ArticlePDF AvailableLiterature Review

Abstract

Pancreatic cancer is without any doubt the malignancy with the poorest prognosis and the lowest survival rate. This highly aggressive disease is rarely diagnosed at an early stage and difficult to treat due to its resistance to radiotherapy and chemotherapy. Therefore, there is an urgent need to clarify the causes responsible for pancreatic cancer and to identify preventive strategies to reduce its incidence in the population. Some circumstances, such as smoking habits, being overweight and diabetes, have been identified as potentially predisposing factors to pancreatic cancer, suggesting that diet might play a role. A diet low in fat and sugars, together with a healthy lifestyle, regular exercise, weight reduction and not smoking, may contribute to prevent pancreatic cancer and many other cancer types. In addition, increasing evidence suggests that some food may have chemo preventive properties. Indeed, a high dietary intake of fresh fruit and vegetables has been shown to reduce the risk of developing pancreatic cancer, and recent epidemiological studies have associated nut consumption with a protective effect against it. Therefore, diet could have an impact on the development of pancreatic cancer and further investigations are needed to assess the potential chemo preventive role of specific foods against this disease. This review summarizes the key evidence for the role of dietary habits and their effect on pancreatic cancer and focuses on possible mechanisms for the association between diet and risk of pancreatic cancer.
Review
Diet and Pancreatic Cancer Prevention
Ilaria Casari and Marco Falasca *
Received: 12 August 2015; Accepted: 10 November 2015; Published: 23 November 2015
Academic Editor: Hildegard M. Schuller
Metabolic Signalling Group, School of Biomedical Sciences, CHIRI Biosciences, Curtin University,
Perth 6102, Australia; ilaria.casari@curtin.edu.au
*Correspondence: marco.falasca@curtin.edu.au; Tel.: +61-8-9266-9712
Abstract: Pancreatic cancer is without any doubt the malignancy with the poorest prognosis and
the lowest survival rate. This highly aggressive disease is rarely diagnosed at an early stage and
difficult to treat due to its resistance to radiotherapy and chemotherapy. Therefore, there is an urgent
need to clarify the causes responsible for pancreatic cancer and to identify preventive strategies
to reduce its incidence in the population. Some circumstances, such as smoking habits, being
overweight and diabetes, have been identified as potentially predisposing factors to pancreatic
cancer, suggesting that diet might play a role. A diet low in fat and sugars, together with a healthy
lifestyle, regular exercise, weight reduction and not smoking, may contribute to prevent pancreatic
cancer and many other cancer types. In addition, increasing evidence suggests that some food may
have chemo preventive properties. Indeed, a high dietary intake of fresh fruit and vegetables has
been shown to reduce the risk of developing pancreatic cancer, and recent epidemiological studies
have associated nut consumption with a protective effect against it. Therefore, diet could have an
impact on the development of pancreatic cancer and further investigations are needed to assess
the potential chemo preventive role of specific foods against this disease. This review summarizes
the key evidence for the role of dietary habits and their effect on pancreatic cancer and focuses on
possible mechanisms for the association between diet and risk of pancreatic cancer.
Keywords: pancreatic cancer; cancer prevention; diet; obesity
1. Introduction
Pancreatic cancer is an aggressive disease which holds the gloomy record of having become one
of the most deadly malignancies in the USA, being the fourth leading cause of cancer-related death
in the USA, despite holding the 10th place in the incident rate scale [1,2]. Due to the difficulty in
obtaining an early diagnosis and to its resistance to treatment, pancreatic cancer has a very poor
prognosis, with a five-year survival rate of 7%, according to the American Cancer Society. Although
recent studies have pointed out the diversity and complexity of pancreatic cancer genetics, some
predisposing factors have been identified for this disease. Genetic factors are the main overt factors
responsible, followed by other DNA damage-inducing causes such as age, smoking (implicated in
20%–25% of cases), type 2 diabetes and chronic pancreatitis [3,4]. However, dietary components are
also thought to play a part in the development of this disease, as obesity, and high consumption of
red meat and fried foods are all risk factors. Conversely and according to some studies, a diet rich in
vegetables, fresh fruit, nuts and whole grain is useful in the prevention of pancreatic cancer [1,57].
In this review, we will discuss the possible links between diet and pancreatic cancer, analysing the
potential role of diet in promoting or preventing the onset of this disease.
Cancers 2015,7, 2309–2317; doi:10.3390/cancers7040892 www.mdpi.com/journal/cancers
Cancers 2015,7, 2309–2317
2. Pancreatic Cancer Progression Model
The pancreas is an elongated organ situated behind the stomach that holds digestive and
hormonal functions. Digestive enzymes are secreted by its exocrine gland, while the islets of
Langerhans, which constitute the endocrine gland, are devolved to hormones secretion. The vast
majority of tumours originate in the exocrine gland and, amongst them, 90% are pancreatic ductal
adenocarcinoma (PDAC), which is also the most aggressive form of pancreatic cancer. Due to the fact
that, especially in its early stages, pancreatic cancer can be asymptomatic or associated to non-specific
symptoms (they may include nausea, indigestion, weight loss, back pain, abdominal pain, jaundice,
steatorrhea and depression), it is rarely possible to obtain an early diagnosis and this therefore
implies an even greater difficulty of treatment [8]. The development of PDAC is very complex
and it is characterized by a sequence of precursor lesions, which occur at different stages of time
and ultimately evolve into invasive cancer. These lesions are known as pancreatic intraepithelial
neoplasia (PanIN), intraductal papillary mucinous neoplasms and mucinous cystic neoplasm. The
majority of PDACs develop from PanINs, and have been classified according to their morphological,
cytological, and genetic transformations, each of which leads to significant alterations of the signalling
pathway. Three stages, characterized by increasingly atypical cells, have been identified and called
PanIN1 (sub classified into PanIN1a and PanIN1B), PanIN2 and PanIN3 [9]. One of the early events
in the progression of the disease is the occurrence of Kras gene mutations at codon 12 in the normal
pancreatic epithelial cells, which are a characteristic of 90% of all PDAC. These mutations induce
cellular proliferation, invasion and survival. In the intermediate stage of the disease, the inhibition
of the p16 tumour suppressor gene occurs, promoting further cytological and architectural atypia in
the duct cells. In a later stage, the inactivation of the p53 tumour suppressor gene (TP53), which is
normally responsible for DNA repairing, cell division blocking and apoptosis activation, takes place
in 70% of all PDAC. In the late stage of the disease, inactivation of the SMAD4 gene involved in the
cell signalling pathway also occurs in 55% of all PDAC [10].
