation, proliferation, and apoptosis [202]. 25-(OH)-D3 and 1,25-vitamin D analogs can inhibit cancer cell proliferation and promote apoptosis [203,204,205]. In vitro studies have shown that vitamin D regulates insulin synthesis [206]. Deficiency correlates with impaired pancreatic insulin synthesis and secretion in animals and humans [207,208]. Thus, the contradictory results highlighted by the studies can be explained at a speculative level by a possible protective effect of vitamin D on the proliferation and differentiation of pancreatic cells. Still, at the same time, an increased risk is determined by increased insulin secretion.

3.2.4. Pancreatitis

Pancreatitis has a complex pathophysiology that is not fully understood. The most common etiologies of acute pancreatitis (AP) are excessive alcohol consumption and gallstone disease [209]. The development involves irregular autophagy, leading to an enhanced inflammatory response in pancreatic tissue [210].
Pancreatitis is frequently associated with PC, but similar to DM, it can also increase the risk of PC as well as develop as a result of underlying PC. The strongest correlation between acute pancreatitis and pancreatic cancer is observed in patients with AP aged 56–75 without the two common underlying causes or during the initial year following an episode of acute pancreatitis (with an effective estimate of 23.47); conversely, this correlation diminishes two years after the episode of acute pancreatitis [211,212]. The episode of AP may be, in fact, the first manifestation of PC.
After the first episode of AP, approximately 8% develop chronic pancreatitis (CP) [213]. Most cases of CP are triggered by the variable combination of environmental factors such as alcohol and nicotine consumption [213] and some genetic mutations in the PRSS1 Gene, SPINK 1, and CTFR genes [214,215].
The risk of developing PC in patients with CP is higher in patients with CP due to PRSS1 mutations [216], in those with obesity, a history of alcohol, and nicotine abuse, and in those with CP with ductal dilatation [217]. The prevailing hypothesis is that chronic inflammation leads to increased cell proliferation and decreased immunosurveillance by the downregulation of the tumor-suppressing genes p16, p53, and SMAD 4 and the upregulation of the oncogenic KRAS gene, thus favoring the onset and growth of cancer [17,79,218]. A recent meta-analysis showed that the risk of PC in CP patients increases with the duration of the disease, excluding the potential cases where PC was diagnosed in close temporal proximity to CP diagnosis [219].
Another risk of CP is being misdiagnosed as CP while having PC, which can happen in about 5% of cases [220]. Similar to DM and AP, also in the case of CP, the highest association was seen in the first two years, especially in patients between the ages of 61 and 70 years, which progressively decreased until no correlation between AP or CP with PC was observed [221], thus suggesting that the patients were either misdiagnosed from the beginning or had both CP and PC [222].
Autoimmune pancreatitis (AIP) represents a rare cause of CP. Type 1 AIP is more common and is associated with increased IgG4. It may also be associated with the involvement of other organs, such as bile ducts, salivary glands, and lungs. Type 2 AIP accounts for a maximum of 5% of AIP cases and is not associated with elevated IgG4 and other type-specific extrapancreatic manifestations, except for inflammatory bowel diseases [223]. Studies on the possible link between AIP and PC have failed to identify an increased risk for PC compared to other causes of CP, possibly due to the small number of individuals included [219,224,225,226,227]. Another explanation may be that even though there is an increase in inflammation in AIP, this may not be associated with a decrease in immunosurveillance and the downregulation of tumor-suppressor genes.
It has been observed that pancreatitis may be followed by post-pancreatitis diabetes mellitus (PPDM) in up to 80% of cases [228], which is associated with poorer glycemic control compared with DM2 [229] and also with a higher risk of developing pancreatic cancer [230].

3.2.5. Alcohol

Inconsistent results have been reported for the association between alcohol consumption and PC. Some studies showed that alcohol consumption is associated with PC in a dose-dependent manner: low to moderate doses do not appear to increase the risk, while doses of >15–30–60 g of alcohol per day increase the risk up to 1.36-fold [231,232,233,234]. Other studies failed to show a link, even in genetically susceptible individuals [235,236]. Regarding the type of alcohol consumed, the data are also inconsistent. However, some meta-analyses showed that a high spirits/liquors intake was associated with an increased risk of PC in men but not in women [232,233,234]. The mechanism may be similar to tobacco use, meaning that high doses of alcohol contribute to inflammation and CP. In addition, alcohol metabolites such as acetaldehyde and even ethanol itself can cause differentiation defects in stem cells and promote inflammatory lesions and carcinogenesis by inhibiting DNA repair proteins [112,237,238].

3.2.6. Coffee

The relationship between coffee consumption and pancreatic cancer is not yet clearly defined. A systematic review and meta-analysis that included 959,992 participants revealed a dose-dependent increased risk, such that each cup of coffee a day increased the risk by 1 to 6% [239,240]. On the opposite side, other prospective studies and meta-analyses have found either no association [241,242] or a decrease in risk in the same dose-dependent manner [243,244,245]. The beneficial mechanism of coffee could probably be explained by its antioxidant properties [246] and its ability to trigger tissue antioxidant gene expression and protect against gastrointestinal oxidative stress [247]. More studies are needed to determine whether and how coffee consumption is associated with PC risk.

3.2.7. Hepatitis

Globally, the prevalence of hepatitis B patients is 3.5%, with higher rates in Africa and the western Pacific and the lowest rates in the USA [248]. The prevalence of hepatitis C is 1%, and the most affected regions are the Eastern Mediterranean and Europe [248]. Both hepatitis B and hepatitis C viruses are established carcinogens [249]. Although both viruses are hepatotropic and induce persistent liver injury [250,251], surface and core HBV antigens and HCV antigens have been identified in pancreatic acinar cells [252,253]. Studies have found an increased risk of pancreatic cancer among HCV- and HBV-infected patients [254,255,256]. The pathophysiological mechanism by which these viruses may contribute to PC development is not fully understood. The common origin of the blood vessels between the liver and the pancreas makes the migration of microorganisms possible [256]. The presence of these viruses in pancreatic tissue induces chronic inflammatory changes [257] that can promote proliferation.

3.2.8. Gallbladder Diseases and Cholecystectomy

Gallstones are highly prevalent in the general population [258], and the standard of care for symptomatic gallstones is cholecystectomy [259]. The overall evidence for the association between gallbladder gallstones and cholecystectomy and PC is inconsistent, probably due to the study design that variably accounts for other risk factors for PC (such as obesity, diabetes, and smoking), as well as the temporal relationship between PC diagnosis and gallbladder disease. A meta-analysis showed that patients with gallstones, cholecystectomy, or both have an increased risk for PC (RR 1.7; 95% Cl 1.3–2.21, respectively; RR 1.31; 95% Cl 1.19–1.43), but the analysis was performed independently of other risk factors [260]. Some studies confirmed these findings [261,262], while others found a correlation only in the first two years after the diagnosis of gallbladder disease, suspecting that a diagnosis bias drives this short-term link, and no long-term effect was identified [263]. A large Danish cohort study including 4,465,962 individuals, who were followed up for more than 30 years, found that symptomatic gallstones and cholecystectomy were associated with a lower risk for PC, with an HR of 0.82 (0.52–1.3) and 0.32 (0.19–0.54); on the other hand, sphincterotomy was associated with an increased risk (HR 3.85 (2.23–6.63)) [264]. This increased risk may be because sphincterotomy made by endoscopic retrograde cholangiography may have an inherent translocation risk of microbial species between the oral cavity and gastrointestinal tract and the biliary system because it disrupts the anatomical and functional barriers. These changes in the microbiota may induce chronic inflammation, thus promoting carcinogenesis.
As for asymptomatic gallstones, the data show an increased risk when compared to the control group (HR 1.25; 95% Cl 0.6–2.5) [265], but with a risk similar to the control group after cholecystectomy [266].

3.2.9. Periodontal Diseases

Periodontal inflammation, such as gingivitis and periodontitis, can determine systemic inflammation and is an independent risk factor for PC. A large Swedish cohort study that included over 5 million individuals found that younger individuals <50 years with mild dental inflammation, periodontitis, and root canal infection had a 35%, 56%, and 58% increased risk of PC, and those between 50 and 70 years old with periodontitis had a 20% risk of PC; meanwhile, in older patients, no association was seen [267], contrary to another study where the association between periodontal disease and PC was seen just in people aged 65 years or older (HR 2.17; 95%CI 1.03–4.57) [268]. Another study suggested that recent tooth loss could be a marker for severe periodontal disease and indirectly for PC, finding that the tooth loss over the last 4 years increased the risk for PC (RR 2.71, 95% Cl 1.70–4.32) [269]. Studies showed that Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans, two oral microorganisms often implicated in periodontitis, can increase the risk of pancreatic cancer by up to 60% also in cases without periodontitis [270,271,272,273,274]. In contrast, other microorganisms, such as Phylum Fusobacteria and its genus Leptotrichia, were associated with a decreased risk of PC [271]. Salivary microbiota analysis revealed decreased levels of Neisseria elongata and Streptococcus Mitis in patients with PC, with a sensitivity of 94.6% and a specificity of 82.1% in differentiating them from healthy individuals using the ROC-plot (AUC value of 0.90 (95%CI 0.78 to 0.96, p < 0.0001)) [275].
The underlying mechanisms between the oral microbiota and PC are not yet elucidated. However, a hypothesis may be that oral dysbiosis could determine changes in the intestinal and pancreatic microbiota that promote inflammation and oncogenesis, but further investigations are needed.

3.2.10. Helicobacter pylori (H. pylori)

H. pylori is an established carcinogen [249], detectable in over 50% of the world’s population [276]. The association between H. pylori infection and the risk of pancreatic cancer remains controversial. It is primarily related to an indirect mechanism based on disrupting the cell-to-cell adhesion and intracellular signaling, triggering inflammatory processes and promoting neoplastic transformation [38,277,278,279].

3.2.11. Autoimmune Diseases

Autoimmune diseases are characterized by an aberrant immune response in which immune cells react against self-antigens. While most autoimmune diseases are associated with a reduced risk of PC [280], cutaneous and systemic lupus appears to be associated with an increased risk [281] (RR 1.41 (95%Cl: 1.13–1.79), especially in the European region, but not for other regions such as America and Asia [282].

3.2.12. Polycystic Ovary Syndrome (POCS)

Limited data have been reported on POCS and PC. PCOS is one of the most common endocrinological diseases affecting women of reproductive age, and it has been associated with a higher prevalence of non-alcoholic fatty pancreas, metabolic syndrome, and insulin resistance [283]. In 2019, a Swedish registry study evaluating the association between different cancer sites and PCOS reported a 3.4-fold higher risk of PC in premenopausal women with PCOS [284]. Another study also found an increased association between these two pathologies, although the association was slightly weaker after accounting for the data regarding diabetes and obesity [285].
Multiple case-control and cohort studies on women taking hormonal contraception reported no association between oral contraceptive use and the risk of PC [286,287,288].

3.2.13. Microbiota

Once considered a sterile organ, recent studies established the presence of pancreatic microbiota, both in a normal and pathogenic state, but with inconsistent data regarding its role in carcinogenesis [273,289,290,291]. The possible mechanism by which the presence of different microorganisms can be involved in carcinogenesis is the ability to induce a pro-inflammatory response [292,293], inhibit the immune response aimed at eliminating tumor cells [294], and modulate cell division [279,295]. Different bacterial strains stimulate the higher release and absorption of lipopolysaccharides (LPS), which are components of the outer membrane of Gram-negative bacteria activating the host’s innate immune system [183,296], promoting chronic inflammation at the beginning, because afterward, LPS reduces the immune response by interacting with NF-kB and upregulating the expression of programmed cell death ligand 1 (PD-L1) [297]. Furthermore, more LPS could trigger Toll-like receptor 4 and thus inhibit tumor suppressor genes like PTEN and p53 [298,299]. Data emerging in recent years promote the idea that the host microbiome and the pancreas are interconnected and that the microbes included in that microbiome, such as Mycoplasma Hyorhinits, can even influence chemotherapy regimens used in PC [289,300,301]. Also, tumor microbiota may influence survival; patients with high alpha diversity (Pseudoxanthomonas/Streptomyces/Saccharopolyspora/Bacillus clausii) have significantly longer overall survival than those with low alpha diversity [302]. Fecal microbiota analysis showed that patients with PC have a significant increase in Bacteroides and Gammaproteobacteria and a reduction in butyrate-producing bacteria compared to healthy controls [184,303,304]. Animal studies have found that probiotics (containing Lactobacillus strain) can increase the efficacy of gemcitabine and may also increase the patient’s tolerance to chemotherapy [305]. Patients with positive intraoperative bile cultures for Klebsiella pneumoniae had better progression-free survival only after adding quinolone to gemcitabine and not on gemcitabine alone, suggesting that Klebsiella pneumoniae may promote chemoresistance [306].
Summarizing the data available, intestinal and tumoral microbiota and their products are implicated in both pancreatic carcinogenesis, tumor progression, and prognostics. However, further studies may lead to findings that may be applicable in clinical settings.

3.2.14. Psychological Stress

The possible mechanism by which stress may be associated with an increased risk of cancer is the dysregulation of the thalamic–pituitary–adrenal axis and sympathetic nervous system and the inappropriate secretion of glucocorticoid and catecholamines [307], which can alter the cellular immune response to cancer cells.
Exposure to severe psychological stress in childhood and young adulthood, such as the loss of a parent by death, was assessed in a Swedish cohort study and associated with the early onset of PC, regardless of the age of loss [308]. Similarly, an increased risk for PC was also found in women but not in men, who experienced severe stress, such as the death of a child. The increased risk for PC became significant when considering the first five years after the loss of the child when the loss was due to suicide and when considering people with a history of psychiatric illnesses [309].
Several studies show that B-blockers, which are used for treating arrhythmia, chronic stress, and depression, have an inhibitory effect on tumorigenesis, inhibiting the damage induced by the catecholamines stimulation of adrenoreceptors in patients with PC, inhibiting the progression of PC in patients without metastasis [310,311,312,313,314].

3.2.15. Renin-Angiotensin Inhibitors

Studies performed on animal models have found that angiotensin II has a pro-proliferative effect on smooth muscle [315]. Angiotensin I-converting enzyme inhibitors (ACI) that inhibit the conversion of angiotensin I to angiotensin II have an oncoprotective role by reducing oxidative stress and inflammation and by downregulating vascular endothelial growth factor, responsible for angiogenesis and nuclear factor kappa beta, which promotes the production of oncogenic proteins [316,317]. Recent cohort studies based on two populations showed a reduced risk for pancreaticobiliary cancer in patients taking ACI (HR 0.62 (0.54–0.72)) (HR 0.69 (0.53–0.9)) [318,319], while another one failed to find an association between ACI consumption and PC [320].
The use of ACI in patients with resected PC has been associated with more prolonged overall survival [321,322].

3.2.16. Allergies

Several epidemiological studies and meta-analyses that examined the association between allergy history and PC risk found that allergies, especially respiratory ones, such as hay fever and animal and plant allergies, statistically significantly decrease the risk [323,324,325,326,327,328,329], but other prospective studies have failed to show an association [330].
The mechanism by which allergies could reduce the risk of PC still needs to be fully elucidated. Enhanced immune surveillance may have a role [323,331], leading to the elimination of neoplastic cells [332] by favoring the IgE response against tumor antigens [333].
While the potential role of the medical treatment of allergies in PC is an intriguing area of study, our current understanding is limited [330,334]. Further research is needed to explore this association and its implications.

3.2.17. Opioids

Opioids are drugs used for pain management and recreational purposes, with increased use over the past decade. It has been shown that opioids can promote cancer progression in various cancer types by promoting angiogenesis and epithelial-to-mesenchymal transition [335] or by altering the gut microbiome [336]. Multiple studies have suggested that opium use increases the risk of PC [337,338,339,340].

3.2.18. Proton Pump Inhibitors (PPI)

PPIs are, in most countries, over-the-counter medications commonly used for acid-related disorders. Two recent independent meta-analyses found that PPI use was associated with an approximately 63% increase in PC risk [341,342]. The mechanism underlying the possible link between PPI use and PC is given by the effects of PPI use, such as decreased gastric acid production and hypergastrinemia. Decreased gastric acid may promote bacterial overgrowth and nitrosamine secretion, increasing pancreatic cell proliferation [343]. Second, gastrin also has a trophic effect on epithelial cells, thus favoring the development of pancreatic neoplasia [344]. On the other hand, in healthy PPI users, a gut dysbiosis that was also associated with PC was seen, thus suggesting that the impact of PPI on microbiota may also play a role in pathogenesis [345].

3.2.19. Nonsteroidal Anti-Inflammatory Drugs (NSAID)

Data on PC risk and the use of NSAIDs is inconsistent. A study including 88,378 women reported a possible increased risk of PC among the individuals who used acetylsalicylic acid regularly [346]. A more recent meta-analysis, which included approximately 258,000 participants, indicated a possible protective effect of aspirin use that increases with the frequency of administrations but failed to show a similar effect for other NSAIDs [347].

3.2.20. Statins

Statins have anti-inflammatory properties, but studies suggest that their use in CP patients does not decrease the risk of PC [348].
The data regarding modifiable risk factors and potential interventions have been synthesized in the table below (Table 2).