PDAC Defining Features
PDAC defining features can be summarized in: altered metabolism, desmoplasia, and hypo
vascularization. In order to survive and proliferate in the new micro-environment, pancreatic
cancer cells are forced to be subjected to a metabolic reprogramming and they rely on anabolic
reactions to synthetized de novo proteins, nucleic acids and lipids. Oncogenic Kras has been found
to play a key part in PDAC metabolism rearrangement. One of the most important constituents of
pancreatic cancer cells metabolism is autophagy. Autophagy is a very important catabolic process,
regulated by several protein complexes, whose function is to recycle unneeded or damaged cellular
particles, protein or molecular complexes, in order to maintain cells homeostasis. This function is
also anti-tumorigenic as it contributes to control pro-tumorigenic elements such as tissue damage,
oxidative stress and genomic imbalance. On the contrary, when increased above the baseline in
established tumours, autophagy becomes a pro-tumorigenic factor by furnishing cancer cells with
nutrients and energy [11]. Desmoplasia, which is another key feature of PDAC, is a complex reaction
of pancreatic stroma that involves stroma components such as stellate cells, leukocytes, endothelial
cells, fibroblasts, and extracellular matrix, as well as invading tumour cells and growth factors, and
results in fibrotic tissue formation. The characteristic hypo vascularization of the pancreatic tissue is
likely to be the cause of the presence of hypoxic areas within PDAC, which stimulate cancer cell to
adapt their metabolism to the new microenvironment and have also been demonstrated to affect the
efficacy of chemotherapy treatments [1113].
3. Obesity and Pancreatic Cancer
The link between obesity (body mass index > 30.0 Kg/m2), type 2 diabetes, cardiovascular
diseases and cancer has long been established, even though the mechanisms by which extra fat
2310
Cancers 2015,7, 2309–2317
deposits increase cancer risk have yet to be fully elucidated. This is a dreary perspective considering
that, according to the 2010 Health and Nutrition Examination Survey (NHANES), 35.5% of the USA
adult population and 17% of children and teenagers are obese [14,15]. Pancreatic cancer is currently
within the list of obesity-related cancers, together with colon, oesophageal, kidney, endometrial and
postmenopausal breast cancer [15]. Obesity or a high-fat diet, is one of the factors that can increase
the risk of developing acute pancreatitis [16,17], by changing the balance of digesting enzymes
within acinar cells and lowering pancreatic enzyme secretion. Acute pancreatitis is characterized
by an inflammatory state of the pancreas and by dysfunctional autophagy in pancreatic cells. In
addition, by increasing the levels of the pro-inflammatory hormone leptin and decreasing the levels
of the anti-inflammatory hormone adiponectin, obesity promotes inflammation. While normally
inflammation is a natural response of the body, which activates immune cells using cytokines,
chemokines and other mediators [18], persistent inflammation can lead to several cell damages
caused by metabolic changes and oxidative stress. Similarly, obesity, promoting the activation of
Akt and mTOR signalling pathways and down-regulating autophagy genes, such as Ulk1/Atg1
and Atg5, Atg6/Beclin1, inhibits autophagy, a cell defence mechanism which involves degradation
and recycling of damaged cellular components and that controls inflammation [19]. Autophagy
can also mediate mechanisms of chemoresitance of cancer cells to anticancer drugs. In response
to metabolic and therapeutic stresses, autophagy induces cell death, increases inflammation and
promotes tumorigenesis [2022]. Moreover, the breakdown of excessive pancreatic fat caused by
obesity produces a surplus of unsaturated fatty acids that can increase inflammation, parenchymal
necrosis and lead to multi-organ damage and death [17]. Furthermore, unresolved or recurrent
acute pancreatitis that shows a persistent low-grade inflammation can activate pancreatic stellate
cells. These cells, which normally have the function of storing Vitamin-A lipid droplets in the
cytoplasm, upon activation during pancreatic injuries, promote fibrogenesis, leading to chronic
pancreatitis with an increased risk (around 5% of patients) of developing pancreatic cancer. In
addition, the exceeding production of cytokines induced by the excessive number of immune
cells stimulated by inflammation, can lead to the activation of oncogenic Kras, representing the
initial switch for Kras activation followed by mutations to an oncogenic form, a typical feature of
almost 90% of all pancreatic adenocarcinoma [19]. As well as many other cancer cells, one of the
characteristic features of pancreatic cancer cells is the substantial alteration of cellular metabolism,
which, together with genetic and epigenetic alteration, promotes tumour growth. In the early stages
of cancer development, lipid biosynthesis, de novo lipogenesis, provides extra lipids necessary for
the generation of biological membranes, energy store, and signalling functions [23]. It has been
demonstrated that lipoprotein catabolism and cholesterol synthesis are very stimulated in PDAC,
as tumour cells require high level of cholesterol [24]. Recent findings indicate that cancer cells can
utilize diet-derived fats present in blood, together with de novo lipogenesis, to satisfy lipid necessities,
strengthening the link between obesity, high fat diet, and cancer risk [2528].
3.1. Epidemiological Studies
Recent epidemiological studies have also collected evidence supporting the connection between
obesity and risk of pancreatic cancer. In the Metabolic Syndrome and Cancer Project, a study
population of 577,315 individuals was observed for about 12 years follow-up and 315 women and
547 men were diagnosed with pancreatic cancer. As a result, a positive correlation between body
mass index and risk of pancreatic cancer emerged, although only for women [29]. Results from a
study conducted between 2008 and 2015 on 110 patients, where half of them were overweight or
obese, showed a direct link between precancerous lesions of the pancreas, pancreatic fatty infiltration,
intralobular fibrosis, subcutaneous and intravisceral fat and a high BMI. The authors also found the
number of PanIN lesions to be correlated with the percentage of intravisceral fat, which was not
found to be localized around the lesions. For this reason, they hypothesised fatty infiltrations to be the
cause of PanIN lesions and not vice versa [30]. A large case-control study conducted in USA associated
2311
Cancers 2015,7, 2309–2317
obesity to a 50%–60% elevated risk of developing pancreatic cancer, particularly amongst women and
black subjects [31]. A positive association between pancreatic cancer risk and obesity and an increased
risk, especially for women in the presence of waist localized fat, was also underlined by a pooled
analysis from the National Cancer Institute Pancreatic Cancer Cohort Consortium (PanScan) [32]. A
case-control study, conducted in the USA on 841 patients with pancreatic adenocarcinoma and 754
healthy individuals, found that obesity and overweight in early adulthood determined an increased
risk of developing pancreatic cancer and led to precocity in the onset of this disease. Moreover, this
study showed that survival rates of patients with pancreatic cancer were affected by the presence of
obesity at an older age [33]. High BMI was also associated to poor survival in a retrospective study
conducted on advanced or metastatic pancreatic cancer patients between 1994 and 2004 [34]. A pooled
analysis of 14 cohort studies involving 846,340 persons found a 54% higher risk of pancreatic cancer
for people obese at baseline and those who were overweight in their early adulthood. In addition,
a 40% increased risk for individuals who had gained weight, compared to individuals who had a
constant weight, and also an increase for persons with a high waist-to hip ratio was observed [35].
3.2. Experimental Studies
Experimental studies on animals also provided data to corroborate the connection between
obesity and pancreatic cancer risk. Hamsters fed with a high fat diet and treated with
N-nitrosobis(2-oxopropyl)amine (BOP) developed hyperlipidaemia and intrapancreatic fatty
infiltration which led to pancreatic ductal adenocarcinoma in 67% of the animals, compared to 0%
in the control group fed with a standard diet [36]. A study conducted on a genetically engineered
mouse model revealed that mice fed with a HFD had an increased activation of oncogenic Kras via
pro-inflammatory factor COX-2, compared to the control group. This resulted in an enhancement
of the number of precancerous lesions of the pancreas and pancreatic ductal adenocarcinoma in the
HFD group [37]. Taken together these data strongly support the theory that obesity and a high fat
diet, by causing intrapancreatic fatty infiltration and promoting inflammation in the pancreas, trigger
a chain reaction leading to the activation of oncogenic Kras signalling and to the developing of chronic
pancreatitis and PanIn lesions, all well-known prerequisites of pancreatic cancer [38].
4. Diet and Pancreatic Cancer
Epidemiological and experimental studies have consistently shown the direct link between
obesity, high BMI, weight gain and elevated risk of developing pancreatic cancer. As a consequence, it
is logical to affirm that a high-calorie diet, and/or high consumptions of fats and sugars, predisposing
over time to overweight or obesity, has a negative impact on pancreatic cancer risk. In addition, a
high consumption of red meat has been found to elevate the risk of several types of cancer, including
pancreatic cancer [39]. On the other hand, there is also evidence that a healthy diet can have a role
in protecting against pancreatic cancer. In the 2010 Dietary guidelines for Americans (DGA), the
recommendation focuses on maintaining a healthy weight through consuming the right amount of
calories and nutrient-dense foods, together with the suggestion of eating a diet rich in fruit, vegetables
and whole grains [5].
4.1. Phytochemicals and Dietary Fibre
It is well known that fruit, vegetables, whole grains and nuts contain elevated amounts
of phytochemicals, bioactive compounds that can provide protection against several chronic
diseases and cancer and that are classified as carotenoids, phenolics, alkaloids, nitrogen-containing
compounds and organosulfur compounds [4043]. Although their mechanisms of action are yet
to be fully elucidated, phytochemicals have been discovered to possess an additive and synergistic
action, which would account for their anticancer properties [44]. Some of the mechanisms proposed
to explain phytochemicals anticancer properties include: antioxidant and anti-inflammatory action;
inhibition of cell proliferation, differentiation, adhesion and invasion; anti-bacterial and anti-viral
2312
Cancers 2015,7, 2309–2317
effects and stimulation of immune functions; DNA damage repair; regulation of steroid hormone and
oestrogen metabolism; regulation of signal transduction pathways; enzyme regulation; inhibition of
oncogene suppression and induction of tumour suppress gene expression; activation of cell cycle G
arrest; induction of cell differentiation and apoptosis [44]. In addition to the role of phytochemicals,
dietary fibre, one of the main components of fruit, vegetables, whole grains and nuts, has been
found to have an inverse correlation with cancer risk. A case control study on 326 pancreatic cancer
patients in Italy found soluble and insoluble fibre and fibre from fruit to be inversely associated to
pancreatic cancer, even though no association between grain fibre and risk of pancreatic cancer was
established [6]. On the contrary, whole grains were found to have a protective effect against several
types of cancers, including pancreatic cancer, in a study on Mediterranean diet and cancer risk [39].
In the Nurses’ health study, a prospective study on 75,680 women, individuals who consumed a
28 g portion of nuts, two or more times per week were associated with a significant diminished
risk of developing pancreatic cancer [45]. A case control study within EPIC cohort study (European
Prospective Investigation into Cancer and Nutrition) recently reported an inverse correlation between
plasma levels of beta-carotene (contained in orange fruit and vegetables and dark green leafy
vegetables), zeaxantin (contained in paprika, corn and wolfberries) and alpha-tocopherol (contained
in green and orange vegetables and tomatoes) and pancreatic cancer risk [46]. Moreover, some
isothiocyanates, compounds contained in cruciferous vegetables (i.e., broccoli, cauliflower, cabbage,
and Brussels sprouts), such as sulforaphane, benzyl isothiocyanate and phenethyl isothiocyanate,
have been shown to have an inhibitory effect on pancreatic cancer cells in in vitro and in animal
studies [4751].
4.2. Dietary Compounds and Autophagy
Autophagy can have different roles in cancer depending on tumour types and context [20].