4. Discussion and Conclusions

The high mortality rate of PC due to a late diagnosis emphasizes the importance of developing methods for reducing the modifiable risk factors and for helping detect resectable tumors. The main goals in improving the survival of patients with PC are decreasing the factors contributing to the appearance of this neoplasia, identifying high-risk populations, and implementing efficient and cost-effective screening methods. The present review highlights the factors associated with an increased or reduced risk of PC.
With regard to non-modifiable risk factors, the CAPS guidelines suggest that screening for multiple mutations should commence at the age of 40–50 years. The issue is that, despite being recommended in many countries, the high costs and limited accessibility make it unfeasible in practice. Furthermore, the recommendations regarding BRCA mutations assume that at least one family member has already been affected. However, 50% of those with germline BRCA mutations may not have a positive family medical history of BRCA-associated cancer. In addition to the elevated risk of pancreatic cancer in individuals with BRCA mutations, another crucial consideration is the impact on therapeutic options and prognosis. BRCA-mutated pancreatic cancers are markedly sensitive to platinum-based therapies, suggesting that testing all patients with pancreatic cancer for BRCA mutations could lead to the optimization of early therapy to improve survival.
Pancreatic cancer risk increases sharply with smoking, even in the case of a small number of cigarettes or after several years of smoking, and it decreases slowly, taking over 20 years to reach the risk of never-smokers. Thus, a campaign against conventional tobacco consumption could reduce the incidence, although studies regarding alternative nicotine delivery methods are still unclear.
Aiming for a normal BMI and a balanced diet, decreasing red meat and fat consumption, increasing physical activity and fruit and vegetable consumption, associating higher nutrient density and better diet quality with a low to moderate consumption of alcohol and coffee, and rigorous oral hygiene are wise choices that could contribute to a lower risk of PC, although the data are inconsistent.
In patients over 50–60 years of age, diabetes, gallbladder disease, and recently diagnosed pancreatitis are frequently associated with pancreatic cancer in the first three years, so additional attention for monitoring could help in early diagnosis. In the case of diabetes, screening with END-PAC could help differentiate between NOD and PCRD. At the same time, treatment with metformin should be the first treatment choice for glycemic control to reduce the risk of PC.
Last but not least, avoiding stressful events and controlling them with adequate medication and therapy could reduce the risk of PC.
A new front is changing the microbiota, but as the understanding of the mechanism of association with PC is still limited, it is clear that this area can benefit from further research. Microbiota can play a role in the early diagnosis of PC, and a new therapeutic intervention based on bacteria-related function could be generated.
Although there are screening programs for high-risk patients either due to genetics or the presence of precursor lesions, or both, they do not consider other modifiable risk factors. Smoking, for example, which is known to reduce the time of PC development by up to 10–20 years, along with other risk factors, could be included to initiate early screening in high-risk patients.

Author Contributions

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

Funding

This research received no external funding.

Institutional Review Board Statement

The study did not require ethical approval.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

We would like to express our gratitude to Radu Cristina for providing a stimulating and supportive academic environment.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ACIAngiotensin I-converting enzyme inhibitors
ACRAmerican College of Radiology
AGAAmerican Gastroenterological Association
APAcute pancreatitis
APCAdenomatous polyposis coli
BD-IPMNBranch duct intraductal papillary mucinous neoplasm
BMIBody mass index
CA 19-9Cancer antigen 19-9
CAPSInternational Cancer of the Pancreas Screening
CEACarcinoembryonic antigen
CPChronic pancreatitis
DMDiabetes mellitus
DM1Diabetes mellitus type 1
DM2Diabetes mellitus type 2
DNADeoxyribonucleic acid
ECOGEastern Cooperative Oncology Group
END-PACEnriching New-Onset Diabetes for Pancreatic Cancer
EOPCEarly onset pancreatic cancer
EUSEndoscopic ultrasound
FAMMM-PCFamilial atypical mole melanoma pancreatic carcinoma syndrome
FAPFamilial adenomatous polyposis
FDRFirst-degree relative
68Ga-FAPI PET PETusing 68Ga-labeled fibroblast activation protein inhibitor
HBVHepatitis B virus
HCVHepatitis C virus
HNPCCHereditary nonpolyposis colorectal cancer (Lynch syndrome)
IGFInsulin-like growth factor
IPMNintraductal papillary mucinous neoplasm
KRASKirsten-rat sarcoma
LOPClate-onset pancreatic cancer
LPSLipopolysaccharides
MCNMucinous cystic neoplasm
MD-IPMNMain duct intraductal papillary mucinous neoplasm
MPDMain pancreatic duct
MRCPMagnetic resonance cholangiopancreatography
MRIMagnet resonance imaging
NODNew-onset diabetes mellitus
NSAIDNonsteroidal Anti-Inflammatory Drugs
PCPancreatic cancer
PCNPancreatic cystic neoplasm
PCRDPancreatic cancer-related diabetes
PD-L1Programmed cell death ligand 1
PETPositron Emission Tomography
PJSPeutz-Jeghers syndrome
POCSPolycystic Ovary Syndrome
PPDMPost-pancreatitis diabetes mellitus
PPIProton Pump Inhibitors
SAMFsmoking-associated mortality fraction
SDRsecond-degree relative
SEERSurveillance, Epidemiology and End Results
STKSerine-threonine kinase
WHOWord Health Organization

References

  1. Pancreatic Neuroendocrine Tumors (PNETs). Available online: https://pancan.org/facing-pancreatic-cancer/about-pancreatic-cancer/types-of-pancreatic-cancer/endocrine-pancreatic-neuroendocrine-tumors/ (accessed on 19 July 2024).
  2. Becker, A.E.; Hernandez, Y.G.; Frucht, H.; Lucas, A.L. Pancreatic ductal adenocarcinoma: Risk factors, screening, and early detection. World J. Gastroenterol. 2014, 20, 11182–11198. [Google Scholar] [CrossRef] [PubMed]
  3. Cancer of the Pancreas—Cancer Stat Facts. Available online: https://seer.cancer.gov/statfacts/html/pancreas.html (accessed on 12 March 2023).
  4. Dyba, T.; Randi, G.; Bray, F.; Martos, C.; Giusti, F.; Nicholson, N.; Gavin, A.; Flego, M.; Neamtiu, L.; Dimitrova, N.; et al. The European cancer burden in 2020: Incidence and mortality estimates for 40 countries and 25 major cancers. Eur. J. Cancer 2021, 157, 308–347. [Google Scholar] [CrossRef] [PubMed]
  5. Hu, J.-X.; Zhao, C.-F.; Chen, W.-B.; Liu, Q.-C.; Li, Q.-W.; Lin, Y.-Y.; Gao, F. Pancreatic cancer: A review of epidemiology, trend, and risk factors. World J. Gastroenterol. 2021, 27, 4298–4321. [Google Scholar] [CrossRef] [PubMed]
  6. Huang, J.; Lok, V.; Ngai, C.H.; Zhang, L.; Yuan, J.; Lao, X.Q.; Ng, K.; Chong, C.; Zheng, Z.-J.; Wong, M.C.S. Worldwide Burden of, Risk Factors for, and Trends in Pancreatic Cancer. Gastroenterology 2021, 160, 744–754. [Google Scholar] [CrossRef] [PubMed]
  7. Bilimoria, K.Y.; Bentrem, D.J.; Ko, C.Y.; Ritchey, J.; Stewart, A.K.; Winchester, D.P.; Talamonti, M.S. Validation of the 6th edition AJCC Pancreatic Cancer Staging System: Report from the National Cancer Database. Cancer 2007, 110, 738–744. [Google Scholar] [CrossRef] [PubMed]
  8. Zhou, B.; Xu, J.-W.; Cheng, Y.-G.; Gao, J.-Y.; Hu, S.-Y.; Wang, L.; Zhan, H.-X. Early detection of pancreatic cancer: Where are we now and where are we going? Int. J. Cancer 2017, 141, 231–241. [Google Scholar] [CrossRef] [PubMed]
  9. Cho, J.; Petrov, M.S. Pancreatitis, Pancreatic Cancer, and Their Metabolic Sequelae: Projected Burden to 2050. Clin. Transl. Gastroenterol. 2020, 11, e00251. [Google Scholar] [CrossRef] [PubMed]
  10. Rahib, L.; Smith, B.D.; Aizenberg, R.; Rosenzweig, A.B.; Fleshman, J.M.; Matrisian, L.M. Projecting cancer incidence and deaths to 2030: The unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014, 74, 2913–2921. [Google Scholar] [CrossRef] [PubMed]
  11. Brat, D.J.; Lillemoe, K.D.; Yeo, C.J.; Warfield, P.B.; Hruban, R.H. Progression of pancreatic intraductal neoplasias to infiltrating adenocarcinoma of the pancreas. Am. J. Surg. Pathol. 1998, 22, 163–169. [Google Scholar] [CrossRef]
  12. Brockie, E.; Anand, A.; Albores-Saavedra, J. Progression of atypical ductal hyperplasia/carcinoma in situ of the pancreas to invasive adenocarcinoma. Ann. Diagn. Pathol. 1998, 2, 286–292. [Google Scholar] [CrossRef]
  13. Berthélemy, P.; Bouisson, M.; Escourrou, J.; Vaysse, N.; Rumeau, J.L.; Pradayrol, L. Identification of K-ras mutations in pancreatic juice in the early diagnosis of pancreatic cancer. Ann. Intern. Med. 1995, 123, 188–191. [Google Scholar] [CrossRef] [PubMed]
  14. Boadas, J.; Mora, J.; Urgell, E.; Puig, P.; Roca, M.; Cussó, X.; Capellà, G.; Lluís, F.; Farré, A. Clinical usefulness of K-ras gene mutation detection and cytology in pancreatic juice in the diagnosis and screening of pancreatic cancer. Eur. J. Gastroenterol. Hepatol. 2001, 13, 1153–1159. [Google Scholar] [CrossRef] [PubMed]
  15. Lu, X.; Xu, T.; Qian, J.; Wen, X.; Wu, D. Detecting K-ras and p53 gene mutation from stool and pancreatic juice for diagnosis of early pancreatic cancer. Chin. Med. J. 2002, 115, 1632–1636. [Google Scholar] [PubMed]
  16. Wakabayashi, T.; Sawabu, N.; Watanabe, H.; Morimoto, H.; Sugioka, G.; Takita, Y. Detection of K-ras point mutation at codon 12 in pure pancreatic juice collected 3 years and 6 months before the clinical diagnosis of pancreatic cancer. Am. J. Gastroenterol. 1996, 91, 1848–1851. [Google Scholar] [PubMed]
  17. Maire, F.; Micard, S.; Hammel, P.; Voitot, H.; Lévy, P.; Cugnenc, P.-H.; Ruszniewski, P.; Puig, P.L. Differential diagnosis between chronic pancreatitis and pancreatic cancer: Value of the detection of KRAS2 mutations in circulating DNA. Br. J. Cancer 2002, 87, 551–554. [Google Scholar] [CrossRef]
  18. Ansari, D.; Althini, C.; Ohlsson, H.; Andersson, R. Early-onset pancreatic cancer: A population-based study using the SEER registry. Langenbecks Arch. Surg. 2019, 404, 565–571. [Google Scholar] [CrossRef] [PubMed]
  19. Ansari, D.; Tingstedt, B.; Andersson, B.; Holmquist, F.; Sturesson, C.; Williamsson, C.; Sasor, A.; Borg, D.; Bauden, M.; Andersson, R. Pancreatic cancer: Yesterday, today and tomorrow. Future Oncol. 2016, 12, 1929–1946. [Google Scholar] [CrossRef] [PubMed]
  20. Stark, A.P.; Sacks, G.D.; Rochefort, M.M.; Donahue, T.R.; Reber, H.A.; Tomlinson, J.S.; Dawson, D.W.; Eibl, G.; Hines, O.J. Long-term Survival in Patients with Pancreatic Ductal Adenocarcinoma. Surgery 2016, 159, 1520–1527. [Google Scholar] [CrossRef] [PubMed]
  21. World Health Organization. Japan Data. Available online: https://data.who.int/countries/392 (accessed on 1 October 2023).
  22. Newgard, C.B.; Sharpless, N.E. Coming of age: Molecular drivers of aging and therapeutic opportunities. J. Clin. Investig. 2013, 123, 946–950. [Google Scholar] [CrossRef]
  23. Piciucchi, M.; Capurso, G.; Valente, R.; Larghi, A.; Archibugi, L.; Signoretti, M.; Stigliano, S.; Zerboni, G.; Barucca, V.; La Torre, M.; et al. Early onset pancreatic cancer: Risk factors, presentation and outcome. Pancreatology 2015, 15, 151–155. [Google Scholar] [CrossRef]
  24. Lin, J.-C.; Chan, D.-C.; Chen, P.-J.; Chu, H.-C.; Chueh, T.-H.; Huang, H.-H.; Chang, P.-Y.; Yu, C.-P.; Chang, W.-K.; Hsieh, T.-Y. Clinical Characteristics of Early Onset Pancreatic Adenocarcinoma: A Medical Center Experience and Review of the Literature. Pancreas 2011, 40, 638. [Google Scholar] [CrossRef]
  25. Ntala, C.; Debernardi, S.; Feakins, R.M.; Crnogorac-Jurcevic, T. Demographic, clinical, and pathological features of early onset pancreatic cancer patients. BMC Gastroenterol. 2018, 18, 139. [Google Scholar] [CrossRef]
  26. Raimondi, S.; Maisonneuve, P.; Löhr, J.-M.; Lowenfels, A.B. Early onset pancreatic cancer: Evidence of a major role for smoking and genetic factors. Cancer Epidemiol. Biomark. Prev. 2007, 16, 1894–1897. [Google Scholar] [CrossRef]
  27. International Agency for Research on Cancer (IARC). Global Cancer Observatory. Available online: https://gco.iarc.fr/ (accessed on 20 July 2024).
  28. Berrington de González, A.; Spencer, E.A.; Bueno-de-Mesquita, H.B.; Roddam, A.; Stolzenberg-Solomon, R.; Halkjaer, J.; Tjønneland, A.; Overvad, K.; Clavel-Chapelon, F.; Boutron-Ruault, M.-C.; et al. Anthropometry, physical activity, and the risk of pancreatic cancer in the European prospective investigation into cancer and nutrition. Cancer Epidemiol. Biomark. Prev. 2006, 15, 879–885. [Google Scholar] [CrossRef]
  29. Michaud, D.S.; Giovannucci, E.; Willett, W.C.; Colditz, G.A.; Stampfer, M.J.; Fuchs, C.S. Physical Activity, Obesity, Height, and the Risk of Pancreatic Cancer. JAMA 2001, 286, 921–929. [Google Scholar] [CrossRef]
  30. Stevens, R.J.; Roddam, A.W.; Spencer, E.A.; Pirie, K.L.; Reeves, G.K.; Green, J.; Beral, V.; Million Women Study Collaborators. Factors associated with incident and fatal pancreatic cancer in a cohort of middle-aged women. Int. J. Cancer 2009, 124, 2400–2405. [Google Scholar] [CrossRef]
  31. Genkinger, J.M.; Spiegelman, D.; Anderson, K.E.; Bernstein, L.; van den Brandt, P.A.; Calle, E.E.; English, D.R.; Folsom, A.R.; Freudenheim, J.L.; Fuchs, C.S.; et al. A pooled analysis of 14 cohort studies of anthropometric factors and pancreatic cancer risk. Int. J. Cancer 2011, 129, 1708–1717. [Google Scholar] [CrossRef]
  32. Green, J.; Cairns, B.J.; Casabonne, D.; Wright, F.L.; Reeves, G.; Beral, V. Height and cancer incidence in the Million Women Study: Prospective cohort, and meta-analysis of prospective studies of height and total cancer risk. Lancet Oncol. 2011, 12, 785–794. [Google Scholar] [CrossRef] [PubMed]
  33. Iodice, S.; Maisonneuve, P.; Botteri, E.; Sandri, M.T.; Lowenfels, A.B. ABO blood group and cancer. Eur. J. Cancer 2010, 46, 3345–3350. [Google Scholar] [CrossRef] [PubMed]
  34. Wolpin, B.M.; Chan, A.T.; Hartge, P.; Chanock, S.J.; Kraft, P.; Hunter, D.J.; Giovannucci, E.L.; Fuchs, C.S. ABO blood group and the risk of pancreatic cancer. J. Natl. Cancer Inst. 2009, 101, 424–431. [Google Scholar] [CrossRef] [PubMed]
  35. Vioque, J.; Walker, A.M. Pancreatic cancer and ABO blood types: A study of cases and controls. Med. Clin. 1991, 96, 761–764. [Google Scholar]
  36. Hakomori, S. Antigen structure and genetic basis of histo-blood groups A, B and O: Their changes associated with human cancer. Biochim. Biophys. Acta 1999, 1473, 247–266. [Google Scholar] [CrossRef] [PubMed]
  37. Paré, G.; Chasman, D.I.; Kellogg, M.; Zee, R.Y.L.; Rifai, N.; Badola, S.; Miletich, J.P.; Ridker, P.M. Novel Association of ABO Histo-Blood Group Antigen with Soluble ICAM-1: Results of a Genome-Wide Association Study of 6578 Women. PLoS Genet. 2008, 4, e1000118. [Google Scholar] [CrossRef]
  38. Risch, H.A.; Yu, H.; Lu, L.; Kidd, M.S. ABO Blood Group, Helicobacter pylori Seropositivity, and Risk of Pancreatic Cancer: A Case–Control Study. J. Natl. Cancer Inst. 2010, 102, 502–505. [Google Scholar] [CrossRef]
  39. Lee, A.A.; Wang, Q.-L.; Kim, J.; Babic, A.; Zhang, X.; Perez, K.; Ng, K.; Nowak, J.; Rifai, N.; Sesso, H.D.; et al. Helicobacter pylori Seropositivity, ABO Blood Type, and Pancreatic Cancer Risk from 5 Prospective Cohorts. Clin. Transl. Gastroenterol. 2023, 14, e00573. [Google Scholar] [CrossRef] [PubMed]
  40. Venkitaraman, A.R. Cancer susceptibility and the functions of BRCA1 and BRCA2. Cell 2002, 108, 171–182. [Google Scholar] [CrossRef] [PubMed]
  41. Tutt, A.; Ashworth, A. The relationship between the roles of BRCA genes in DNA repair and cancer predisposition. Trends Mol. Med. 2002, 8, 571–576. [Google Scholar] [CrossRef] [PubMed]
  42. Paluch-Shimon, S.; Cardoso, F.; Sessa, C.; Balmana, J.; Cardoso, M.J.; Gilbert, F.; Senkus, E.; ESMO Guidelines Committee. Prevention and screening in BRCA mutation carriers and other breast/ovarian hereditary cancer syndromes: ESMO Clinical Practice Guidelines for cancer prevention and screening. Ann. Oncol. 2016, 27, v103–v110. [Google Scholar] [CrossRef]
  43. Lucas, A.L.; Shakya, R.; Lipsyc, M.D.; Mitchel, E.B.; Kumar, S.; Hwang, C.; Deng, L.; Devoe, C.; Chabot, J.A.; Szabolcs, M.; et al. High Prevalence of BRCA1 and BRCA2 Germline Mutations with Loss of Heterozygosity in a Series of Resected Pancreatic Adenocarcinoma and Other Neoplastic Lesions. Clin. Cancer Res. 2013, 19, 3396–3403. [Google Scholar] [CrossRef]
  44. Tai, Y.C.; Domchek, S.; Parmigiani, G.; Chen, S. Breast cancer risk among male BRCA1 and BRCA2 mutation carriers. J. Natl. Cancer Inst. 2007, 99, 1811–1814. [Google Scholar] [CrossRef]
  45. Brose, M.S.; Rebbeck, T.R.; Calzone, K.A.; Stopfer, J.E.; Nathanson, K.L.; Weber, B.L. Cancer risk estimates for BRCA1 mutation carriers identified in a risk evaluation program. J. Natl. Cancer Inst. 2002, 94, 1365–1372. [Google Scholar] [CrossRef] [PubMed]
  46. Thompson, D.; Easton, D.F.; Breast Cancer Linkage Consortium. Cancer Incidence in BRCA1 mutation carriers. J. Natl. Cancer Inst. 2002, 94, 1358–1365. [Google Scholar] [CrossRef]
  47. Hahn, S.A.; Greenhalf, B.; Ellis, I.; Sina-Frey, M.; Rieder, H.; Korte, B.; Gerdes, B.; Kress, R.; Ziegler, A.; Raeburn, J.A.; et al. BRCA2 germline mutations in familial pancreatic carcinoma. J. Natl. Cancer Inst. 2003, 95, 214–221. [Google Scholar] [CrossRef]
  48. Pishvaian, M.J.; Blais, E.M.; Brody, J.R.; Lyons, E.; DeArbeloa, P.; Hendifar, A.; Mikhail, S.; Chung, V.; Sahai, V.; Sohal, D.P.S.; et al. Overall survival in patients with pancreatic cancer receiving matched therapies following molecular profiling: A retrospective analysis of the Know Your Tumor registry trial. Lancet Oncol. 2020, 21, 508–518. [Google Scholar] [CrossRef]
  49. Couch, F.J.; Johnson, M.R.; Rabe, K.; Boardman, L.; McWilliams, R.; de Andrade, M.; Petersen, G. Germ line Fanconi anemia complementation group C mutations and pancreatic cancer. Cancer Res. 2005, 65, 383–386. [Google Scholar] [CrossRef]
  50. van der Heijden, M.S.; Yeo, C.J.; Hruban, R.H.; Kern, S.E. Fanconi anemia gene mutations in young-onset pancreatic cancer. Cancer Res. 2003, 63, 2585–2588. [Google Scholar]
  51. Kastrinos, F.; Stoffel, E.M. History, Genetics, and Strategies for Cancer Prevention in Lynch Syndrome. Clin. Gastroenterol. Hepatol. 2014, 12, 715–727. [Google Scholar] [CrossRef] [PubMed]
  52. Rustgi, A.K. The genetics of hereditary colon cancer. Genes Dev. 2007, 21, 2525–2538. [Google Scholar] [CrossRef] [PubMed]
  53. Kastrinos, F.; Mukherjee, B.; Tayob, N.; Wang, F.; Sparr, J.; Raymond, V.M.; Bandipalliam, P.; Stoffel, E.M.; Gruber, S.B.; Syngal, S. The Risk of Pancreatic Cancer in Families with Lynch Syndrome. JAMA 2009, 302, 1790–1795. [Google Scholar] [CrossRef]
  54. Dunlop, M.G.; Farrington, S.M.; Carothers, A.D.; Wyllie, A.H.; Sharp, L.; Burn, J.; Liu, B.; Kinzler, K.W.; Vogelstein, B. Cancer Risk Associated with Germline DNA Mismatch Repair Gene Mutations. Hum. Mol. Genet. 1997, 6, 105–110. [Google Scholar] [CrossRef]
  55. Yamamoto, H.; Itoh, F.; Nakamura, H.; Fukushima, H.; Sasaki, S.; Perucho, M.; Imai, K. Genetic and clinical features of human pancreatic ductal adenocarcinomas with widespread microsatellite instability. Cancer Res. 2001, 61, 3139–3144. [Google Scholar] [PubMed]
  56. Wilentz, R.E.; Goggins, M.; Redston, M.; Marcus, V.A.; Adsay, N.V.; Sohn, T.A.; Kadkol, S.S.; Yeo, C.J.; Choti, M.; Zahurak, M.; et al. Genetic, immunohistochemical, and clinical features of medullary carcinoma of the pancreas: A newly described and characterized entity. Am. J. Pathol. 2000, 156, 1641–1651. [Google Scholar] [CrossRef]
  57. Nakata, B.; Wang, Y.Q.; Yashiro, M.; Nishioka, N.; Tanaka, H.; Ohira, M.; Ishikawa, T.; Nishino, H.; Hirakawa, K. Prognostic value of microsatellite instability in resectable pancreatic cancer. Clin. Cancer Res. 2002, 8, 2536–2540. [Google Scholar]
  58. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef]
  59. Galiatsatos, P.; Foulkes, W.D. Familial adenomatous polyposis. Am. J. Gastroenterol. 2006, 101, 385–398. [Google Scholar] [CrossRef]
  60. Giardiello, F.M.; Offerhaus, G.J.; Lee, D.H.; Krush, A.J.; Tersmette, A.C.; Booker, S.V.; Kelley, N.C.; Hamilton, S.R. Increased risk of thyroid and pancreatic carcinoma in familial adenomatous polyposis. Gut 1993, 34, 1394–1396. [Google Scholar] [CrossRef]
  61. Karstensen, J.G.; Bülow, S.; Højen, H.; Jelsig, A.M.; Jespersen, N.; Andersen, K.K.; Wewer, M.D.; Burisch, J.; Pommergaard, H.C. Cancer in Patients with Familial Adenomatous Polyposis: A Nationwide Danish Cohort Study with Matched Controls. Gastroenterology 2023, 165, 573–581.e3. [Google Scholar] [CrossRef]
  62. Hemminki, A.; Markie, D.; Tomlinson, I.; Avizienyte, E.; Roth, S.; Loukola, A.; Bignell, G.; Warren, W.; Aminoff, M.; Höglund, P.; et al. A serine/threonine kinase gene defective in Peutz-Jeghers syndrome. Nature 1998, 391, 184–187. [Google Scholar] [CrossRef] [PubMed]
  63. Jenne, D.E.; Reimann, H.; Nezu, J.; Friedel, W.; Loff, S.; Jeschke, R.; Müller, O.; Back, W.; Zimmer, M. Peutz-Jeghers syndrome is caused by mutations in a novel serine threonine kinase. Nat. Genet. 1998, 18, 38–43. [Google Scholar] [CrossRef] [PubMed]
  64. Giardiello, F.M.; Brensinger, J.D.; Tersmette, A.C.; Goodman, S.N.; Petersen, G.M.; Booker, S.V.; Cruz-Correa, M.; Offerhaus, J.A. Very high risk of cancer in familial Peutz-Jeghers syndrome. Gastroenterology 2000, 119, 1447–1453. [Google Scholar] [CrossRef]
  65. Korsse, S.E.; Harinck, F.; van Lier, M.G.F.; Biermann, K.; Offerhaus, G.J.A.; Krak, N.; Looman, C.W.N.; van Veelen, W.; Kuipers, E.J.; Wagner, A.; et al. Pancreatic cancer risk in Peutz-Jeghers syndrome patients: A large cohort study and implications for surveillance. J. Med. Genet. 2013, 50, 59–64. [Google Scholar] [CrossRef] [PubMed]