Indeed, during the first stages of tumour progression autophagy prevents genomic instability
and blocks tumour initiation, whereas in advanced states of the disease, autophagy, through
the degradation and recycling of cellular components, contributes to the increased demand for
rapid growth of cancer cells. PDAC are characterized by high levels of basal autophagy, and
pharmacological or genetic suppression of autophagy have been shown to inhibit pancreatic cancer
growth in vivo and in vitro [11]. Therefore, autophagy is required for tumour growth of PDAC,
and drugs that inhibit this process have been proposed for clinical testing in PDAC patients, as
well as in other tumour showing a similar dependence on autophagy. As a consequence of this
increasing interest in targeting this process, there are currently several clinical trials involving
autophagy inhibitors, such as chloroquine and its derivatives, worldwide. Among the different
mechanisms through which dietary compounds can affect the risk of cancer, autophagy is emerging as
an important process affected by diet [52]. This is not surprising considering that nutrient availability
is the major regular of autophagy. Several dietary compounds have been shown to affect autophagy
such as quercetin, genistein, curcumin, sulforaphane and resveratrol [53]. In addition, Vitamin D has
been shown to influence autophagy and increasing evidence supports a potential role of vitamin D
and its analogues in preventing or treating pancreatic cancer [54,55]. Nevertheless, several questions
remain to be addressed, such as the dose and duration of exposures and tissue specificity in response
to bioactive compounds as well as the implications of changes in autophagy during the early stages
of tumour initiation.
4.3. Calorie Restriction
In addition to a diet rich in fruit, vegetables, nuts, and whole grains, calorie restriction is
another promising strategy that seems to be effective in protecting against cancer risk [56]. In
fact, by chronically controlling the amount of calories consumed (in animal model a 20% to 40%
reduction is usually implemented), a reduction in the level of insulin, insulin-like growth factor,
leptin, adiponectin, plasminogen activator inhibitor, cytokines and vascular endothelial growth factor
2313
Cancers 2015,7, 2309–2317
is obtained. These changes contribute to lower inflammation and growth factor signalling, and to
diminish vascular disorders, causing a reduction in cancer risk and cancer progression [56]. These
findings clearly show a link between a high-calorie diet and the probability of developing pancreatic
cancer and suggest a plan of action from a preventive perspective.
5. Conclusions
Given the extreme aggressiveness of pancreatic cancer and the difficulties in achieving an early
diagnosis and an efficacious cure, it is mandatory to concentrate not only on finding new adequate
treatments, but also on effective preventive strategies. As overweightness, high BMI and obesity are
increasingly common in our society and can play a role in increasing the risk of pancreatic cancer,
targeting these conditions and implementing a healthier lifestyle in the global population could be a
method for having an impact in the prevention of pancreatic cancer, and also on other types of cancer
and chronic diseases. A diet rich in fruit and vegetables, nuts and whole grain foods, together with a
balanced calorie control, could be a valuable tool in future pancreatic cancer prevention strategies.
Acknowledgments: M.F. is supported by Pancreatic Cancer Research Fund.
Author Contributions: Ilaria Casari: idea, concept, collection of data, review, paper writing, and paper revision.
Marco Falasca: Idea, Collection of data, paper revision, and editing, concept, and review of paper.
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Li, Y.; Go, V.L.; Sarkar, F.H. The role of nutraceuticals in pancreatic cancer prevention and therapy: Targeting
cellular signaling, microRNAs, and epigenome. Pancreas 2015,44, 1–10. [CrossRef] [PubMed]
2. Li, H.-Y.; Cui, Z.M.; Chen, J.; Guo, X.Z.; Li, Y.Y. Pancreatic cancer: Diagnosis and treatments. Tumour Biol.
2015,36, 1375–1384. [CrossRef] [PubMed]
3. Stan, S.D.; Singh, S.V.; Brand, R.E. Chemoprevention strategies for pancreatic cancer. Nat. Rev.
Gastroenterol. Hepatol. 2010,7, 347–356. [CrossRef] [PubMed]
4. Magruder, J.T.; Elahi, D.; Andersen, D.K. Diabetes and pancreatic cancer: Chicken or egg? Pancreas 2011,
40, 339–351. [CrossRef] [PubMed]
5. Harris, D.M.; Srihari, P.; Go, V.L. Pancreatic cancer prevention and the 2010 Dietary Guidelines for
Americans. Pancreas 2011,40, 641–643. [CrossRef] [PubMed]
6. Bidoli, E.; Pelucchi, C.; Zucchetto, A.; Negri, E.; dal Maso, L.; Polesel, J.; Boz, G.; Montella, M.; Franceschi, S.;
Serraino, D.; et al. Fiber intake and pancreatic cancer risk: A case-control study. Ann Oncol. 2012,23,
264–268. [CrossRef] [PubMed]
7. Bao, Y.; Hu, F.B.; Giovannucci, E.L.; Wolpin, B.M.; Stampfer, M.J.; Willett, W.C.; Fuchs, C.S. Nut
consumption and risk of pancreatic cancer in women. Br. J. Cancer 2013,109, 2911–2916. [CrossRef]
[PubMed]
8. Garrido-Laguna, I.; Hidalgo, M. Pancreatic cancer: From state-of-the-art treatments to promising novel
therapies. Nat. Rev. Clin. Oncol. 2015,12, 319–334. [CrossRef] [PubMed]
9. Hruban, R.H.; Goggins, M.; Parsons, J.; Kern, S.E. Progression model for pancreatic cancer. Clin. Cancer Res.
2000,6, 2969–2972. [PubMed]
10. Mohammed, A.; Janakiram, N.B.; Lightfoot, S.; Gali, H.; Vibhudutta, A.; Rao, C.V. Early detection and
prevention of pancreatic cancer: Use of genetically engineered mouse models and advanced imaging
technologies. Curr. Med. Chem. 2012,19, 3701–3713. [CrossRef] [PubMed]
11. Sousa, C.M.; Kimmelman, A.C. The complex landscape of pancreatic cancer metabolism. Carcinogenesis
2014,35, 1441–1450. [CrossRef] [PubMed]
12. Provenzano, P.P.; Cuevas, C.; Chang, A.E.; Goel, V.K.; von Hoff, D.D.; Hingorani, S.R. Enzymatic targeting
of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell 2012,
21, 418–429. [CrossRef] [PubMed]
13. Oberstein, P.E.; Olive, K.P. Pancreatic cancer: Why is it so hard to treat? Ther. Adv. Gastroenterol. 2013,6,
321–337. [CrossRef] [PubMed]
2314
Cancers 2015,7, 2309–2317
14. Schmitz, K.H.; Neuhouser, M.L.; Agurs-Collins, T.; Zanetti, K.A.; Cadmus-Bertram, L.; Dean, L.T.;
Drake, B.F. Impact of obesity on cancer survivorship and the potential relevance of race and ethnicity.