  66. de Snoo, F.A.; Bishop, D.T.; Bergman, W.; van Leeuwen, I.; van der Drift, C.; van Nieuwpoort, F.A.; Out-Luiting, C.J.; Vasen, H.F.; ter Huurne, J.A.C.; Frants, R.R.; et al. Increased risk of cancer other than melanoma in CDKN2A founder mutation (p16-Leiden)-positive melanoma families. Clin. Cancer Res. 2008, 14, 7151–7157. [Google Scholar] [CrossRef] [PubMed]
  67. Borg, A.; Sandberg, T.; Nilsson, K.; Johannsson, O.; Klinker, M.; Måsbäck, A.; Westerdahl, J.; Olsson, H.; Ingvar, C. High frequency of multiple melanomas and breast and pancreas carcinomas in CDKN2A mutation-positive melanoma families. J. Natl. Cancer Inst. 2000, 92, 1260–1266. [Google Scholar] [CrossRef] [PubMed]
  68. Goldstein, A.M.; Fraser, M.C.; Struewing, J.P.; Hussussian, C.J.; Ranade, K.; Zametkin, D.P.; Fontaine, L.S.; Organic, S.M.; Dracopoli, N.C.; Clark, W.H. Increased risk of pancreatic cancer in melanoma-prone kindreds with p16INK4 mutations. N. Engl. J. Med. 1995, 333, 970–974. [Google Scholar] [CrossRef] [PubMed]
  69. Lynch, H.T.; Fusaro, R.M.; Lynch, J.F.; Brand, R. Pancreatic cancer and the FAMMM syndrome. Fam. Cancer 2008, 7, 103–112. [Google Scholar] [CrossRef] [PubMed]
  70. Goldstein, A.M.; Chan, M.; Harland, M.; Hayward, N.K.; Demenais, F.; Bishop, D.T.; Azizi, E.; Bergman, W.; Bianchi-Scarra, G.; Bruno, W.; et al. Features associated with germline CDKN2A mutations: A GenoMEL study of melanoma-prone families from three continents. J. Med. Genet. 2007, 44, 99–106. [Google Scholar] [CrossRef] [PubMed]
  71. Lynch, H.T.; Brand, R.E.; Hogg, D.; Deters, C.A.; Fusaro, R.M.; Lynch, J.F.; Liu, L.; Knezetic, J.; Lassam, N.J.; Goggins, M.; et al. Phenotypic variation in eight extended CDKN2A germline mutation familial atypical multiple mole melanoma-pancreatic carcinoma-prone families: The familial atypical mole melanoma-pancreatic carcinoma syndrome. Cancer 2002, 94, 84–96. [Google Scholar] [CrossRef] [PubMed]
  72. Parker, J.F.; Florell, S.R.; Alexander, A.; DiSario, J.A.; Shami, P.J.; Leachman, S.A. Pancreatic carcinoma surveillance in patients with familial melanoma. Arch. Dermatol. 2003, 139, 1019–1025. [Google Scholar] [CrossRef]
  73. Vasen, H.; Ibrahim, I.; Ponce, C.G.; Slater, E.P.; Matthäi, E.; Carrato, A.; Earl, J.; Robbers, K.; van Mil, A.M.; Potjer, T.; 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. J. Clin. Oncol. 2016, 34, 2010–2019. [Google Scholar] [CrossRef]
  74. Schneider, A.; Suman, A.; Rossi, L.; Barmada, M.M.; Beglinger, C.; Parvin, S.; Sattar, S.; Ali, L.; Khan, A.K.A.; Gyr, N.; et al. SPINK1/PSTI mutations are associated with tropical pancreatitis and type II diabetes mellitus in Bangladesh. Gastroenterology 2002, 123, 1026–1030. [Google Scholar] [CrossRef]
  75. LaRusch, J.; Whitcomb, D.C. Genetics of pancreatitis. Curr. Opin. Gastroenterol. 2011, 27, 467–474. [Google Scholar] [CrossRef] [PubMed]
  76. Teich, N.; Rosendahl, J.; Tóth, M.; Mössner, J.; Sahin-Tóth, M. Mutations of human cationic trypsinogen (PRSS1) and chronic pancreatitis. Hum. Mutat. 2006, 27, 721–730. [Google Scholar] [CrossRef] [PubMed]
  77. Sahin-Tóth, M.; Tóth, M. Gain-of-function mutations associated with hereditary pancreatitis enhance autoactivation of human cationic trypsinogen. Biochem. Biophys. Res. Commun. 2000, 278, 286–289. [Google Scholar] [CrossRef]
  78. Sossenheimer, M.J.; Aston, C.E.; Preston, R.A.; Gates, L.K.; Ulrich, C.D.; Martin, S.P.; Zhang, Y.; Gorry, M.C.; Ehrlich, G.D.; Whitcomb, D.C. Clinical characteristics of hereditary pancreatitis in a large family, based on high-risk haplotype. The Midwest Multicenter Pancreatic Study Group (MMPSG). Am. J. Gastroenterol. 1997, 92, 1113–1116. [Google Scholar] [PubMed]
  79. Gukovsky, I.; Li, N.; Todoric, J.; Gukovskaya, A.; Karin, M. Inflammation, Autophagy, and Obesity: Common Features in the Pathogenesis of Pancreatitis and Pancreatic Cancer. Gastroenterology 2013, 144, 1199–1209.e4. [Google Scholar] [CrossRef] [PubMed]
  80. Lowenfels, A.B.; Maisonneuve, P.; DiMagno, E.P.; Elitsur, Y.; Gates, L.K.; Perrault, J.; Whitcomb, D.C. Hereditary pancreatitis and the risk of pancreatic cancer. International Hereditary Pancreatitis Study Group. J. Natl. Cancer Inst. 1997, 89, 442–446. [Google Scholar] [CrossRef]
  81. Lowenfels, A.B.; Maisonneuve, P.; Whitcomb, D.C.; Lerch, M.M.; DiMagno, E.P. Cigarette smoking as a risk factor for pancreatic cancer in patients with hereditary pancreatitis. JAMA 2001, 286, 169–170. [Google Scholar] [CrossRef] [PubMed]
  82. Yu, C.; Kotsimbos, T. Respiratory Infection and Inflammation in Cystic Fibrosis: A Dynamic Interplay among the Host, Microbes, and Environment for the Ages. Int. J. Mol. Sci. 2023, 24, 4052. [Google Scholar] [CrossRef]
  83. Maisonneuve, P.; Marshall, B.C.; Lowenfels, A.B. Risk of pancreatic cancer in patients with cystic fibrosis. Gut 2007, 56, 1327–1328. [Google Scholar] [CrossRef]
  84. Roberts, N.J.; Jiao, Y.; Yu, J.; Kopelovich, L.; Petersen, G.M.; Bondy, M.; Gallinger, S.; Schwartz, A.G.; Syngal, S.; Cote, M.L.; et al. ATM mutations in hereditary pancreatic cancer patients. Cancer Discov. 2012, 2, 41–46. [Google Scholar] [CrossRef]
  85. Swift, M.; Reitnauer, P.J.; Morrell, D.; Chase, C.L. Breast and other cancers in families with ataxia-telangiectasia. N. Engl. J. Med. 1987, 316, 1289–1294. [Google Scholar] [CrossRef]
  86. Taylor, A.M.R.; Byrd, P.J. Molecular pathology of ataxia telangiectasia. J. Clin. Pathol. 2005, 58, 1009–1015. [Google Scholar] [CrossRef] [PubMed]
  87. Goggins, M.; Overbeek, K.A.; Brand, R.; Syngal, S.; Del Chiaro, M.; Bartsch, D.K.; Bassi, C.; Carrato, A.; Farrell, J.; Fishman, E.K.; 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. [Google Scholar] [CrossRef]
  88. Rodriguez, J.R.; Salvia, R.; Crippa, S.; Warshaw, A.L.; Bassi, C.; Falconi, M.; Thayer, S.P.; Lauwers, G.Y.; Capelli, P.; Mino-Kenudson, M.; et al. Branch-Duct Intraductal Papillary Mucinous Neoplasms: Observations in 145 Patients Who Underwent Resection. Gastroenterology 2007, 133, 72–310. [Google Scholar] [CrossRef] [PubMed]
  89. Salvia, R.; Castillo, C.F.; Bassi, C.; Thayer, S.P.; Falconi, M.; Mantovani, W.; Pederzoli, P.; Warshaw, A.L. Main-Duct Intraductal Papillary Mucinous Neoplasms of the Pancreas. Ann. Surg. 2004, 239, 678–687. [Google Scholar] [CrossRef] [PubMed]
  90. van Huijgevoort, N.C.M.; del Chiaro, M.; Wolfgang, C.L.; van Hooft, J.E.; Besselink, M.G. Diagnosis and management of pancreatic cystic neoplasms: Current evidence and guidelines. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 676–689. [Google Scholar] [CrossRef]
  91. Brugge, W.R. Role of endoscopic ultrasound in the diagnosis of cystic lesions of the pancreas. Pancreatology 2001, 1, 637–640. [Google Scholar] [CrossRef]
  92. Zhang, H.; Cao, Y.; Ren, S.; Guo, K.; Zhang, Y.; Lin, T.; Wang, Y.; Chen, X.; Wang, Z. Threshold of Main Pancreatic Duct Diameter in Identifying Malignant Intraductal Papillary Mucinous Neoplasm by Magnetic Resonance Imaging. Technol. Cancer Res. Treat. 2023, 22, 15330338231170942. [Google Scholar] [CrossRef]
  93. Ohno, E.; Hirooka, Y.; Kawashima, H.; Ishikawa, T.; Kanamori, A.; Ishikawa, H.; Sasaki, Y.; Nonogaki, K.; Hara, K.; Hashimoto, S.; et al. Natural history of pancreatic cystic lesions: A multicenter prospective observational study for evaluating the risk of pancreatic cancer. J. Gastroenterol. Hepatol. 2018, 33, 320–328. [Google Scholar] [CrossRef]
  94. Shimizu, Y.; Yamaue, H.; Maguchi, H.; Yamao, K.; Hirono, S.; Osanai, M.; Hijioka, S.; Hosoda, W.; Nakamura, Y.; Shinohara, T.; et al. Predictors of malignancy in intraductal papillary mucinous neoplasm of the pancreas: Analysis of 310 pancreatic resection patients at multiple high-volume centers. Pancreas 2013, 42, 883–888. [Google Scholar] [CrossRef]
  95. Lafemina, J.; Katabi, N.; Klimstra, D.; Correa-Gallego, C.; Gaujoux, S.; Kingham, T.P.; Dematteo, R.P.; Fong, Y.; D’Angelica, M.I.; Jarnagin, W.R.; et al. Malignant progression in IPMN: A cohort analysis of patients initially selected for resection or observation. Ann. Surg. Oncol. 2013, 20, 440–447. [Google Scholar] [CrossRef]
  96. Tanaka, M. Intraductal Papillary Mucinous Neoplasm of the Pancreas as the Main Focus for Early Detection of Pancreatic Adenocarcinoma. Pancreas 2018, 47, 544. [Google Scholar] [CrossRef] [PubMed]
  97. Scherer, J.A.; Gebhard, R.; Firkins, S.A.; Shah, Z.K.; Urbina Andersson, I.K.; Barker, S.J.; Fiorillo, L.E.; Hollander, E.; Shaheen, N.; Koay, E.J.; et al. Lower Interobserver Reliability for Nondimensional Intracystic Features among Abdominal Radiologists for Characterizing Intraductal Papillary Mucinous Neoplasms Using Magnetic Resonance Imaging. Pancreas 2022, 51, 1225–1230. [Google Scholar] [CrossRef]
  98. Li, H.; Feng, X.; Gao, F.; Chen, Q.; Linghu, E. Diagnostic value of EUS-guided SF6 pancreatography for pancreatic cystic lesions on cyst communication with the pancreatic duct. Endosc. Ultrasound 2023, 12, 245–250. [Google Scholar] [CrossRef] [PubMed]
  99. Lang, M.; Spektor, A.-M.; Hielscher, T.; Hoppner, J.; Glatting, F.M.; Bicu, F.; Hackert, T.; Heger, U.; Pausch, T.; Gutjahr, E.; et al. Static and Dynamic 68Ga-FAPI PET/CT for the Detection of Malignant Transformation of Intraductal Papillary Mucinous Neoplasia of the Pancreas. J. Nucl. Med. 2023, 64, 244–251. [Google Scholar] [CrossRef]
  100. Levink, I.; Bruno, M.; Cahen, D. Management of Intraductal Papillary Mucinous Neoplasms: Controversies in Guidelines and Future Perspectives. Curr. Treat. Options Gastroenterol. 2018, 16, 316–332. [Google Scholar] [CrossRef]
  101. Gentiluomo, M.; Corradi, C.; Arcidiacono, P.G.; Crippa, S.; Falconi, M.; Belfiori, G.; Farinella, R.; Apadula, L.; Lauri, G.; Bina, N.; et al. Role of pancreatic ductal adenocarcinoma risk factors in intraductal papillary mucinous neoplasm progression. Front. Oncol. 2023, 13, 1172606. [Google Scholar] [CrossRef] [PubMed]
  102. Sofi, A.A.; Ahmad, S.; Peerzada, M.; Hackett, L. Diabetes mellitus and the risk of progression or malignancy of pancreatic cystic neoplasms in patients undergoing surveillance: A systematic review and meta-analysis. Pancreatology 2022, 22, 1195–1201. [Google Scholar] [CrossRef] [PubMed]
  103. Schweber, A.B.; Brooks, C.; Agarunov, E.; Sethi, A.; Poneros, J.M.; Schrope, B.A.; Kluger, M.D.; Chabot, J.A.; Gonda, T.A. New onset diabetes predicts progression of low risk pancreatic mucinous cysts. Pancreatology 2020, 20, 1755–1763. [Google Scholar] [CrossRef] [PubMed]
  104. Capurso, G.; Boccia, S.; Salvia, R.; Del Chiaro, M.; Frulloni, L.; Arcidiacono, P.G.; Zerbi, A.; Manta, R.; Fabbri, C.; Ventrucci, M.; et al. Risk Factors for Intraductal Papillary Mucinous Neoplasm (IPMN) of the Pancreas: A Multicentre Case–Control Study. Am. J. Gastroenterol. 2013, 108, 1003. [Google Scholar] [CrossRef]
  105. Huang, X.; Guo, T.; Zhang, Z.; Cai, M.; Guo, X.; Zhang, J.; Yu, Y. Prediction of malignant intraductal papillary mucinous neoplasm: A nomogram based on clinical information and radiological outcomes. Cancer Med. 2023, 12, 16958–16971. [Google Scholar] [CrossRef]
  106. Gausman, V.; Kandel, P.; Van Riet, P.A.; Moris, M.; Kayal, M.; Do, C.; Poneros, J.M.; Sethi, A.; Gress, F.G.; Schrope, B.A.; et al. Predictors of Progression among Low-Risk Intraductal Papillary Mucinous Neoplasms in a Multicenter Surveillance Cohort. Pancreas 2018, 47, 471–476. [Google Scholar] [CrossRef] [PubMed]
  107. Armstrong, M.T.; Saadat, L.V.; Chou, J.F.; Gönen, M.; Balachandran, V.P.; D’Angelica, M.I.; Drebin, J.A.; Flood, J.; Jarnagin, W.R.; Kingham, T.P.; et al. Risk Factors for Progression in Patients Undergoing Surveillance for Pancreatic Cysts. Ann. Surg. 2024, 279, 119–124. [Google Scholar] [CrossRef] [PubMed]
  108. Levink, I.J.M.; Jaarsma, S.C.; Koopmann, B.D.M.; van Riet, P.A.; Overbeek, K.A.; Meziani, J.; Sprij, M.L.J.A.; Casadei, R.; Ingaldi, C.; Polkowski, M.; et al. The additive value of CA19.9 monitoring in a pancreatic cyst surveillance program. United Eur. Gastroenterol. J. 2023, 11, 601–611. [Google Scholar] [CrossRef] [PubMed]
  109. Crippa, S.; Pezzilli, R.; Bissolati, M.; Capurso, G.; Romano, L.; Brunori, M.P.; Calculli, L.; Tamburrino, D.; Piccioli, A.; Ruffo, G.; et al. Active Surveillance Beyond 5 Years Is Required for Presumed Branch-Duct Intraductal Papillary Mucinous Neoplasms Undergoing Non-Operative Management. Am. J. Gastroenterol. 2017, 112, 1153–1161. [Google Scholar] [CrossRef]
  110. Pergolini, I.; Sahora, K.; Ferrone, C.R.; Morales-Oyarvide, V.; Wolpin, B.M.; Mucci, L.A.; Brugge, W.R.; Mino-Kenudson, M.; Patino, M.; Sahani, D.V.; et al. Long-term Risk of Pancreatic Malignancy in Patients with Branch Duct Intraductal Papillary Mucinous Neoplasm in a Referral Center. Gastroenterology 2017, 153, 1284–1294.e1. [Google Scholar] [CrossRef]
  111. Marinelli, V.; Secchettin, E.; Andrianello, S.; Moretti, C.; Donvito, S.; Marchegiani, G.; Esposito, A.; Casetti, L.; Salvia, R. Psychological distress in patients under surveillance for intraductal papillary mucinous neoplasms of the pancreas: The “Sword of Damocles” effect calls for an integrated medical and psychological approach a prospective analysis. Pancreatology 2020, 20, 505–510. [Google Scholar] [CrossRef]
  112. Duell, E.J. Epidemiology and potential mechanisms of tobacco smoking and heavy alcohol consumption in pancreatic cancer. Mol. Carcinog. 2012, 51, 40–52. [Google Scholar] [CrossRef] [PubMed]
  113. Underwood, P.W.; Zhang, D.Y.; Cameron, M.E.; Gerber, M.H.; Delitto, D.; Maduka, M.U.; Cooper, K.J.; Han, S.; Hughes, S.J.; Judge, S.M.; et al. Nicotine Induces IL-8 Secretion from Pancreatic Cancer Stroma and Worsens Cancer-Induced Cachexia. Cancers 2020, 12, 329. [Google Scholar] [CrossRef]
  114. Iodice, S.; Gandini, S.; Maisonneuve, P.; Lowenfels, A.B. Tobacco and the risk of pancreatic cancer: A review and meta-analysis. Langenbecks Arch. Surg. 2008, 393, 535–545. [Google Scholar] [CrossRef]
  115. Bosetti, C.; Lucenteforte, E.; Silverman, D.T.; Petersen, G.; Bracci, P.M.; Ji, B.T.; Negri, E.; Li, D.; Risch, H.A.; Olson, S.H.; et al. Cigarette smoking and pancreatic cancer: An analysis from the International Pancreatic Cancer Case-Control Consortium (Panc4). Ann. Oncol. 2012, 23, 1880–1888. [Google Scholar] [CrossRef] [PubMed]
  116. Anderson, M.A.; Zolotarevsky, E.; Cooper, K.L.; Sherman, S.; Shats, O.; Whitcomb, D.C.; Lynch, H.T.; Ghiorzo, P.; Rubinstein, W.S.; Vogel, K.J.; et al. Alcohol and tobacco lower the age of presentation in sporadic pancreatic cancer in a dose-dependent manner: A multicenter study. Am. J. Gastroenterol. 2012, 107, 1730–1739. [Google Scholar] [CrossRef]
  117. Lugo, A.; Peveri, G.; Bosetti, C.; Bagnardi, V.; Crippa, A.; Orsini, N.; Rota, M.; Gallus, S. Strong excess risk of pancreatic cancer for low frequency and duration of cigarette smoking: A comprehensive review and meta-analysis. Eur. J. Cancer 2018, 104, 117–126. [Google Scholar] [CrossRef] [PubMed]
  118. Janssen, F. Similarities and Differences between Sexes and Countries in the Mortality Imprint of the Smoking Epidemic in 34 Low-Mortality Countries, 1950–2014. Nicotine Tob. Res. 2019, 22, 1210–1220. [Google Scholar] [CrossRef] [PubMed]
  119. Janssen, F.; El Gewily, S.; Bardoutsos, A. Smoking epidemic in Europe in the 21st century. Tob. Control 2021, 30, 523–529. [Google Scholar] [CrossRef] [PubMed]
  120. Tyczynski, J.E.; Bray, F.; Aareleid, T.; Dalmas, M.; Kurtinaitis, J.; Plesko, I.; Pompe-Kirn, V.; Stengrevics, A.; Parkin, D.M. Lung cancer mortality patterns in selected Central, Eastern and Southern European countries. Int. J. Cancer 2004, 109, 598–610. [Google Scholar] [CrossRef] [PubMed]
  121. Bao, Y.; Giovannucci, E.; Fuchs, C.S.; Michaud, D.S. Passive Smoking and Pancreatic Cancer in Women: A Prospective Cohort Study. Cancer Epidemiol. Biomark. Prev. 2009, 18, 2292–2296. [Google Scholar] [CrossRef] [PubMed]
  122. Zhou, J.; Wellenius, G.A.; Michaud, D.S. Environmental tobacco smoke and the risk of pancreatic cancer among non-smokers: A meta-analysis. Occup. Environ. Med. 2012, 69, 853–857. [Google Scholar] [CrossRef] [PubMed]
  123. Bao, W.; Xu, G.; Lu, J.; Snetselaar, L.G.; Wallace, R.B. Changes in Electronic Cigarette Use among Adults in the United States, 2014–2016. JAMA 2018, 319, 2039–2041. [Google Scholar] [CrossRef] [PubMed]
  124. McMillen, R.C.; Gottlieb, M.A.; Shaefer, R.M.W.; Winickoff, J.P.; Klein, J.D. Trends in Electronic Cigarette Use among U.S. Adults: Use is Increasing in Both Smokers and Nonsmokers. Nicotine Tob. Res. 2015, 17, 1195–1202. [Google Scholar] [CrossRef]
  125. Soneji, S.S.; Sung, H.-Y.; Primack, B.A.; Pierce, J.P.; Sargent, J.D. Quantifying population-level health benefits and harms of e-cigarette use in the United States. PLoS ONE 2018, 13, e0193328. [Google Scholar] [CrossRef] [PubMed]
  126. Stoklosa, M.; Cahn, Z.; Liber, A.; Nargis, N.; Drope, J. Effect of IQOS introduction on cigarette sales: Evidence of decline and replacement. Tob. Control 2020, 29, 381–387. [Google Scholar] [CrossRef] [PubMed]
  127. Araghi, M.; Rosaria Galanti, M.; Lundberg, M.; Lager, A.; Engström, G.; Alfredsson, L.; Knutsson, A.; Norberg, M.; Sund, M.; Wennberg, P.; et al. Use of moist oral snuff (snus) and pancreatic cancer: Pooled analysis of nine prospective observational studies. Int. J. Cancer 2017, 141, 687–693. [Google Scholar] [CrossRef] [PubMed]
  128. Clarke, E.; Thompson, K.; Weaver, S.; Thompson, J.; O’Connell, G. Snus: A compelling harm reduction alternative to cigarettes. Harm Reduct. J. 2019, 16, 62. [Google Scholar] [CrossRef]
  129. Kosmider, L.; Sobczak, A.; Fik, M.; Knysak, J.; Zaciera, M.; Kurek, J.; Goniewicz, M.L. Carbonyl compounds in electronic cigarette vapors: Effects of nicotine solvent and battery output voltage. Nicotine Tob. Res. 2014, 16, 1319–1326. [Google Scholar] [CrossRef] [PubMed]
  130. Stephens, W.E. Comparing the cancer potencies of emissions from vapourised nicotine products including e-cigarettes with those of tobacco smoke. Tob. Control 2018, 27, 10–17. [Google Scholar] [CrossRef] [PubMed]
  131. Schober, W.; Szendrei, K.; Matzen, W.; Osiander-Fuchs, H.; Heitmann, D.; Schettgen, T.; Jörres, R.A.; Fromme, H. Use of electronic cigarettes (e-cigarettes) impairs indoor air quality and increases FeNO levels of e-cigarette consumers. Int. J. Hyg. Environ. Health 2014, 217, 628–637. [Google Scholar] [CrossRef] [PubMed]
  132. Lerner, C.A.; Sundar, I.K.; Yao, H.; Gerloff, J.; Ossip, D.J.; McIntosh, S.; Robinson, R.; Rahman, I. Vapors produced by electronic cigarettes and e-juices with flavorings induce toxicity, oxidative stress, and inflammatory response in lung epithelial cells and in mouse lung. PLoS ONE 2015, 10, e0116732. [Google Scholar] [CrossRef] [PubMed]
  133. Rodrigo, G.; Jaccard, G.; Tafin Djoko, D.; Korneliou, A.; Esposito, M.; Belushkin, M. Cancer potencies and margin of exposure used for comparative risk assessment of heated tobacco products and electronic cigarettes aerosols with cigarette smoke. Arch. Toxicol. 2021, 95, 283–298. [Google Scholar] [CrossRef]
  134. Schaller, J.-P.; Keller, D.; Poget, L.; Pratte, P.; Kaelin, E.; McHugh, D.; Cudazzo, G.; Smart, D.; Tricker, A.R.; Gautier, L.; et al. Evaluation of the Tobacco Heating System 2.2. Part 2: Chemical composition, genotoxicity, cytotoxicity, and physical properties of the aerosol. Regul. Toxicol. Pharmacol. 2016, 81, S27–S47. [Google Scholar] [CrossRef]
  135. Li, X.; Luo, Y.; Jiang, X.; Zhang, H.; Zhu, F.; Hu, S.; Hou, H.; Hu, Q.; Pang, Y. Chemical Analysis and Simulated Pyrolysis of Tobacco Heating System 2.2 Compared to Conventional Cigarettes. Nicotine Tob. Res. 2019, 21, 111–118. [Google Scholar] [CrossRef] [PubMed]
  136. Diabetes. Available online: https://www.who.int/news-room/fact-sheets/detail/diabetes (accessed on 14 April 2023).
  137. Wang, F.; Herrington, M.; Larsson, J.; Permert, J. The relationship between diabetes and pancreatic cancer. Mol. Cancer 2003, 2, 4. [Google Scholar] [CrossRef] [PubMed]
  138. Pannala, R.; Leirness, J.B.; Bamlet, W.R.; Basu, A.; Petersen, G.M.; Chari, S.T. Prevalence and Clinical Profile of Pancreatic Cancer-associated Diabetes mellitus. Gastroenterology 2008, 134, 981–987. [Google Scholar] [CrossRef] [PubMed]
  139. Pereira, S.P.; Oldfield, L.; Ney, A.; Hart, P.A.; Keane, M.G.; Pandol, S.J.; Li, D.; Greenhalf, W.; Jeon, C.Y.; Koay, E.J.; et al. Early detection of pancreatic cancer. Lancet Gastroenterol. Hepatol. 2020, 5, 698–710. [Google Scholar] [CrossRef] [PubMed]
  140. Chari, S.T.; Klee, G.G.; Miller, L.J.; Raimondo, M.; DiMagno, E.P. Islet amyloid polypeptide is not a satisfactory marker for detecting pancreatic cancer. Gastroenterology 2001, 121, 640–645. [Google Scholar] [CrossRef] [PubMed]
  141. Chari, S.T.; Leibson, C.L.; Rabe, K.G.; Timmons, L.J.; Ransom, J.; de Andrade, M.; Petersen, G.M. Pancreatic Cancer-associated Diabetes Mellitus: Prevalence and Temporal Association with Diagnosis of Cancer. Gastroenterology 2008, 134, 95–101. [Google Scholar] [CrossRef]
  142. Cui, Y.; Andersen, D.K. Diabetes and pancreatic cancer. Endocr. Relat. Cancer 2012, 19, F9–F26. [Google Scholar] [CrossRef] [PubMed]
  143. White, M.J.; Sheka, A.C.; LaRocca, C.J.; Irey, R.L.; Ma, S.; Wirth, K.M.; Benner, A.; Denbo, J.W.; Jensen, E.H.; Ankeny, J.S.; et al. The association of new-onset diabetes with subsequent diagnosis of pancreatic cancer—Novel use of a large administrative database. J. Public Health 2022, 45, e266–e274. [Google Scholar] [CrossRef]
  144. Dankner, R.; Boffetta, P.; Balicer, R.D.; Boker, L.K.; Sadeh, M.; Berlin, A.; Olmer, L.; Goldfracht, M.; Freedman, L.S. Time-Dependent Risk of Cancer after a Diabetes Diagnosis in a Cohort of 2.3 Million Adults. Am. J. Epidemiol. 2016, 183, 1098–1106. [Google Scholar] [CrossRef]
  145. Li, D. Diabetes and pancreatic cancer. Mol. Carcinog. 2012, 51, 64–74. [Google Scholar] [CrossRef]
  146. Li, D.; Tang, H.; Hassan, M.M.; Holly, E.A.; Bracci, P.M.; Silverman, D.T. Diabetes and risk of pancreatic cancer: A pooled analysis of three large case-control studies. Cancer Causes Control 2011, 22, 189–197. [Google Scholar] [CrossRef] [PubMed]
  147. Sharma, A.; Kandlakunta, H.; Nagpal, S.J.S.; Feng, Z.; Hoos, W.; Petersen, G.M.; Chari, S.T. Model to Determine Risk of Pancreatic Cancer in Patients with New-Onset Diabetes. Gastroenterology 2018, 155, 730–739.e3. [Google Scholar] [CrossRef] [PubMed]
  148. Boursi, B.; Patalon, T.; Webb, M.; Margalit, O.; Beller, T.; Yang, Y.-X.; Chodick, G. Validation of the Enriching New-Onset Diabetes for Pancreatic Cancer Model: A Retrospective Cohort Study Using Real-World Data. Pancreas 2022, 51, 196–199. [Google Scholar] [CrossRef]
  149. Chen, W.; Butler, R.K.; Lustigova, E.; Chari, S.T.; Wu, B.U. Validation of the Enriching New-Onset Diabetes for Pancreatic Cancer Model in a Diverse and Integrated Healthcare Setting. Dig. Dis. Sci. 2021, 66, 78–87. [Google Scholar] [CrossRef] [PubMed]
  150. Khan, S.; Safarudin, R.F.; Kupec, J.T. Validation of the ENDPAC model: Identifying new-onset diabetics at risk of pancreatic cancer. Pancreatology 2021, 21, 550–555. [Google Scholar] [CrossRef] [PubMed]
  151. Mellenthin, C.; Balaban, V.D.; Dugic, A.; Cullati, S. Risk Factors for Pancreatic Cancer in Patients with New-Onset Diabetes: A Systematic Review and Meta-Analysis. Cancers 2022, 14, 4684. [Google Scholar] [CrossRef] [PubMed]
  152. Deng, Z.; Gu, Y.; Hou, X.; Zhang, L.; Bao, Y.; Hu, C.; Jia, W. Association between uric acid, cancer incidence and mortality in patients with type 2 diabetes: Shanghai diabetes registry study. Diabetes/Metab. Res. Rev. 2016, 32, 325–332. [Google Scholar] [CrossRef]
  153. Batty, G.D.; Shipley, M.J.; Marmot, M.; Smith, G.D. Diabetes status and post-load plasma glucose concentration in relation to site-specific cancer mortality: Findings from the original Whitehall study. Cancer Causes Control 2004, 15, 873–881. [Google Scholar] [CrossRef] [PubMed]
  154. Smith, G.D.; Egger, M.; Shipley, M.J.; Marmot, M.G. Post-challenge glucose concentration, impaired glucose tolerance, diabetes, and cancer mortality in men. Am. J. Epidemiol. 1992, 136, 1110–1114. [Google Scholar] [CrossRef]
  155. Gapstur, S.M.; Gann, P.H.; Lowe, W.; Liu, K.; Colangelo, L.; Dyer, A. Abnormal glucose metabolism and pancreatic cancer mortality. JAMA 2000, 283, 2552–2558. [Google Scholar] [CrossRef]
  156. Bosetti, C.; Rosato, V.; Li, D.; Silverman, D.; Petersen, G.M.; Bracci, P.M.; Neale, R.E.; Muscat, J.; Anderson, K.; Gallinger, S.; et al. Diabetes, antidiabetic medications, and pancreatic cancer risk: An analysis from the International Pancreatic Cancer Case-Control Consortium. Ann. Oncol. 2014, 25, 2065–2072. [Google Scholar] [CrossRef]
  157. Ding, X.Z.; Fehsenfeld, D.M.; Murphy, L.O.; Permert, J.; Adrian, T.E. Physiological concentrations of insulin augment pancreatic cancer cell proliferation and glucose utilization by activating MAP kinase, PI3 kinase and enhancing GLUT-1 expression. Pancreas 2000, 21, 310–320. [Google Scholar] [CrossRef]
  158. Stoeltzing, O.; Liu, W.; Reinmuth, N.; Fan, F.; Parikh, A.A.; Bucana, C.D.; Evans, D.B.; Semenza, G.L.; Ellis, L.M. Regulation of hypoxia-inducible factor-1alpha, vascular endothelial growth factor, and angiogenesis by an insulin-like growth factor-I receptor autocrine loop in human pancreatic cancer. Am. J. Pathol. 2003, 163, 1001–1011. [Google Scholar] [CrossRef]
  159. Zeng, H.; Datta, K.; Neid, M.; Li, J.; Parangi, S.; Mukhopadhyay, D. Requirement of different signaling pathways mediated by insulin-like growth factor-I receptor for proliferation, invasion, and VPF/VEGF expression in a pancreatic carcinoma cell line. Biochem. Biophys. Res. Commun. 2003, 302, 46–55. [Google Scholar] [CrossRef]
  160. Tao, L.V.; Topham, J.T.; Karasinska, J.M.; Tsang, E.S.; Metcalfe, A.; Ali, H.; Ashforth, D.; Goodwin, R.; Tang, P.A.; Bathe, O.F.; et al. Abstract B066: Elucidating the role of insulin receptor isoform expression in metastatic pancreatic ductal adenocarcinoma. Cancer Res. 2022, 82, B066. [Google Scholar] [CrossRef]
  161. Schneider, M.B.; Matsuzaki, H.; Haorah, J.; Ulrich, A.; Standop, J.; Ding, X.Z.; Adrian, T.E.; Pour, P.M. Prevention of pancreatic cancer induction in hamsters by metformin. Gastroenterology 2001, 120, 1263–1270. [Google Scholar] [CrossRef]
  162. Decensi, A.; Puntoni, M.; Goodwin, P.; Cazzaniga, M.; Gennari, A.; Bonanni, B.; Gandini, S. Metformin and cancer risk in diabetic patients: A systematic review and meta-analysis. Cancer Prev. Res. 2010, 3, 1451–1461. [Google Scholar] [CrossRef]
  163. Li, D.; Yeung, S.-C.J.; Hassan, M.M.; Konopleva, M.; Abbruzzese, J.L. Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology 2009, 137, 482–488. [Google Scholar] [CrossRef]
  164. Zhang, P.; Li, H.; Tan, X.; Chen, L.; Wang, S. Association of metformin use with cancer incidence and mortality: A meta-analysis. Cancer Epidemiol. 2013, 37, 207–218. [Google Scholar] [CrossRef]
  165. Kirpichnikov, D.; McFarlane, S.I.; Sowers, J.R. Metformin: An update. Ann. Intern. Med. 2002, 137, 25–33. [Google Scholar] [CrossRef]
  166. Dowling, R.J.O.; Zakikhani, M.; Fantus, I.G.; Pollak, M.; Sonenberg, N. Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells. Cancer Res. 2007, 67, 10804–10812. [Google Scholar] [CrossRef] [PubMed]
  167. Carstensen, B.; Read, S.H.; Friis, S.; Sund, R.; Keskimäki, I.; Svensson, A.-M.; Ljung, R.; Wild, S.H.; Kerssens, J.J.; Harding, J.L.; et al. Cancer incidence in persons with type 1 diabetes: A five-country study of 9000 cancers in type 1 diabetic individuals. Diabetologia 2016, 59, 980–988. [Google Scholar] [CrossRef] [PubMed]
  168. Stevens, R.J.; Roddam, A.W.; Beral, V. Pancreatic cancer in type 1 and young-onset diabetes: Systematic review and meta-analysis. Br. J. Cancer 2007, 96, 507–509. [Google Scholar] [CrossRef] [PubMed]
  169. Wideroff, L.; Gridley, G.; Mellemkjaer, L.; Chow, W.H.; Linet, M.; Keehn, S.; Borch-Johnsen, K.; Olsen, J.H. Cancer incidence in a population-based cohort of patients hospitalized with diabetes mellitus in Denmark. J. Natl. Cancer Inst. 1997, 89, 1360–1365. [Google Scholar] [CrossRef] [PubMed]
  170. Obesity and Overweight. Available online: https://www.who.int/news-room/fact-sheets/detail/obesity-and-overweight (accessed on 14 April 2023).
  171. Larsson, S.C.; Orsini, N.; Wolk, A. Body mass index and pancreatic cancer risk: A meta-analysis of prospective studies. Int. J. Cancer 2007, 120, 1993–1998. [Google Scholar] [CrossRef] [PubMed]
  172. Stocks, T.; Rapp, K.; Bjørge, T.; Manjer, J.; Ulmer, H.; Selmer, R.; Lukanova, A.; Johansen, D.; Concin, H.; Tretli, S.; et al. Blood Glucose and Risk of Incident and Fatal Cancer in the Metabolic Syndrome and Cancer Project (Me-Can): Analysis of Six Prospective Cohorts. PLoS Med. 2009, 6, e1000201. [Google Scholar] [CrossRef] [PubMed]
  173. Silverman, D.T.; Swanson, C.A.; Gridley, G.; Wacholder, S.; Greenberg, R.S.; Brown, L.M.; Hayes, R.B.; Swanson, G.M.; Schoenberg, J.B.; Pottern, L.M.; et al. Dietary and nutritional factors and pancreatic cancer: A case-control study based on direct interviews. J. Natl. Cancer Inst. 1998, 90, 1710–1719. [Google Scholar] [CrossRef] [PubMed]
  174. Malone, J.I.; Hansen, B.C. Does obesity cause type 2 diabetes mellitus (T2DM)? Or is it the opposite? Pediatr. Diabetes 2019, 20, 5–9. [Google Scholar] [CrossRef] [PubMed]
  175. Incio, J.; Liu, H.; Suboj, P.; Chin, S.M.; Chen, I.X.; Pinter, M.; Ng, M.R.; Nia, H.T.; Grahovac, J.; Kao, S.; et al. Obesity-Induced Inflammation and Desmoplasia Promote Pancreatic Cancer Progression and Resistance to Chemotherapy. Cancer Discov. 2016, 6, 852–869. [Google Scholar] [CrossRef]
  176. Abate, N. Insulin resistance and obesity. The role of fat distribution pattern. Diabetes Care 1996, 19, 292–294. [Google Scholar] [CrossRef]
  177. Butler, A.E.; Galasso, R.; Matveyenko, A.; Rizza, R.A.; Dry, S.; Butler, P.C. Pancreatic duct replication is increased with obesity and type 2 diabetes in humans. Diabetologia 2010, 53, 21–26. [Google Scholar] [CrossRef] [PubMed]
  178. Stolzenberg-Solomon, R.Z.; Adams, K.; Leitzmann, M.; Schairer, C.; Michaud, D.S.; Hollenbeck, A.; Schatzkin, A.; Silverman, D.T. Adiposity, physical activity, and pancreatic cancer in the National Institutes of Health-AARP Diet and Health Cohort. Am. J. Epidemiol. 2008, 167, 586–597. [Google Scholar] [CrossRef]
  179. Arslan, A.A.; Helzlsouer, K.J.; Kooperberg, C.; Shu, X.-O.; Steplowski, E.; Bueno-de-Mesquita, H.B.; Fuchs, C.S.; Gross, M.D.; Jacobs, E.J.; LaCroix, A.Z.; 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. [Google Scholar] [CrossRef]
  180. Sreedhar, U.L.; DeSouza, S.V.; Park, B.; Petrov, M.S. A Systematic Review of Intra-pancreatic Fat Deposition and Pancreatic Carcinogenesis. J. Gastrointest. Surg. 2020, 24, 2560–2569. [Google Scholar] [CrossRef] [PubMed]
  181. Kashiwagi, K.; Seino, T.; Fukuhara, S.; Minami, K.; Horibe, M.; Iwasaki, E.; Takaishi, H.; Itoh, K.; Sugino, Y.; Inoue, N.; et al. Pancreatic Fat Content Detected by Computed Tomography and Its Significant Relationship with Intraductal Papillary Mucinous Neoplasm. Pancreas 2018, 47, 1087–1092. [Google Scholar] [CrossRef] [PubMed]
  182. DiPietro, L.; Seeman, T.E.; Stachenfeld, N.S.; Katz, L.D.; Nadel, E.R. Moderate-Intensity Aerobic Training Improves Glucose Tolerance in Aging Independent of Abdominal Adiposity. J. Am. Geriatr. Soc. 1998, 46, 875–879. [Google Scholar] [CrossRef] [PubMed]
  183. Djuric, Z. Obesity-associated cancer risk: The role of intestinal microbiota in the etiology of the host proinflammatory state. Transl. Res. 2017, 179, 155–167. [Google Scholar] [CrossRef] [PubMed]
  184. Ren, Z.; Jiang, J.; Xie, H.; Li, A.; Lu, H.; Xu, S.; Zhou, L.; Zhang, H.; Cui, G.; Chen, X.; et al. Gut microbial profile analysis by MiSeq sequencing of pancreatic carcinoma patients in China. Oncotarget 2017, 8, 95176–95191. [Google Scholar] [CrossRef] [PubMed]