J. Natl. Cancer Inst. 2013,105, 1344–1354. [CrossRef] [PubMed]
15. Azvolinsky, A. Cancer risk: The fat tissue-BMI-obesity connection. J. Natl. Cancer Inst. 2014,106. [CrossRef]
[PubMed]
16. Frossard, J.L.; Lescuyer, P.; Pastor, C.M. Experimental evidence of obesity as a risk factor for severe acute
pancreatitis. World J. Gastroenterol. 2009,15, 5260–5265. [CrossRef] [PubMed]
17. Navina, S.; Acharya, C.; DeLany, J.P.; Orlichenko, L.S.; Baty, C.J.; Shiva, S.S.; Durgampudi, C.; Karlsson, J.M.;
Lee, K.; Bae, K.T.; et al. Lipotoxicity causes multisystem organ failure and exacerbates acute pancreatitis in
obesity. Sci. Transl. Med. 2011,3. [CrossRef] [PubMed]
18. Mantovani, A.; Cassatella, M.A.; Costantini, C.; Jaillon, S. Neutrophils in the activation and regulation of
innate and adaptive immunity. Nat. Rev. Immunol. 2011,11, 519–531. [CrossRef] [PubMed]
19. Kolodecik, T.; Shugrue, C.; Ashat, M.; Thrower, E.C. Risk factors for pancreatic cancer: Underlying
mechanisms and potential targets. Front. Physiol. 2014,4, 415. [CrossRef] [PubMed]
20. Levine, B.; Kroemer, G. Autophagy in the pathogenesis of disease. Cell 2008,132, 27–42. [CrossRef]
[PubMed]
21. 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.
[CrossRef] [PubMed]
22. Kuraishy, A.; Karin, M.; Grivennikov, S.I. Tumor promotion via injury- and death-induced inflammation.
Immunity 2011,35, 467–477. [CrossRef] [PubMed]
23. Baenke, F.; Peck, B.; Miess, H.; Schulze, A. Hooked on fat: The role of lipid synthesis in cancer metabolism
and tumour development. Dis. Model Mech. 2013,6, 1353–1363. [CrossRef] [PubMed]
24. Guillaumond, F.; Bidaut, G.; Ouaissi, M.; Servais, S.; Gouirand, V.; Olivares, O.; Lac, S.; Borge, L.; Roques, J.;
Gayet, O.; et al. Cholesterol uptake disruption, in association with chemotherapy, is a promising combined
metabolic therapy for pancreatic adenocarcinoma. Proc. Natl. Acad. Sci. USA 2015,112, 2473–2478.
[CrossRef] [PubMed]
25. Nomura, D.K.; Long, J.Z.; Niessen, S.; Hoover, H.S.; Ng, S.W.; Cravatt, B.F. Monoacylglycerol lipase
regulates a fatty acid network that promotes cancer pathogenesis. Cell 2010,140, 49–61. [CrossRef]
[PubMed]
26. Louie, S.M.; Roberts, L.S.; Mulvihill, M.M.; Luo, K.; Nomura, D.K. Cancer cells incorporate and remodel
exogenous palmitate into structural and oncogenic signaling lipids. Biochim. Biophys. Acta 2013,1831,
1566–1572. [CrossRef] [PubMed]
27. Kuemmerle, N.B.; Rysman, E.; Lombardo, P.S.; Flanagan, A.J.; Lipe, B.C.; Wells, W.A.; Pettus, J.R.;
Froehlich, H.M.; Memoli, V.A.; Morganelli, P.M.; et al. Lipoprotein lipase links dietary fat to solid tumor
cell proliferation. Mol. Cancer Ther. 2011,10, 427–436. [CrossRef] [PubMed]
28. Zaidi, N.; Lupien, L.; Kuemmerle, N.B.; Kinlaw, W.B.; Swinnen, J.V.; Smans, K. Lipogenesis and lipolysis:
The pathways exploited by the cancer cells to acquire fatty acids. Prog. Lipid Res. 2013,52, 585–589.
[CrossRef] [PubMed]
29. Johansen, D.; Stocks, T.; Jonsson, H.; Lindkvist, B.; Björge, T.; Concin, H.; Almquist, M.; Häggström, C.;
Engeland, A.; Ulmer, H.; et al. Metabolic factors and the risk of pancreatic cancer: A prospective analysis
of almost 580,000 men and women in the Metabolic Syndrome and Cancer Project. Cancer Epidemiol.