  185. Stolzenberg-Solomon, R.Z.; Pietinen, P.; Taylor, P.R.; Virtamo, J.; Albanes, D. Prospective Study of Diet and Pancreatic Cancer in Male Smokers. Am. J. Epidemiol. 2002, 155, 783–792. [Google Scholar] [CrossRef]
  186. Casari, I.; Falasca, M. Diet and Pancreatic Cancer Prevention. Cancers 2015, 7, 2309–2317. [Google Scholar] [CrossRef]
  187. Garcia, C.C.; Lawres, L.; Agabiti, S.; Singh, J.; Tong, A.; Venkat, A.; Burkhardt, D.B.; Cardone, R.; Kibbey, R.G.; Krishnaswamy, S.; et al. Abstract C050: Elucidating mechanisms of endocrine-exocrine signaling in obesity-driven pancreatic cancer. Cancer Res. 2022, 82, C050. [Google Scholar] [CrossRef]
  188. Jarosz, M.; Sekuła, W.; Rychlik, E. Influence of Diet and Tobacco Smoking on Pancreatic Cancer Incidence in Poland in 1960–2008. Gastroenterol. Res. Pract. 2012, 2012, 682156. [Google Scholar] [CrossRef] [PubMed]
  189. Paluszkiewicz, P.; Smolińska, K.; Dębińska, I.; Turski, W.A. Main dietary compounds and pancreatic cancer risk. The quantitative analysis of case-control and cohort studies. Cancer Epidemiol. 2012, 36, 60–67. [Google Scholar] [CrossRef] [PubMed]
  190. Rossi, M.; Lugo, A.; Lagiou, P.; Zucchetto, A.; Polesel, J.; Serraino, D.; Negri, E.; Trichopoulos, D.; Vecchia, C.L. Proanthocyanidins and other flavonoids in relation to pancreatic cancer: A case–control study in Italy. Ann. Oncol. 2012, 23, 1488–1493. [Google Scholar] [CrossRef] [PubMed]
  191. Heinen, M.M.; Verhage, B.A.J.; Goldbohm, R.A.; van den Brandt, P.A. Intake of vegetables, fruits, carotenoids and vitamins C and E and pancreatic cancer risk in The Netherlands Cohort Study. Int. J. Cancer 2012, 130, 147–158. [Google Scholar] [CrossRef]
  192. Falasca, M.; Casari, I.; Maffucci, T. Cancer chemoprevention with nuts. J. Natl. Cancer Inst. 2014, 106, dju238. [Google Scholar] [CrossRef]
  193. Awad, A.B.; Fink, C.S. Phytosterols as anticancer dietary components: Evidence and mechanism of action. J. Nutr. 2000, 130, 2127–2130. [Google Scholar] [CrossRef]
  194. Liu, R.H. Dietary bioactive compounds and their health implications. J. Food Sci. 2013, 78 (Suppl. 1), A18–A25. [Google Scholar] [CrossRef] [PubMed]
  195. Bao, Y.; Han, J.; Hu, F.B.; Giovannucci, E.L.; Stampfer, M.J.; Willett, W.C.; Fuchs, C.S. Association of nut consumption with total and cause-specific mortality. N. Engl. J. Med. 2013, 369, 2001–2011. [Google Scholar] [CrossRef]
  196. Jeurnink, S.M.; Ros, M.M.; Leenders, M.; van Duijnhoven, F.J.B.; Siersema, P.D.; Jansen, E.H.J.M.; van Gils, C.H.; Bakker, M.F.; Overvad, K.; Roswall, N.; et al. Plasma carotenoids, vitamin C, retinol and tocopherols levels and pancreatic cancer risk within the European Prospective Investigation into Cancer and Nutrition: A nested case-control study: Plasma micronutrients and pancreatic cancer risk. Int. J. Cancer 2015, 136, E665–E676. [Google Scholar] [CrossRef]
  197. Sahu, R.P.; Zhang, R.; Batra, S.; Shi, Y.; Srivastava, S.K. Benzyl isothiocyanate-mediated generation of reactive oxygen species causes cell cycle arrest and induces apoptosis via activation of MAPK in human pancreatic cancer cells. Carcinogenesis 2009, 30, 1744–1753. [Google Scholar] [CrossRef] [PubMed]
  198. Basu, A.; Haldar, S. Anti-proliferative and proapoptotic effects of benzyl isothiocyanate on human pancreatic cancer cells is linked to death receptor activation and RasGAP/Rac1 down-modulation. Int. J. Oncol. 2009, 35, 593–599. [Google Scholar] [CrossRef] [PubMed]
  199. Giovannucci, E.; Liu, Y.; Rimm, E.B.; Hollis, B.W.; Fuchs, C.S.; Stampfer, M.J.; Willett, W.C. Prospective study of predictors of vitamin D status and cancer incidence and mortality in men. J. Natl. Cancer Inst. 2006, 98, 451–459. [Google Scholar] [CrossRef] [PubMed]
  200. Skinner, H.G.; Michaud, D.S.; Giovannucci, E.; Willett, W.C.; Colditz, G.A.; Fuchs, C.S. Vitamin D intake and the risk for pancreatic cancer in two cohort studies. Cancer Epidemiol. Biomark. Prev. 2006, 15, 1688–1695. [Google Scholar] [CrossRef] [PubMed]
  201. Stolzenberg-Solomon, R.Z.; Vieth, R.; Azad, A.; Pietinen, P.; Taylor, P.R.; Virtamo, J.; Albanes, D. A Prospective Nested Case-Control Study of Vitamin D Status and Pancreatic Cancer Risk in Male Smokers. Cancer Res. 2006, 66, 10213–10219. [Google Scholar] [CrossRef] [PubMed]
  202. Whiting, S.J.; Calvo, M.S. Dietary recommendations to meet both endocrine and autocrine needs of Vitamin D. J. Steroid Biochem. Mol. Biol. 2005, 97, 7–12. [Google Scholar] [CrossRef]
  203. Schwartz, G.G.; Eads, D.; Rao, A.; Cramer, S.D.; Willingham, M.C.; Chen, T.C.; Jamieson, D.P.; Wang, L.; Burnstein, K.L.; Holick, M.F.; et al. Pancreatic cancer cells express 25-hydroxyvitamin D-1 alpha-hydroxylase and their proliferation is inhibited by the prohormone 25-hydroxyvitamin D3. Carcinogenesis 2004, 25, 1015–1026. [Google Scholar] [CrossRef]
  204. Pettersson, F.; Colston, K.W.; Dalgleish, A.G. Differential and antagonistic effects of 9-cis-retinoic acid and vitamin D analogues on pancreatic cancer cells in vitro. Br. J. Cancer 2000, 83, 239–245. [Google Scholar] [CrossRef] [PubMed]
  205. Kawa, S.; Yoshizawa, K.; Tokoo, M.; Imai, H.; Oguchi, H.; Kiyosawa, K.; Homma, T.; Nikaido, T.; Furihata, K. Inhibitory effect of 220-oxa-1,25-dihydroxyvitamin D3 on the proliferation of pancreatic cancer cell lines. Gastroenterology 1996, 110, 1605–1613. [Google Scholar] [CrossRef]
  206. Maestro, B.; Dávila, N.; Carranza, M.C.; Calle, C. Identification of a Vitamin D response element in the human insulin receptor gene promoter. J. Steroid Biochem. Mol. Biol. 2003, 84, 223–230. [Google Scholar] [CrossRef]
  207. Cade, C.; Norman, A.W. Vitamin D3 improves impaired glucose tolerance and insulin secretion in the vitamin D-deficient rat in vivo. Endocrinology 1986, 119, 84–90. [Google Scholar] [CrossRef]
  208. Norman, A.W.; Frankel, J.B.; Heldt, A.M.; Grodsky, G.M. Vitamin D deficiency inhibits pancreatic secretion of insulin. Science 1980, 209, 823–825. [Google Scholar] [CrossRef]
  209. Roberts, S.E.; Morrison-Rees, S.; John, A.; Williams, J.G.; Brown, T.H.; Samuel, D.G. The incidence and aetiology of acute pancreatitis across Europe. Pancreatology 2017, 17, 155–165. [Google Scholar] [CrossRef]
  210. Gukovskaya, A.S.; Gukovsky, I.; Algül, H.; Habtezion, A. Autophagy, Inflammation, and Immune Dysfunction in the Pathogenesis of Pancreatitis. Gastroenterology 2017, 153, 1212–1226. [Google Scholar] [CrossRef]
  211. Liu, J.; Wang, Y.; Yu, Y. Meta-analysis reveals an association between acute pancreatitis and the risk of pancreatic cancer. World J. Clin. Cases 2020, 8, 4416–4430. [Google Scholar] [CrossRef]
  212. Duell, E.J.; Lucenteforte, E.; Olson, S.H.; Bracci, P.M.; Li, D.; Risch, H.A.; Silverman, D.T.; Ji, B.T.; Gallinger, S.; Holly, E.A.; et al. Pancreatitis and pancreatic cancer risk: A pooled analysis in the International Pancreatic Cancer Case-Control Consortium (PanC4). Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2012, 23, 2964–2970. [Google Scholar] [CrossRef]
  213. Ahmed Ali, U.; Issa, Y.; Hagenaars, J.C.; Bakker, O.J.; van Goor, H.; Nieuwenhuijs, V.B.; Bollen, T.L.; van Ramshorst, B.; Witteman, B.J.; Brink, M.A.; et al. Risk of Recurrent Pancreatitis and Progression to Chronic Pancreatitis after a First Episode of Acute Pancreatitis. Clin. Gastroenterol. Hepatol. 2016, 14, 738–746. [Google Scholar] [CrossRef]
  214. Whitcomb, D.C. Value of genetic testing in the management of pancreatitis. Gut 2004, 53, 1710–1717. [Google Scholar] [CrossRef]
  215. Beyer, G.; Habtezion, A.; Werner, J.; Lerch, M.M.; Mayerle, J. Chronic pancreatitis. Lancet 2020, 396, 499–512. [Google Scholar] [CrossRef] [PubMed]
  216. Raimondi, S.; Lowenfels, A.B.; Morselli-Labate, A.M.; Maisonneuve, P.; Pezzilli, R. Pancreatic cancer in chronic pancreatitis; aetiology, incidence, and early detection. Best. Pract. Res. Clin. Gastroenterol. 2010, 24, 349–358. [Google Scholar] [CrossRef] [PubMed]
  217. Jeon, C.Y.; Chen, Q.; Yu, W.; Dong, E.Y.; Chung, J.; Pandol, S.J.; Yadav, D.; Conwell, D.L.; Wu, B.U. Identification of Individuals at Increased Risk for Pancreatic Cancer in a Community-Based Cohort of Patients with Suspected Chronic Pancreatitis. Clin. Transl. Gastroenterol. 2020, 11, e00147. [Google Scholar] [CrossRef]
  218. Kong, X.; Sun, T.; Kong, F.; Du, Y.; Li, Z. Chronic Pancreatitis and Pancreatic Cancer. Gastrointest. Tumors 2014, 1, 123–134. [Google Scholar] [CrossRef]
  219. Gandhi, S.; de la Fuente, J.; Murad, M.H.; Majumder, S. Chronic Pancreatitis Is a Risk Factor for Pancreatic Cancer, and Incidence Increases with Duration of Disease: A Systematic Review and Meta-analysis. Clin. Transl. Gastroenterol. 2022, 13, e00463. [Google Scholar] [CrossRef]
  220. Munigala, S.; Kanwal, F.; Xian, H.; Agarwal, B. New diagnosis of chronic pancreatitis: Risk of missing an underlying pancreatic cancer. Am. J. Gastroenterol. 2014, 109, 1824–1830. [Google Scholar] [CrossRef]
  221. Ma, D.-M.; Dong, X.-W.; Han, X.; Ling, Z.; Lu, G.-T.; Sun, Y.-Y.; Yin, X.-D. Pancreatitis and Pancreatic Cancer Risk. Technol. Cancer Res. Treat. 2023, 22, 15330338231164875. [Google Scholar] [CrossRef]
  222. Kirkegård, J.; Mortensen, F.V.; Cronin-Fenton, D. Chronic Pancreatitis and Pancreatic Cancer Risk: A Systematic Review and Meta-analysis. Am. J. Gastroenterol. 2017, 112, 1366–1372. [Google Scholar] [CrossRef]
  223. Blaho, M.; Dítě, P.; Kunovský, L.; Kianička, B. Autoimmune pancreatitis—An ongoing challenge. Adv. Med. Sci. 2020, 65, 403–408. [Google Scholar] [CrossRef]
  224. Gupta, R.; Khosroshahi, A.; Shinagare, S.; Fernandez, C.; Ferrone, C.; Lauwers, G.Y.; Stone, J.H.; Deshpande, V. Does autoimmune pancreatitis increase the risk of pancreatic carcinoma?: A retrospective analysis of pancreatic resections. Pancreas 2013, 42, 506–510. [Google Scholar] [CrossRef]
  225. Ikeura, T.; Miyoshi, H.; Uchida, K.; Fukui, T.; Shimatani, M.; Fukui, Y.; Sumimoto, K.; Matsushita, M.; Takaoka, M.; Okazaki, K. Relationship between autoimmune pancreatitis and pancreatic cancer: A single-center experience. Pancreatology 2014, 14, 373–379. [Google Scholar] [CrossRef] [PubMed]
  226. Hart, P.A.; Kamisawa, T.; Brugge, W.R.; Chung, J.B.; Culver, E.L.; Czakó, L.; Frulloni, L.; Go, V.L.W.; Gress, T.M.; Kim, M.-H.; et al. Long-term outcomes of autoimmune pancreatitis: A multicentre, international analysis. Gut 2013, 62, 1771–1776. [Google Scholar] [CrossRef] [PubMed]
  227. Yamamoto, M.; Takahashi, H.; Tabeya, T.; Suzuki, C.; Naishiro, Y.; Ishigami, K.; Yajima, H.; Shimizu, Y.; Obara, M.; Yamamoto, H.; et al. Risk of malignancies in IgG4-related disease. Mod. Rheumatol. 2012, 22, 414–418. [Google Scholar] [CrossRef]
  228. Petrov, M.S.; Yadav, D. Global epidemiology and holistic prevention of pancreatitis. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 175–184. [Google Scholar] [CrossRef]
  229. Woodmansey, C.; McGovern, A.P.; McCullough, K.A.; Whyte, M.B.; Munro, N.M.; Correa, A.C.; Gatenby, P.A.C.; Jones, S.A.; de Lusignan, S. Incidence, Demographics, and Clinical Characteristics of Diabetes of the Exocrine Pancreas (Type 3c): A Retrospective Cohort Study. Diabetes Care 2017, 40, 1486–1493. [Google Scholar] [CrossRef]
  230. Cho, J.; Scragg, R.; Petrov, M.S. Postpancreatitis Diabetes Confers Higher Risk for Pancreatic Cancer Than Type 2 Diabetes: Results from a Nationwide Cancer Registry. Diabetes Care 2020, 43, 2106–2112. [Google Scholar] [CrossRef]
  231. Tramacere, I.; Scotti, L.; Jenab, M.; Bagnardi, V.; Bellocco, R.; Rota, M.; Corrao, G.; Bravi, F.; Boffetta, P.; La Vecchia, C. Alcohol drinking and pancreatic cancer risk: A meta-analysis of the dose-risk relation. Int. J. Cancer 2010, 126, 1474–1486. [Google Scholar] [CrossRef]
  232. Wang, Y.-T.; Gou, Y.-W.; Jin, W.-W.; Xiao, M.; Fang, H.-Y. Association between alcohol intake and the risk of pancreatic cancer: A dose–response meta-analysis of cohort studies. BMC Cancer 2016, 16, 212. [Google Scholar] [CrossRef]
  233. Gapstur, S.M.; Jacobs, E.J.; Deka, A.; McCullough, M.L.; Patel, A.V.; Thun, M.J. Association of alcohol intake with pancreatic cancer mortality in never smokers. Arch. Intern. Med. 2011, 171, 444–451. [Google Scholar] [CrossRef]
  234. Naudin, S.; Li, K.; Jaouen, T.; Assi, N.; Kyrø, C.; Tjønneland, A.; Overvad, K.; Boutron-Ruault, M.C.; Rebours, V.; Védié, A.L.; et al. Lifetime and baseline alcohol intakes and risk of pancreatic cancer in the European Prospective Investigation into Cancer and Nutrition study. Int. J. Cancer 2018, 143, 801–812. [Google Scholar] [CrossRef]
  235. Shan, Y.-S.; Chen, L.-T.; Wu, C.-H.; Chang, Y.-F.; Lee, C.-T.; Chiang, N.-J.; Chao, Y.-J.; Yen, C.-J.; Tsai, H.-J.; Huang, H.-E.; et al. No association between alcohol consumption and pancreatic cancer even among individuals genetically susceptible to the carcinogenicity of alcohol. Sci. Rep. 2021, 11, 14567. [Google Scholar] [CrossRef] [PubMed]
  236. Jayasekara, H.; English, D.R.; Hodge, A.M.; Room, R.; Hopper, J.L.; Milne, R.L.; Giles, G.G.; MacInnis, R.J. Lifetime alcohol intake and pancreatic cancer incidence and survival: Findings from the Melbourne Collaborative Cohort Study. Cancer Causes Control 2019, 30, 323–331. [Google Scholar] [CrossRef] [PubMed]
  237. Yu, W.; Ma, Y.; Shankar, S.; Srivastava, R.K. Chronic ethanol exposure of human pancreatic normal ductal epithelial cells induces cancer stem cell phenotype through SATB2. J. Cell. Mol. Med. 2018, 22, 3920–3928. [Google Scholar] [CrossRef] [PubMed]
  238. Serio, R.N.; Gudas, L.J. Modification of stem cell states by alcohol and acetaldehyde. Chem. Biol. Interact. 2020, 316, 108919. [Google Scholar] [CrossRef]
  239. Li, T.-D.; Yang, H.-W.; Wang, P.; Song, C.-H.; Wang, K.-J.; Dai, L.-P.; Shi, J.-X.; Zhang, J.-Y.; Ye, H. Coffee consumption and risk of pancreatic cancer: A systematic review and dose–response meta-analysis. Int. J. Food Sci. Nutr. 2019, 70, 519–529. [Google Scholar] [CrossRef]
  240. Nie, K.; Xing, Z.; Huang, W.; Wang, W.; Liu, W. Coffee intake and risk of pancreatic cancer: An updated meta-analysis of prospective studies. Minerva Med. 2016, 107, 270–278. [Google Scholar]
  241. Bae, J.-M.; Shim, S.R. Coffee Consumption and Pancreatic Cancer Risk: A Meta-Epidemiological Study of Population-based Cohort Studies. Asian Pac. J. Cancer Prev. 2020, 21, 2793–2798. [Google Scholar] [CrossRef]
  242. Turati, F.; Galeone, C.; Edefonti, V.; Ferraroni, M.; Lagiou, P.; La Vecchia, C.; Tavani, A. A meta-analysis of coffee consumption and pancreatic cancer. Ann. Oncol. 2012, 23, 311–318. [Google Scholar] [CrossRef]
  243. Zhou, C.D.; Kuan, A.S.; Reeves, G.K.; Green, J.; Floud, S.; Beral, V.; Yang, T.O.; Million Women Study Collaborators. Coffee and pancreatic cancer risk among never-smokers in the UK prospective Million Women Study. Int. J. Cancer 2019, 145, 1484–1492. [Google Scholar] [CrossRef]
  244. Ran, H.-Q.; Wang, J.-Z.; Sun, C.-Q. Coffee Consumption and Pancreatic Cancer Risk: An Update Meta-analysis of Cohort Studies. Pak. J. Med. Sci. 2016, 32, 253–259. [Google Scholar] [CrossRef]
  245. Lukic, M.; Nilsson, L.M.; Skeie, G.; Lindahl, B.; Braaten, T. Coffee consumption and risk of rare cancers in Scandinavian countries. Eur. J. Epidemiol. 2018, 33, 287–302. [Google Scholar] [CrossRef]
  246. Nuhu, A.A. Bioactive Micronutrients in Coffee: Recent Analytical Approaches for Characterization and Quantification. ISRN Nutr. 2014, 2014, 384230. [Google Scholar] [CrossRef]
  247. Liang, N.; Kitts, D.D. Antioxidant property of coffee components: Assessment of methods that define mechanisms of action. Molecules 2014, 19, 19180–19208. [Google Scholar] [CrossRef]
  248. World Health Organization. Global Hepatitis Report 2017; World Health Organization: Geneva, Switzerland, 2017; ISBN 978-92-4-156545-5. [Google Scholar]
  249. de Martel, C.; Georges, D.; Bray, F.; Ferlay, J.; Clifford, G.M. Global burden of cancer attributable to infections in 2018: A worldwide incidence analysis. Lancet Glob. Health 2020, 8, e180–e190. [Google Scholar] [CrossRef]
  250. Poynard, T.; Ratziu, V.; Charlotte, F.; Goodman, Z.; McHutchison, J.; Albrecht, J. Rates and risk factors of liver fibrosis progression in patients with chronic hepatitis c. J. Hepatol. 2001, 34, 730–739. [Google Scholar] [CrossRef]
  251. Dandri, M.; Locarnini, S. New insight in the pathobiology of hepatitis B virus infection. Gut 2012, 61 (Suppl. 1), i6–i17. [Google Scholar] [CrossRef]
  252. Yoshimura, M.; Sakurai, I.; Shimoda, T.; Abe, K.; Okano, T.; Shikata, T. Detection of HBsAg in the pancreas. Acta Pathol. Jpn. 1981, 31, 711–717. [Google Scholar] [CrossRef]
  253. Montalbano, M.; Neff, G.W. Management of recurrent viral hepatitis B and C after liver transplantation. Curr. Gastroenterol. Rep. 2006, 8, 60–66. [Google Scholar] [CrossRef]
  254. Huang, J.; Magnusson, M.; Törner, A.; Ye, W.; Duberg, A.-S. Risk of pancreatic cancer among individuals with hepatitis C or hepatitis B virus infection: A nationwide study in Sweden. Br. J. Cancer 2013, 109, 2917–2923. [Google Scholar] [CrossRef]
  255. Hassan, M.M.; Li, D.; El-Deeb, A.S.; Wolff, R.A.; Bondy, M.L.; Davila, M.; Abbruzzese, J.L. Association between Hepatitis B Virus and Pancreatic Cancer. J. Clin. Oncol. 2008, 26, 4557–4562. [Google Scholar] [CrossRef]
  256. Xu, J.-H.; Fu, J.-J.; Wang, X.-L.; Zhu, J.-Y.; Ye, X.-H.; Chen, S.-D. Hepatitis B or C viral infection and risk of pancreatic cancer: A meta-analysis of observational studies. World J. Gastroenterol. 2013, 19, 4234–4241. [Google Scholar] [CrossRef]
  257. Parsa, I.; Longnecker, D.S.; Scarpelli, D.G.; Pour, P.; Reddy, J.K.; Lefkowitz, M. Ductal metaplasia of human exocrine pancreas and its association with carcinoma. Cancer Res. 1985, 45, 1285–1290. [Google Scholar]
  258. Peery, A.F.; Crockett, S.D.; Barritt, A.S.; Dellon, E.S.; Eluri, S.; Gangarosa, L.M.; Jensen, E.T.; Lund, J.L.; Pasricha, S.; Runge, T.; et al. Burden of Gastrointestinal, Liver, and Pancreatic Diseases in the United States. Gastroenterology 2015, 149, 1731–1741.e3. [Google Scholar] [CrossRef]
  259. European Association for the Study of the Liver (EASL). EASL Clinical Practice Guidelines on the prevention, diagnosis and treatment of gallstones. J. Hepatol. 2016, 65, 146–181. [Google Scholar] [CrossRef]
  260. Fan, Y.; Hu, J.; Feng, B.; Wang, W.; Yao, G.; Zhai, J.; Li, X. Increased Risk of Pancreatic Cancer Related to Gallstones and Cholecystectomy: A Systematic Review and Meta-Analysis. Pancreas 2016, 45, 503–509. [Google Scholar] [CrossRef]
  261. Luo, X.; Yang, W.; Joshi, A.D.; Wu, K.; Simon, T.G.; Yuan, C.; Jin, L.; Long, L.; Kim, M.N.; Lo, C.-H.; et al. Gallstones and risk of cancers of the liver, biliary tract and pancreas: A prospective study within two U.S. cohorts. Br. J. Cancer 2022, 127, 1069–1075. [Google Scholar] [CrossRef]
  262. Rosato, V.; Gómez-Rubio, P.; Molina-Montes, E.; Márquez, M.; Löhr, M.; O’Rorke, M.; Michalski, C.W.; Molero, X.; Farré, A.; Perea, J.; et al. Gallbladder disease and pancreatic cancer risk: A multicentric case-control European study. Eur. J. Cancer Prev. 2021, 30, 423–430. [Google Scholar] [CrossRef]
  263. Rosato, V.; Negri, E.; Bosetti, C.; Malats, N.; Gomez-Rubio, P.; Consortium, P.; Maisonneuve, P.; Miller, A.B.; Bueno-de-Mesquita, H.B.; Baghurst, P.A.; et al. Gallbladder disease, cholecystectomy, and pancreatic cancer risk in the International Pancreatic Cancer Case-Control Consortium (PanC4). Eur. J. Cancer Prev. 2020, 29, 408–415. [Google Scholar] [CrossRef]
  264. Shabanzadeh, D.M.; Martinussen, T.; Sørensen, L.T. Development of upper gastrointestinal cancer in patients with symptomatic gallstones, cholecystectomy, and sphincterotomy: A nationwide cohort study. Scand. J. Surg. 2022, 111, 39–47. [Google Scholar] [CrossRef]
  265. Shabanzadeh, D.M.; Sørensen, L.T.; Jørgensen, T. Association between Screen-Detected Gallstone Disease and Cancer in a Cohort Study. Gastroenterology 2017, 152, 1965–1974.e1. [Google Scholar] [CrossRef]
  266. Wang, C.-C.; Tseng, M.-H.; Wu, S.-W.; Yang, T.-W.; Chen, H.-Y.; Sung, W.-W.; Su, C.-C.; Wang, Y.-T.; Chen, W.-L.; Lai, H.-C.; et al. Symptomatic cholelithiasis patients have an increased risk of pancreatic cancer: A population-based study. J. Gastroenterol. Hepatol. 2021, 36, 1187–1196. [Google Scholar] [CrossRef]
  267. Yu, J.; Ploner, A.; Chen, M.S.; Zhang, J.; Sandborgh-Englund, G.; Ye, W. Poor dental health and risk of pancreatic cancer: A nationwide registry-based cohort study in Sweden, 2009–2016. Br. J. Cancer 2022, 127, 2133–2140. [Google Scholar] [CrossRef]
  268. Chang, J.S.; Tsai, C.-R.; Chen, L.-T.; Shan, Y.-S. Investigating the Association between Periodontal Disease and Risk of Pancreatic Cancer. Pancreas 2016, 45, 134–141. [Google Scholar] [CrossRef]
  269. Michaud, D.S.; Joshipura, K.; Giovannucci, E.; Fuchs, C.S. A Prospective Study of Periodontal Disease and Pancreatic Cancer in US Male Health Professionals. JNCI J. Natl. Cancer Inst. 2007, 99, 171–175. [Google Scholar] [CrossRef]
  270. Ahn, J.; Segers, S.; Hayes, R.B. Periodontal disease, Porphyromonas gingivalis serum antibody levels and orodigestive cancer mortality. Carcinogenesis 2012, 33, 1055–1058. [Google Scholar] [CrossRef]
  271. Fan, X.; Alekseyenko, A.V.; Wu, J.; Peters, B.A.; Jacobs, E.J.; Gapstur, S.M.; Purdue, M.P.; Abnet, C.C.; Stolzenberg-Solomon, R.; Miller, G.; et al. Human oral microbiome and prospective risk for pancreatic cancer: A population-based nested case-control study. Gut 2018, 67, 120–127. [Google Scholar] [CrossRef]
  272. Michaud, D.S.; Izard, J.; Wilhelm-Benartzi, C.S.; You, D.-H.; Grote, V.A.; Tjønneland, A.; Dahm, C.C.; Overvad, K.; Jenab, M.; Fedirko, V.; et al. Plasma antibodies to oral bacteria and risk of pancreatic cancer in a large European prospective cohort study. Gut 2013, 62, 1764–1770. [Google Scholar] [CrossRef]
  273. del Castillo, E.; Meier, R.; Chung, M.; Koestler, D.C.; Chen, T.; Paster, B.J.; Charpentier, K.P.; Kelsey, K.T.; Izard, J.; Michaud, D.S. The Microbiomes of Pancreatic and Duodenum Tissue Overlap and are Highly Subject Specific but Differ between Pancreatic Cancer and Non-Cancer Subjects. Cancer Epidemiol. Biomark. Prev. 2019, 28, 370–383. [Google Scholar] [CrossRef]
  274. Maisonneuve, P.; Amar, S.; Lowenfels, A.B. Periodontal disease, edentulism, and pancreatic cancer: A meta-analysis. Ann. Oncol. 2017, 28, 985–995. [Google Scholar] [CrossRef]
  275. Farrell, J.J.; Zhang, L.; Zhou, H.; Chia, D.; Elashoff, D.; Akin, D.; Paster, B.J.; Joshipura, K.; Wong, D.T.W. Variations of oral microbiota are associated with pancreatic diseases including pancreatic cancer. Gut 2012, 61, 582–588. [Google Scholar] [CrossRef]
  276. Hooi, J.K.Y.; Lai, W.Y.; Ng, W.K.; Suen, M.M.Y.; Underwood, F.E.; Tanyingoh, D.; Malfertheiner, P.; Graham, D.Y.; Wong, V.W.S.; Wu, J.C.Y.; et al. Global Prevalence of Helicobacter pylori Infection: Systematic Review and Meta-Analysis. Gastroenterology 2017, 153, 420–429. [Google Scholar] [CrossRef]
  277. Huang, J.; Zagai, U.; Hallmans, G.; Nyrén, O.; Engstrand, L.; Stolzenberg-Solomon, R.; Duell, E.J.; Overvad, K.; Katzke, V.A.; Kaaks, R.; et al. Helicobacter pylori infection, chronic corpus atrophic gastritis and pancreatic cancer risk in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort: A nested case-control study. Int. J. Cancer 2017, 140, 1727–1735. [Google Scholar] [CrossRef] [PubMed]
  278. Bulajic, M.; Panic, N.; Löhr, J.M. Helicobacter pylori and pancreatic diseases. World J. Gastrointest. Pathophysiol. 2014, 5, 380–383. [Google Scholar] [CrossRef] [PubMed]
  279. Cullin, N.; Antunes, C.A.; Straussman, R.; Stein-Thoeringer, C.K.; Elinav, E. Microbiome and cancer. Cancer Cell 2021, 39, 1317–1341. [Google Scholar] [CrossRef] [PubMed]
  280. Gomez-Rubio, P.; Piñero, J.; Molina-Montes, E.; Gutiérrez-Sacristán, A.; Marquez, M.; Rava, M.; Michalski, C.W.; Farré, A.; Molero, X.; Löhr, M.; et al. Pancreatic cancer and autoimmune diseases: An association sustained by computational and epidemiological case–control approaches. Int. J. Cancer 2019, 144, 1540–1549. [Google Scholar] [CrossRef] [PubMed]
  281. Westermann, R.; Zobbe, K.; Cordtz, R.; Haugaard, J.H.; Dreyer, L. Increased cancer risk in patients with cutaneous lupus erythematosus and systemic lupus erythematosus compared with the general population: A Danish nationwide cohort study. Lupus 2021, 30, 752–761. [Google Scholar] [CrossRef]
  282. Zhang, M.; Wang, Y.; Wang, Y.; Bai, Y.; Gu, D. Association between Systemic Lupus Erythematosus and Cancer Morbidity and Mortality: Findings from Cohort Studies. Front. Oncol. 2022, 12, 860794. [Google Scholar] [CrossRef]
  283. Osman, M.A.; Alkhouly, M.; Elmohaseb, G.F.; Nassef, E.M.; Mohamed, I.G.R.; Mancy, I.M.E.; Sabry, S.; Abdulrehim, M.M.; Eliwa, A.; Eisa, Y.H.; et al. Relation between Non-Alcoholic Fatty Pancreas and Clinical and Biochemical Parameters in Women with Polycystic Ovary Syndrome: A Multi-Centric Study. Int. J. Gen. Med. 2022, 15, 8225–8233. [Google Scholar] [CrossRef] [PubMed]
  284. Yin, W.; Falconer, H.; Yin, L.; Xu, L.; Ye, W. Association between Polycystic Ovary Syndrome and Cancer Risk. JAMA Oncol. 2019, 5, 106–107. [Google Scholar] [CrossRef] [PubMed]
  285. Peeri, N.C.; Landicino, M.V.; Saldia, C.A.; Kurtz, R.C.; Rolston, V.S.; Du, M. Association between Polycystic Ovary Syndrome and Risk of Pancreatic Cancer. JAMA Oncol. 2022, 8, 1845–1847. [Google Scholar] [CrossRef] [PubMed]
  286. Butt, S.A.; Lidegaardi, Ø.; Skovlund, C.; Hannaford, P.C.; Iversen, L.; Fielding, S.; Mørch, L.S. Hormonal contraceptive use and risk of pancreatic cancer—A cohort study among premenopausal women. PLoS ONE 2018, 13, e0206358. [Google Scholar] [CrossRef] [PubMed]
  287. Tang, B.; Lv, J.; Li, Y.; Yuan, S.; Wang, Z.; He, S. Relationship between female hormonal and menstrual factors and pancreatic cancer: A meta-analysis of observational studies. Medicine 2015, 94, e177. [Google Scholar] [CrossRef] [PubMed]
  288. Duell, E.J.; Travier, N.; Lujan-Barroso, L.; Dossus, L.; Boutron-Ruault, M.-C.; Clavel-Chapelon, F.; Tumino, R.; Masala, G.; Krogh, V.; Panico, S.; et al. Menstrual and reproductive factors in women, genetic variation in CYP17A1, and pancreatic cancer risk in the European prospective investigation into cancer and nutrition (EPIC) cohort. Int. J. Cancer 2013, 132, 2164–2175. [Google Scholar] [CrossRef]
  289. Pushalkar, S.; Hundeyin, M.; Daley, D.; Zambirinis, C.P.; Kurz, E.; Mishra, A.; Mohan, N.; Aykut, B.; Usyk, M.; Torres, L.E.; et al. The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression. Cancer Discov. 2018, 8, 403–416. [Google Scholar] [CrossRef] [PubMed]
  290. Thomas, R.M.; Gharaibeh, R.Z.; Gauthier, J.; Beveridge, M.; Pope, J.L.; Guijarro, M.V.; Yu, Q.; He, Z.; Ohland, C.; Newsome, R.; et al. Intestinal microbiota enhances pancreatic carcinogenesis in preclinical models. Carcinogenesis 2018, 39, 1068–1078. [Google Scholar] [CrossRef] [PubMed]
  291. Li, S.; Fuhler, G.M.; BN, N.; Jose, T.; Bruno, M.J.; Peppelenbosch, M.P.; Konstantinov, S.R. Pancreatic cyst fluid harbors a unique microbiome. Microbiome 2017, 5, 147. [Google Scholar] [CrossRef] [PubMed]
  292. Kostic, A.D.; Chun, E.; Robertson, L.; Glickman, J.N.; Gallini, C.A.; Michaud, M.; Clancy, T.E.; Chung, D.C.; Lochhead, P.; Hold, G.L.; et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor immune microenvironment. Cell Host Microbe 2013, 14, 207–215. [Google Scholar] [CrossRef] [PubMed]
  293. Li, Q.; Jin, M.; Liu, Y.; Jin, L. Gut Microbiota: Its Potential Roles in Pancreatic Cancer. Front. Cell. Infect. Microbiol. 2020, 10, 572492. [Google Scholar] [CrossRef]
  294. Gur, C.; Ibrahim, Y.; Isaacson, B.; Yamin, R.; Abed, J.; Gamliel, M.; Enk, J.; Bar-On, Y.; Stanietsky-Kaynan, N.; Coppenhagen-Glazer, S.; et al. Binding of the Fap2 Protein of Fusobacterium nucleatum to Human Inhibitory Receptor TIGIT Protects Tumors from Immune Cell Attack. Immunity 2015, 42, 344–355. [Google Scholar] [CrossRef] [PubMed]
  295. Rubinstein, M.R.; Wang, X.; Liu, W.; Hao, Y.; Cai, G.; Han, Y.W. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe 2013, 14, 195–206. [Google Scholar] [CrossRef]
  296. Sharif, R.; Dawra, R.; Wasiluk, K.; Phillips, P.; Dudeja, V.; Kurt-Jones, E.; Finberg, R.; Saluja, A. Impact of toll-like receptor 4 on the severity of acute pancreatitis and pancreatitis-associated lung injury in mice. Gut 2009, 58, 813–819. [Google Scholar] [CrossRef]
  297. Yin, H.; Pu, N.; Chen, Q.; Zhang, J.; Zhao, G.; Xu, X.; Wang, D.; Kuang, T.; Jin, D.; Lou, W.; et al. Gut-derived lipopolysaccharide remodels tumoral microenvironment and synergizes with PD-L1 checkpoint blockade via TLR4/MyD88/AKT/NF-κB pathway in pancreatic cancer. Cell Death Dis. 2021, 12, 1033. [Google Scholar] [CrossRef]
  298. Liu, J.; Xu, D.; Wang, Q.; Zheng, D.; Jiang, X.; Xu, L. LPS induced miR-181a promotes pancreatic cancer cell migration via targeting PTEN and MAP2K4. Dig. Dis. Sci. 2014, 59, 1452–1460. [Google Scholar] [CrossRef] [PubMed]
  299. Santoni, M.; Andrikou, K.; Sotte, V.; Bittoni, A.; Lanese, A.; Pellei, C.; Piva, F.; Conti, A.; Nabissi, M.; Santoni, G.; et al. Toll like receptors and pancreatic diseases: From a pathogenetic mechanism to a therapeutic target. Cancer Treat. Rev. 2015, 41, 569–576. [Google Scholar] [CrossRef] [PubMed]
  300. Tilg, H.; Zmora, N.; Adolph, T.E.; Elinav, E. The intestinal microbiota fuelling metabolic inflammation. Nat. Rev. Immunol. 2020, 20, 40–54. [Google Scholar] [CrossRef] [PubMed]
  301. Geller, L.T.; Barzily-Rokni, M.; Danino, T.; Jonas, O.H.; Shental, N.; Nejman, D.; Gavert, N.; Zwang, Y.; Cooper, Z.A.; Shee, K.; et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science 2017, 357, 1156–1160. [Google Scholar] [CrossRef] [PubMed]
  302. Riquelme, E.; Zhang, Y.; Zhang, L.; Montiel, M.; Zoltan, M.; Dong, W.; Quesada, P.; Sahin, I.; Chandra, V.; Lucas, A.S.; et al. Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes. Cell 2019, 178, 795–806.e12. [Google Scholar] [CrossRef] [PubMed]
  303. Zhou, W.; Zhang, D.; Li, Z.; Jiang, H.; Li, J.; Ren, R.; Gao, X.; Li, J.; Wang, X.; Wang, W.; et al. The fecal microbiota of patients with pancreatic ductal adenocarcinoma and autoimmune pancreatitis characterized by metagenomic sequencing. J. Transl. Med. 2021, 19, 215. [Google Scholar] [CrossRef] [PubMed]
  304. Sammallahti, H.; Sarhadi, V.K.; Kokkola, A.; Ghanbari, R.; Rezasoltani, S.; Asadzadeh Aghdaei, H.; Puolakkainen, P.; Knuutila, S. Oncogenomic Changes in Pancreatic Cancer and Their Detection in Stool. Biomolecules 2022, 12, 652. [Google Scholar] [CrossRef] [PubMed]
  305. Chen, S.-M.; Chieng, W.-W.; Huang, S.-W.; Hsu, L.-J.; Jan, M.-S. The synergistic tumor growth-inhibitory effect of probiotic Lactobacillus on transgenic mouse model of pancreatic cancer treated with gemcitabine. Sci. Rep. 2020, 10, 20319. [Google Scholar] [CrossRef] [PubMed]
  306. Weniger, M.; Hank, T.; Qadan, M.; Ciprani, D.; Michelakos, T.; Niess, H.; Heiliger, C.; Ilmer, M.; D’Haese, J.G.; Ferrone, C.R.; et al. Influence of Klebsiella pneumoniae and quinolone treatment on prognosis in patients with pancreatic cancer. Br. J. Surg. 2021, 108, 709–716. [Google Scholar] [CrossRef]
  307. Ross, K. Mapping Pathways from Stress to Cancer Progression. JNCI J. Natl. Cancer Inst. 2008, 100, 914–917. [Google Scholar] [CrossRef]
  308. Kennedy, B.; Valdimarsdóttir, U.; Sundström, K.; Sparén, P.; Lambe, M.; Fall, K.; Fang, F. Loss of a parent and the risk of cancer in early life: A nationwide cohort study. Cancer Causes Control 2014, 25, 499–506. [Google Scholar] [CrossRef] [PubMed]
  309. Huang, J.; Valdimarsdóttir, U.; Fall, K.; Ye, W.; Fang, F. Pancreatic Cancer Risk after Loss of a Child: A Register-based Study in Sweden During 1991–2009. Am. J. Epidemiol. 2013, 178, 582–589. [Google Scholar] [CrossRef] [PubMed]
  310. Kim-Fuchs, C.; Le, C.P.; Pimentel, M.A.; Shackleford, D.; Ferrari, D.; Angst, E.; Hollande, F.; Sloan, E.K. Chronic stress accelerates pancreatic cancer growth and invasion: A critical role for beta-adrenergic signaling in the pancreatic microenvironment. Brain Behav. Immun. 2014, 40, 40–47. [Google Scholar] [CrossRef] [PubMed]