Biomarkers Prev. 2010,19, 2307–2317. [CrossRef] [PubMed]
30. Rebours, V.; Gaujoux, S.; d’Assignies, G.; Sauvanet, A.; Ruszniewski, P.; Levy, P.; Bedossa, P.; Paradis, V.;
Couvelard, A. Obesity and fatty pancreatic infiltration are risk factors for pancreatic precancerous lesions
(PanIN). Clin. Cancer Res. 2015,21, 3522–3528. [CrossRef] [PubMed]
31. 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. [CrossRef]
[PubMed]
2315
Cancers 2015,7, 2309–2317
32. 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. [CrossRef] [PubMed]
33. Li, D.; Morris, J.S.; Liu, J.; Hassan, M.M.; Day, R.S.; Bondy, M.L.; Abbruzzese, J.L. Body mass index and
risk, age of onset, and survival in patients with pancreatic cancer. JAMA 2009,301, 2553–2562. [CrossRef]
[PubMed]
34. 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. [CrossRef] [PubMed]
35. Kasenda, B.; Bass, A.; Koeberle, D.; Pestalozzi, B.; Borner, M.; Herrmann, R.; Jost, L.; Lohri, A. Survival in
overweight patients with advanced pancreatic carcinoma: A multicentre cohort study. BMC Cancer 2014,
14, 728. [CrossRef] [PubMed]
36. Hori, M.; Kitahashi, T.; Imai, T.; Ishigamori, R.; Takasu, S.; Mutoh, M.; Sugimura, T.;
Wakabayashi, K.; Takahashi, M. Enhancement of carcinogenesis and fatty infiltration in the pancreas in
N-nitrosobis(2-oxopropyl)amine-treated hamsters by high-fat diet. Pancreas 2011,40, 1234–1240. [CrossRef]
[PubMed]
37. Philip, B.; Roland, C.L.; Daniluk, J.; Liu, Y.; Chatterjee, D.; Gomez, S.B.; Ji, B.; Huang, H.; Wang, H.;
Fleming, J.B.; et al. A high-fat diet activates oncogenic Kras and COX2 to induce development of pancreatic
ductal adenocarcinoma in mice. Gastroenterology 2013,145, 1449–1458. [CrossRef]
38. Wang, H.; Maitra, A.; Wang, H. Obesity, intrapancreatic fatty infiltration, and pancreatic cancer.
Clin. Cancer Res. 2015,21, 3369–3371. [CrossRef] [PubMed]
39. La Vecchia, C. Association between Mediterranean dietary patterns and cancer risk. Nutr. Rev. 2009,67,
S126–S129. [CrossRef] [PubMed]
40. Liu, R.H. Potential synergy of phytochemicals in cancer prevention: Mechanism of action. J. Nutr. 2004,
134, 3479S–3485S. [PubMed]
41. Awad, A.B.; Fink, C.S. Phytosterols as anticancer dietary components: Evidence and mechanism of action.
J. Nutr. 2000,130, 2127–2130. [PubMed]
42. Falasca, M.; Casari, I.; Maffucci, T. Cancer chemoprevention with nuts. J. Natl. Cancer Inst. 2014,106, 238.
[CrossRef] [PubMed]
43. Falasca, M.; Casari, I. Cancer chemoprevention by nuts: Evidence and promises. Front. Biosci. (Schol. Ed.)
2012,4, 109–120. [CrossRef] [PubMed]
44. Liu, R.H. Dietary bioactive compounds and their health implications. J. Food Sci. 2013,78, A18–A25.
[CrossRef] [PubMed]
45. 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. [CrossRef]
[PubMed]
46. Jeurnink, S.M.; Ros, M.M.; Leenders, M.; van Duijnhoven, F.J.; Siersema, P.D.; Jansen, E.H.; 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. [CrossRef] [PubMed]
47. Kallifatidis, G.; Rausch, V.; Baumann, B.; Apel, A.; Beckermann, B.M.; Groth, A.; Mattern, J.; Li, Z.; Kolb, A.;
Moldenhauer, G.; et al. Sulforaphane targets pancreatic tumour-initiating cells by NF-kappaB-induced
antiapoptotic signalling. Gut 2009,58, 949–963. [CrossRef] [PubMed]
48. Srivastava, S.K.; Singh, S.V. Cell cycle arrest, apoptosis induction and inhibition of nuclear factor kappa
B activation in anti-proliferative activity of benzyl isothiocyanate against human pancreatic cancer cells.
Carcinogenesis 2004,25, 1701–1709. [CrossRef] [PubMed]
49. 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. [CrossRef] [PubMed]
2316
Cancers 2015,7, 2309–2317
50. 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. [CrossRef] [PubMed]
51. Son, H.Y.; Nishikawa, A.; Furukawa, F.; Lee, I.S.; Ikeda, T.; Miyauchi, M.; Nakamura, H.;
Hirose, M. Modifying effects of 4-phenylbutyl isothiocyanate on N-nitrosobis(2-oxopropyl)amine-induced
tumorigenesis in hamsters. Cancer Lett. 2000,160, 141–147. [CrossRef]
52. Hasima, N.; Ozpolat, B. Regulation of autophagy by polyphenolic compounds as a potential therapeutic
strategy for cancer. Cell Death Dis. 2014,5, e1509. [CrossRef] [PubMed]
53. Singletary, K.; Milner, J. Diet, autophagy, and cancer: A review. Cancer Epidemiol. Biomarkers Prev. 2008,17,
1596–1610. [CrossRef] [PubMed]
54. Barreto, S.G.; Neale, R.E. Vitamin D and pancreatic cancer. Cancer Lett. 2015,368, 1–6. [CrossRef] [PubMed]
55. Sherman, M.H.; Yu, R.T.; Engle, D.D.; Ding, N.; Atkins, A.R.; Tiriac, H.; Collisson, E.A.; Connor, F.;
van Dyke, T.; Kozlov, S.; et al. Vitamin D receptor-mediated stromal reprogramming suppresses pancreatitis
and enhancespancreatic cancer therapy. Cell 2014,159, 80–93. [CrossRef] [PubMed]
56. Hursting, S.D.; Dunlap, S.M.; Ford, N.A.; Hursting, M.J.; Lashinger, L.M. Calorie restriction and cancer
prevention: A mechanistic perspective. Cancer Metab. 2013,1, 10. [CrossRef]
© 2015 by the authors; licensee MDPI, Basel, Switzerland. This article is an open
access article distributed under the terms and conditions of the Creative Commons by
Attribution (CC-BY) license (http://creativecommons.org/licenses/by/4.0/).
2317
... Although the incidence rate of pancreatic cancer (PC) is considerably low, its mortality rate is the highest among all major cancers. [1,2] This could be attributed to the inferior prognosis of PC, which is usually diagnosed at developed stages, and the ineffectiveness of treatment; thus poor survival rate, [3][4][5] which necessitates assessing and controlling its risk factors and enhancing the prevention measures. Even though genetics affect the risk of Medicine high carbohydrate consumption was found to have a protective effect against the risk of PC in a cohort study in Finland, [3] nevertheless, the elevated consumption of fructose and sucrose (except soda drinks) was significantly and positively associated with the risk. ...
... [1] Generally, diets rich in fresh fruits, vegetables, nuts, and whole grains might help to reduce the risk of PC; on the other hand, an increased intake of saturated fat, fried foods, and animal products may induce its occurrence. [3,5] PC risk was found to be lower among individuals who highly adhered to the Mediterranean diet, while the opposite association was detected for the Western diet. [9] Given the complexity and the interaction of different dietary components and the narrow picture provided by studying a single dietary component, studying the overall dietary pattern offers an inclusive view of the relationship between a diet and disease risk. ...
... Several potential mechanisms have been suggested to elucidate the potential power to prevent cancer in foods containing those nutrients/ chemicals, mainly fresh fruits and cruciferous vegetables. [25,26] On the other hand, previous studies documented that meat and saturated fat intakes were associated with an increased risk of PC. [3,5,17,24] Here, we detected a nonsignificant positive association. Evidence concerning the positive association between PC and high intake of meat, dairy products, fat, and cholesterol is not confirmative. ...
Article
Full-text available
Diet is an important modifiable lifestyle factor, but epidemiological studies evaluating the association between dietary patterns and pancreatic cancer (PC) have reported inconsistent findings. This study aimed to evaluate the impact of several dietary choices on the risk of PC among newly diagnosed Jordanian patients. A case-control study was conducted at major teaching and general hospitals, including a Jordanian oncology center. The study included 101 patients with incident pancreatic cancer and 314 controls. Data was collected using interview-based questionnaires. Dietary intake was estimated using a validated Arabic and reproducible food-frequency questionnaire. Dietary patterns were derived using Principal Component Analysis. Multinomial logistic regression was used to estimate the association between dietary patterns and PC. Four dietary patterns were identified. The “Traditional” dietary pattern, which presented a diet rich in fresh fruits, vegetables, milk, yogurt, and lentils, was associated with a significant decrease in the odds of PC (OR = 0.42, 95% CI = 0.21–0.84) for the third quartile compared to first one. The “High-fruit” dietary pattern, which was loaded with strawberry, melon, watermelon, and other fruits, significantly reduced the odds of PC (OR = 0.38, 95% CI = 0.19–0.75) for the second quartile compared to the first one. The “Soup” dietary pattern was mainly composed of vermicelli soup, vegetable soup, lentil soup, and mushroom soup, which decreased the odds of PC (OR = 0.18, 95% CI = 0.07–0.38). There was no relation between PC and the “Western” dietary pattern, loaded with beer, wine, roasted lamb, meat, chicken sandwich, beefsteak, and fried fish. The “Traditional,” “High-fruit,” and “Soup” dietary patterns were associated with reduced risk of PC among Jordanians.
... Since obesity was identified as an important risk factor for many cancers, numerous studies were conducted to evaluate how effective the therapies for cancer prevention (surgical or pharmacological) against obesity proved to be [49,50]. Weight reduction (which correlates with a low-fat and -sugar diet and a high dietary intake of fresh fruit and vegetables, and their associated nutrients like fiber, antioxidants, and polyphenols) together with a healthy lifestyle and regular exercise may contribute to the prevention of pancreatic cancer [50,51]. ...
... Since obesity was identified as an important risk factor for many cancers, numerous studies were conducted to evaluate how effective the therapies for cancer prevention (surgical or pharmacological) against obesity proved to be [49,50]. Weight reduction (which correlates with a low-fat and -sugar diet and a high dietary intake of fresh fruit and vegetables, and their associated nutrients like fiber, antioxidants, and polyphenols) together with a healthy lifestyle and regular exercise may contribute to the prevention of pancreatic cancer [50,51]. Some studies suggested that certain anti-inflammatory drugs (such as aspirin), anti-diabetic drugs (such as metformin), and anti-dyslipidemic drugs (such as statins) are considered as potential chemo-preventive agents for pancreatic cancer development, although the results from previous studies were inconsistent [52][53][54]. ...
Article
Full-text available
Simple Summary The burden of pancreatic cancer attributable to a high body mass index (BMI) increased significantly for both sexes, in all ages, across all socio-demographic index (SDI) quintiles, and all GBD regions. There were apparent international variations in ASRs both for mortality and DALYs for pancreatic cancer that could be attributed to a high BMI in 2019: higher ASRs were observed in females, in the high SDI region than in other SDI regions, and in the Central Europe region than in other GBD regions. Growth trends in pancreatic cancer burden that could be attributed to a high BMI were faster in the low than in the high SDI region. Abstract (1) Background: This study aimed to assess the global burden of pancreatic cancer attributable to a high BMI in 1990–2019. (2) Methods: An ecological study was carried out. Data about deaths and Disability-Adjusted Life Years (DALYs) for pancreatic cancer were extracted from the Global Burden of Disease (GBD) study. The age-standardized rates (ASRs, per 100,000) were presented. In order to determine trends of pancreatic cancer burden, joinpoint regression analysis was used to calculate the average annual percent change (AAPC). (3) Results: The highest ASRs of DALYs of pancreatic cancer were found in the United Arab Emirates (47.5 per 100,000), followed by countries with about 25.0 per 100,000 (such as Hungary, Czechia, and Montenegro). From 1990 to 2019, the ASRs of deaths and DALYs of pancreatic cancer attributable to a high BMI significantly increased (p < 0.001) for both sexes in all ages, and across all SDI quintiles and all GBD regions. The highest fraction of DALYs attributable to a high BMI was found in the United States of America and China (equally about 15.0%), followed by the Russian Federation, India, Germany, and Brazil (about 5.0%, equally). (4) Conclusions: Further analytical epidemiological studies are necessary to elucidate the relationship between pancreatic cancer and a high BMI.
... While associations between diet and PDAC have been suggested, randomized trials are needed to better evaluate this relationship due to confounding factors such as smoking and obesity [102]. High dietary intake of fruits, vegetables, and whole grains has been shown to reduce pancreatic cancer risk and a possible protective role of dietary folate consumption has also been suggested [102,103]. Red meat and high-fat diets have been proposed as risk factors for PDAC [102]. ...