  311. Weddle, D.L.; Tithoff, P.; Williams, M.; Schuller, H.M. Beta-adrenergic growth regulation of human cancer cell lines derived from pancreatic ductal carcinomas. Carcinogenesis 2001, 22, 473–479. [Google Scholar] [CrossRef] [PubMed]
  312. Lin, X.; Luo, K.; Lv, Z.; Huang, J. Beta-adrenoceptor action on pancreatic cancer cell proliferation and tumor growth in mice. Hepatogastroenterology 2012, 59, 584–588. [Google Scholar] [CrossRef] [PubMed]
  313. Al-Wadei, H.A.; Al-Wadei, M.H.; Schuller, H.M. Prevention of pancreatic cancer by the beta-blocker propranolol. Anticancer Drugs 2009, 20, 477–482. [Google Scholar] [CrossRef]
  314. Beg, M.S.; Gupta, A.; Sher, D.; Ali, S.; Khan, S.; Gao, A.; Stewart, T.; Ahn, C.; Berry, J.; Mortensen, E.M. Impact of Concurrent Medication Use on Pancreatic Cancer Survival—SEER-Medicare Analysis. Am. J. Clin. Oncol. 2018, 41, 766. [Google Scholar] [CrossRef] [PubMed]
  315. Daemen, M.J.; Lombardi, D.M.; Bosman, F.T.; Schwartz, S.M. Angiotensin II induces smooth muscle cell proliferation in the normal and injured rat arterial wall. Circ. Res. 1991, 68, 450–456. [Google Scholar] [CrossRef] [PubMed]
  316. Kubota, M.; Shimizu, M.; Sakai, H.; Yasuda, Y.; Ohno, T.; Kochi, T.; Tsurumi, H.; Tanaka, T.; Moriwaki, H. Renin-angiotensin system inhibitors suppress azoxymethane-induced colonic preneoplastic lesions in C57BL/KsJ-db/db obese mice. Biochem. Biophys. Res. Commun. 2011, 410, 108–113. [Google Scholar] [CrossRef]
  317. Fendrich, V.; Chen, N.-M.; Neef, M.; Waldmann, J.; Buchholz, M.; Feldmann, G.; Slater, E.P.; Maitra, A.; Bartsch, D.K. The angiotensin-I-converting enzyme inhibitor enalapril and aspirin delay progression of pancreatic intraepithelial neoplasia and cancer formation in a genetically engineered mouse model of pancreatic cancer. Gut 2010, 59, 630–637. [Google Scholar] [CrossRef]
  318. Lee, S.-H.; Park, J.; Park, R.W.; Shin, S.J.; Kim, J.; Sung, J.D.; Kim, D.J.; Yang, K. Renin-Angiotensin-Aldosterone System Inhibitors and Risk of Cancer: A Population-Based Cohort Study Using a Common Data Model. Diagnostics 2022, 12, 263. [Google Scholar] [CrossRef] [PubMed]
  319. Tse, G.; Zhou, J.; Lee, S.; Hung, J.K.F.; Leung, K.S.K.; Liu, Y.; Zhang, Y.; Liu, T.; Wong, W.T.; Wong, I.C.K.; et al. Incidence of pancreatic cancer in angiotensin-converting enzyme inhibitors (ACEIs) versus angiotensin receptor blockers (ARBs): A population-based cohort study. medRxiv 2022. [Google Scholar] [CrossRef]
  320. Mandilaras, V.; Bouganim, N.; Yin, H.; Asselah, J.; Azoulay, L. The use of drugs acting on the renin–angiotensin system and the incidence of pancreatic cancer. Br. J. Cancer 2017, 116, 103–108. [Google Scholar] [CrossRef]
  321. Liu, H.; D’Alesio, M.; Lebowitz, S.; Hammad, A.; Nassour, I.; Singhi, A.; Bahary, N.; Lee, K.; Zureikat, A.; Paniccia, A. Angiotensin system inhibitor use is associated with longer survival in resected pancreatic cancer. HPB 2021, 23, S466–S467. [Google Scholar] [CrossRef]
  322. Tseng, K.-Y.; Chou, C.-W. Abstract 6755: The impact of renin-angiotensin system inhibitors on survival outcomes of pancreatic adenocarcinoma patients. Cancer Res. 2023, 83, 6755. [Google Scholar] [CrossRef]
  323. Gandini, S.; Lowenfels, A.B.; Jaffee, E.M.; Armstrong, T.D.; Maisonneuve, P. Allergies and the risk of pancreatic cancer: A meta-analysis with review of epidemiology and biological mechanisms. Cancer Epidemiol. Biomark. Prev. 2005, 14, 1908–1916. [Google Scholar] [CrossRef]
  324. Cotterchio, M.; Lowcock, E.; Hudson, T.J.; Greenwood, C.; Gallinger, S. Association between allergies and risk of pancreatic cancer. Cancer Epidemiol. Biomark. Prev. 2014, 23, 469–480. [Google Scholar] [CrossRef]
  325. Wang, G.; Xu, Z.; Zhu, J.; Ren, J.; Chen, M.; He, G.; Yu, B. Decreased Risk in the Pancreatic Cancer With History of Hay Fever: A Meta-Analysis. Front. Public Health 2020, 8, 551490. [Google Scholar] [CrossRef]
  326. Olson, S.H.; Hsu, M.; Satagopan, J.M.; Maisonneuve, P.; Silverman, D.T.; Lucenteforte, E.; Anderson, K.E.; Borgida, A.; Bracci, P.M.; Bueno-de-Mesquita, H.B.; et al. Allergies and Risk of Pancreatic Cancer: A Pooled Analysis from the Pancreatic Cancer Case-Control Consortium. Am. J. Epidemiol. 2013, 178, 691–700. [Google Scholar] [CrossRef]
  327. Olson, S.H. Selected medical conditions and risk of pancreatic cancer. Mol. Carcinog. 2012, 51, 75–97. [Google Scholar] [CrossRef]
  328. Santibañez, M.; O’Rorke, M.; O’Leary, E.; Cancela, M.D.C.; Murray, L.; Sharp, L.; on Behalf of the PanCAM Study Group. Allergies, asthma and the risk of pancreatic cancer: A population-based case-control study in Ireland. Eur. Respir. J. 2015, 46, PA3389. [Google Scholar] [CrossRef]
  329. Karim, A.F.; Westenberg, L.E.H.; Eurelings, L.E.M.; Otten, R.; Gerth van Wijk, R. The association between allergic diseases and cancer: A systematic review of the literature. Neth. J. Med. 2019, 77, 42–66. [Google Scholar] [PubMed]
  330. Huang, B.Z.; Le Marchand, L.; Haiman, C.A.; Monroe, K.R.; Wilkens, L.R.; Zhang, Z.-F.; Setiawan, V.W. Atopic allergic conditions and pancreatic cancer risk: Results from the Multiethnic Cohort Study. Int. J. Cancer 2018, 142, 2019–2027. [Google Scholar] [CrossRef]
  331. Tirado-Rodríguez, B.; Huerta-Yépez, S. Allergies: Diseases closely related to cancer. Bol. Med. Hosp. Infant. Mex. 2016, 73, 432–445. [Google Scholar] [CrossRef]
  332. Vesely, M.D.; Kershaw, M.H.; Schreiber, R.D.; Smyth, M.J. Natural innate and adaptive immunity to cancer. Annu. Rev. Immunol. 2011, 29, 235–271. [Google Scholar] [CrossRef] [PubMed]
  333. Fu, S.L.; Pierre, J.; Smith-Norowitz, T.A.; Hagler, M.; Bowne, W.; Pincus, M.R.; Mueller, C.M.; Zenilman, M.E.; Bluth, M.H. Immunoglobulin E antibodies from pancreatic cancer patients mediate antibody-dependent cell-mediated cytotoxicity against pancreatic cancer cells. Clin. Exp. Immunol. 2008, 153, 401–409. [Google Scholar] [CrossRef]
  334. Maisonneuve, P.; Lowenfels, A.B.; Bueno-de-Mesquita, H.B.; Ghadirian, P.; Baghurst, P.A.; Zatonski, W.A.; Miller, A.B.; Duell, E.J.; Boffetta, P.; Boyle, P. Past medical history and pancreatic cancer risk: Results from a multicenter case-control study. Ann. Epidemiol. 2010, 20, 92–98. [Google Scholar] [CrossRef] [PubMed]
  335. Singleton, P.A.; Moss, J.; Karp, D.D.; Atkins, J.T.; Janku, F. The mu opioid receptor: A new target for cancer therapy? Cancer 2015, 121, 2681–2688. [Google Scholar] [CrossRef]
  336. Wang, F.; Roy, S. Gut Homeostasis, Microbial Dysbiosis, and Opioids. Toxicol. Pathol. 2017, 45, 150–156. [Google Scholar] [CrossRef]
  337. Shakeri, R.; Kamangar, F.; Mohamadnejad, M.; Tabrizi, R.; Zamani, F.; Mohamadkhani, A.; Nikfam, S.; Nikmanesh, A.; Sotoudeh, M.; Sotoudehmanesh, R.; et al. Opium use, cigarette smoking, and alcohol consumption in relation to pancreatic cancer. Medicine 2016, 95, e3922. [Google Scholar] [CrossRef]
  338. Moossavi, S.; Mohamadnejad, M.; Pourshams, A.; Poustchi, H.; Islami, F.; Sharafkhah, M.; Mirminachi, B.; Nasseri-Moghaddam, S.; Semnani, S.; Shakeri, R.; et al. Opium Use and Risk of Pancreatic Cancer: A Prospective Cohort Study. Cancer Epidemiol. Biomark. Prev. 2018, 27, 268–273. [Google Scholar] [CrossRef] [PubMed]
  339. Barlass, U.; Deshmukh, A.; Beck, T.; Bishehsari, F. Opioid use as a potential risk factor for pancreatic cancer in the United States: An analysis of state and national level databases. PLoS ONE 2021, 16, e0244285. [Google Scholar] [CrossRef]
  340. Sun, M.; Lin, J.-A.; Chang, C.-L.; Wu, S.-Y.; Zhang, J. Association between long-term opioid use and cancer risk in patients with chronic pain: A propensity score-matched cohort study. Br. J. Anaesth. 2022, 129, 84–91. [Google Scholar] [CrossRef]
  341. Poly, T.N.; Islam, M.M.; Walther, B.A.; Lin, M.-C.; Li, Y.-C.K. Proton Pump Inhibitors Use and the Risk of Pancreatic Cancer: Evidence from Eleven Epidemiological Studies, Comprising 1.5 Million Individuals. Cancers 2022, 14, 5357. [Google Scholar] [CrossRef]
  342. Zhang, M.-L.; Fan, Y.-X.; Meng, R.; Cai, W.-K.; Yin, S.-J.; Zhou, T.; Huang, Y.-H.; Wang, P.; Jiang, F.-F.; Yang, M.; et al. Proton Pump Inhibitors and Cancer Risk: An Umbrella Review and Meta-analysis of Observational Studies. Am. J. Clin. Oncol. 2022, 45, 475–485. [Google Scholar] [CrossRef]
  343. Fried, M.; Siegrist, H.; Frei, R.; Froehlich, F.; Duroux, P.; Thorens, J.; Blum, A.; Bille, J.; Gonvers, J.J.; Gyr, K. Duodenal bacterial overgrowth during treatment in outpatients with omeprazole. Gut 1994, 35, 23–26. [Google Scholar] [CrossRef]
  344. Smith, J.P.; Fonkoua, L.K.; Moody, T.W. The Role of Gastrin and CCK Receptors in Pancreatic Cancer and other Malignancies. Int. J. Biol. Sci. 2016, 12, 283–291. [Google Scholar] [CrossRef] [PubMed]
  345. Nagata, N.; Nishijima, S.; Kojima, Y.; Hisada, Y.; Imbe, K.; Miyoshi-Akiyama, T.; Suda, W.; Kimura, M.; Aoki, R.; Sekine, K.; et al. Metagenomic Identification of Microbial Signatures Predicting Pancreatic Cancer from a Multinational Study. Gastroenterology 2022, 163, 222–238. [Google Scholar] [CrossRef]
  346. Schernhammer, E.S.; Kang, J.-H.; Chan, A.T.; Michaud, D.S.; Skinner, H.G.; Giovannucci, E.; Colditz, G.A.; Fuchs, C.S. A prospective study of aspirin use and the risk of pancreatic cancer in women. J. Natl. Cancer Inst. 2004, 96, 22–28. [Google Scholar] [CrossRef]
  347. Zhang, Y.-P.; Wan, Y.-D.; Sun, Y.-L.; Li, J.; Zhu, R.-T. Aspirin might reduce the incidence of pancreatic cancer: A meta-analysis of observational studies. Sci. Rep. 2015, 5, 15460. [Google Scholar] [CrossRef]
  348. Kirkegård, J.; Lund, J.L.; Mortensen, F.V.; Cronin-Fenton, D. Statins and pancreatic cancer risk in patients with chronic pancreatitis: A Danish nationwide population-based cohort study. Int. J. Cancer 2020, 146, 610–616. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Modifiable and non-modifiable risk factors in pancreatic cancer.
Figure 1. Modifiable and non-modifiable risk factors in pancreatic cancer.
Life 14 00980 g001
Figure 2. Genetic mutations grouped according to the risk of developing PC.
Figure 2. Genetic mutations grouped according to the risk of developing PC.
Life 14 00980 g002
Figure 3. Pancreatic cystic lesions and the risk of PC.
Figure 3. Pancreatic cystic lesions and the risk of PC.
Life 14 00980 g003
Figure 4. Risk of pancreatic cancer related to glucose level and onset of DM2.
Figure 4. Risk of pancreatic cancer related to glucose level and onset of DM2.
Life 14 00980 g004
Table 1. The screening recommendations for those who carry a germline mutation.
Table 1. The screening recommendations for those who carry a germline mutation.
Gene MutationPancreatic Cancer RiskCAPS Consensus for SurveillanceAt What Age Should Surveillance Begin? According to CAPS
BRCA1Inconsistent data about the riskIf at least one affected FDR
BRCA 2Inconsistent data about the riskIf at least one affected FDR or ≥two affected blood relatives50 years
Lynch syndrome—MLH1/MSH2/MSH68.6-fold increased riskIf at least one affected FDR
FAP—APC mutationIncreased up to 4 timesNo data
Peuts–Jeghers syndrome—LKB1/STK11132-increased riskRegardless of family history40 years
FAMMM with CDKN2A mutation leading to changes in the p16 protein13 to 67-fold increased riskRegardless of family history40 years, and if worrisome features → EUS+/−FNA and close follow-up imaging at 3–6 months
Hereditary pancreatitis—SPINK 1 mutation/PRSS1 mutations53-fold increased riskRegardless of family history40 years or 20 years after the first pancreatitis attack
PALB2 At least one affected FDR50 years
CFTR5.3 increased riskNo data
Ataxia-teleangiectasia ATM mutation At least one affected FDR50 years
Table 2. PC risk factors.
Table 2. PC risk factors.
Risk FactorImpact on RiskStudies on RiskWhat Can Be Done?
SmokingUp to 2.7-fold increaseBosetti et al. [115]
Anderson et al. [116]
Lugo et al. [117]
Smoking cessation campaign
Advocating for alternative nicotine-delivering products for those who do not want to quit
Diabetes14–15-fold increased risk in the first year
2.4–5-fold increased risk in the later years
Dankner et al. [144]
Bosetii C et al. [156]
Advocating for a healthy lifestyle and exercise
Validating, in prospective studies, the END-PAC model or refined models (adding a family history of pancreatic cancer, a personal history of pancreatitis, COPD, and uric acid) to find the patient at increased risk and including them in screening programs
Favoring the use of metformin as the first-line anti-diabetic treatment
Obesity, diet and lifestyle20–30% increased riskBerrington de Gonzáles et al. [28]
Genkinger et al. [31]
Stolzenberg-Solomon RZ et al. [178]
Arslan et al. [179]
Advocating for a normal weight, physical activity, and a diet rich in antioxidants such as fruits, nuts, and vegetables, avoiding the use of high amounts of meat and large amounts of soft drinks
PancreatitisUp to a 24-fold increased riskIkeura et al. [225]
Hart et al. [226]
Avoiding excessive alcohol consumption
Screening for patients with AP aged 57–75 years old, without the two most common ethologic factors of acute pancreatitis
Screening for genetic mutations in those with CP and then further screening according to guidelines
Alcohol Up to 1.36-fold increased riskTramacere et al. [231]
Wang et al. [232]
Gapstur et al. [233]
Avoiding excessive alcohol consumption
CoffeeInconsistent dataLi et al. [239]
Nie et al. [240]
Zhou et al. [243]
Ran et al. [244]
Lukic et al. [245]
Moderate consumption until further studies are conducted
Viral hepatitis Huang et al. [254]
Hassan et al. [255]
Xu et al. [256]
Vaccination against Hepatitis B virus, treatment for hepatitis C virus
Gallbladder diseases and cholecystectomyInconsistent dataFan et al. [260]
Luo et al. [261]
Rosato et al. [263]
Shabanzadeh et al. [265]
Promovate
Periodontal diseases Jingru Yu et al. [268]
Chang et al. [268]
Michaud et al. [269]
Good oral hygiene
Helicobacter PyloriControversial dataRisch HA et al. [38]
Huang J et al. [277]
Bulajic M et al. [278]
Cullin N et al. [279]
Promoting a balanced diet
Autoimmune diseasesSystemic and cutaneous lupus may be associated with an increased risk, but the data are controversialZhang M et al. [282]A special focus on patients with autoimmune diseases and their treatment
POCS3.4-fold increased riskPeeri et al. [285]
Yin et al. [284]
Considering the correlation of POCS with obesity and glucose intolerance; perhaps a balanced diet could influence both conditions
MicrobiotaInconsistent dataPushalkar S et al. [289]
Liu J et al. [298]
Santoni M et al. [299]
Further studies needed
Psychological stressIncreased riskKennedy B et al. [308]
Huang et al. [309]
Psychological and/or psychiatric support for very stressful events
Renin-angiotensin inhibitorsMay reduce the risk of pancreatic cancer—further data neededLee et al. [318]
Tse et al. [319]
With a doubtful effect, promoting the use of ACI for those who need antihypertensive treatment
Statins No effect on PC riskKirkegård et al. [348]Not appliable
Allergies May reduce the riskGandini et al. [323]
Cotterchio M et al. [324]
Wang et al. [325]
Gandini et al. [323]
Karim et al. [329]
Not appliable
OpioidsMay increase the riskShakeri et al. [337]
Moossavi et al. [338]
Barlass et al. [339]
Sun et al. [340]
Limiting the use of opioids; anti-drug campaigns
NSAIDInconsistent dataSchernhammer ES [346]
Zhang et al. [347]
Inconsistent data
PPI63% increased riskPoly et al. [341]
Zhang et al. [342]
Limiting the use; use just for those who need it
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Grigorescu, R.R.; Husar-Sburlan, I.A.; Gheorghe, C. Pancreatic Cancer: A Review of Risk Factors. Life 2024, 14, 980. https://doi.org/10.3390/life14080980

AMA Style

Grigorescu RR, Husar-Sburlan IA, Gheorghe C. Pancreatic Cancer: A Review of Risk Factors. Life. 2024; 14(8):980. https://doi.org/10.3390/life14080980

Chicago/Turabian Style

Grigorescu, Raluca Roxana, Ioana Alexandra Husar-Sburlan, and Cristian Gheorghe. 2024. "Pancreatic Cancer: A Review of Risk Factors" Life 14, no. 8: 980. https://doi.org/10.3390/life14080980

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

Article Metrics

Back to TopTop