Article
Full-text available
Pancreatic ductal adenocarcinoma (PDAC) is often diagnosed at an advanced stage, resulting in poor prognosis and low 5-year survival rates. While early evidence suggests increased long-term survival in those with screen-detected resectable cancers, surveillance imaging is currently only recommended for individuals with a lifetime risk of PDAC ≥ 5%. Identification of risk factors for PDAC provides opportunities for early detection, risk reducing interventions, and targeted therapies, thus potentially improving patient outcomes. Here, we summarize modifiable and non-modifiable risk factors for PDAC. We review hereditary cancer syndromes associated with risk for PDAC and their implications for patients and their relatives. In addition, other biologically relevant pathways and environmental and lifestyle risk factors are discussed. Future work may focus on elucidating additional genetic, environmental, and lifestyle risk factors that may modify PDAC risk to continue to identify individuals at increased risk for PDAC who may benefit from surveillance and risk reducing interventions.
... A previous study reported that those free of prior cancer at baseline with a body mass index (BMI) ≥ 30 kg/m 2 have about 70% higher risk of pancreatic cancer compared to those with a BMI < 23 kg/m 2 [9], indicating that the regulation of muscle mass or obesity is directly associated with carcinogenesis in the pancreas [10]. An epidemiological study also reported that physical activity considerably decreases pancreatic cancer incidence by 28% [11], indicating that exercise has anti-cancer effects [12]. ...
Article
Full-text available
Simple Summary This study aimed to investigate the impact of several variables affecting the development of pancreatic cancer through the analysis of a sample cohort of 1,108,369 individuals from the National Health Insurance Sharing Service (NHISS DB). Furthermore, we aimed to develop individualized evidence-based prognostic and preventive programs (e.g., tailored exercise-based programs) through the derivation of cut-off value results of significant parameters to improve pancreatic cancer. There were differences between the pancreatic cancer versus non-pancreatic cancer groups in terms of gender; for example, body mass index, systolic/diastolic blood pressure, fasting blood glucose, and total cholesterol concentrations were lower in women with pancreatic cancer than in women without pancreatic cancer (p < 0.01). Fasting blood glucose and total cholesterol concentrations were significantly different between men with versus without pancreatic cancer (p < 0.05). Logistic regression analysis identified more than 20 parameters, including the impact of exercise. Afterward, the ROC curve analysis revealed significant cut-off values, which differed between men and women (i.e., the ROC curve analysis showed that total cholesterol concentration was the only significant factor associated with pancreatic cancer in men). Our analysis of large-scale data from the NHISS DB suggests that identifying significant parameters helps in developing individualized evidence-based prognostic and preventive programs (e.g., tailored exercise-based programs) for ameliorating pancreatic cancer. Abstract This study aimed to examine the effects of multiple parameters on the incidence of pancreatic cancer. We analyzed data from 1,108,369 individuals in the National Health Insurance Sharing Service Database (NHISS DB; birth to death; 2002 to 2015) and identified 2912 patients with pancreatic cancer. Body mass index, systolic/diastolic blood pressure, and fasting blood glucose and total cholesterol concentrations were lower in women with than without pancreatic cancer (p < 0.01). Fasting blood glucose and total cholesterol concentrations were significantly different between men with and without pancreatic cancer (p < 0.05). In the logistic regression analysis, the total cholesterol concentration (odds ratio (OR), 1.007; 95% confidence interval (CI), 1.005–1.010) was significantly higher in men than women with pancreatic cancer (p < 0.05). Pancreatic cancer rates were highest in men who smoked for 5–9 years or more (OR, 5.332) and in women who smoked for 10–19 years (OR, 18.330). Daily intensive exercise reduced the risk of pancreatic cancer by 56% in men (95% CI, 0.230–0.896). Receiver operating characteristic curve analysis revealed a total cholesterol concentration cut-off point of 188.50 mg/dL (p < 0.05) in men with pancreatic cancer, with a sensitivity and specificity of 53.5% and 54.6%, respectively. For women, the cut-off values for weight and gamma glutamyl transpeptidase concentration were 58.5 kg and 20.50 U/L, respectively. The sex-specific differences in patients with pancreatic cancer identified herein will aid in the development of individualized evidence-based prognostic and preventive programs for the treatment of pancreatic cancer.
... The mechanisms of progression (cellular and molecular) of known PC types, complications and screening methods used to identify precise, effective and cost-effective novel therapeutic drug development for treatment are all still somewhat unknown [10][11][12]. Accordingly, numerous proposed factors such as environmental, genetic and other pathology have been linked to PC disease, thereby translating to the challenges of a novel therapy [7,13,14]. ...
Article
Full-text available
The unexpected rise in cancer and diabetes statistics has been a significant global threat, inciting ongoing research into various biomarkers that can act as innovative therapeutic targets for their management. The recent discovery of how EZH2-PPARs’ regulatory function affects the metabolic and signalling pathways contributing to this disease has posed a significant breakthrough, with the synergistic combination of inhibitors like GSK-126 and bezafibrate for treating these diseases. Nonetheless, no findings on other protein biomarkers involved in the associated side effects have been reported. As a result of this virtual study, we identified the gene–disease association, protein interaction networks between EZH2-PPARs and other protein biomarkers regulating pancreatic cancer and diabetes pathology, ADME/Toxicity profiling, docking simulation and density functional theory of some natural products. The results indicated a correlation between obesity and hypertensive disease for the investigated biomarkers. At the same time, the predicted protein network validates the link to cancer and diabetes, and nine natural products were screened to have versatile binding capacity against the targets. Among all natural products, phytocassane A outperforms the standard drugs’ (GSK-126 and bezafibrate) in silico validation for drug-likeness profiles. Hence, these natural products were conclusively proposed for additional experimental screening to complement the results on their utility in drug development for diabetes and cancer therapy against the EZH2-PPARs’ new target.
... Among the non-modifiable factors, gender, age and genetic factors have been widely studied. Alcohol and smoking, comorbidities (such as obesity, diabetes, and chronic pancreatitis) and other lifestyle factors, such as diet, have also been studied in association with PC [3][4][5]. For instance, one-third of the deaths from all types of cancer are due to lifestyle and diet [6]. ...
Article
Full-text available
Pancreatic cancer (PC) represents the 6th cause of cancer death. Although the aetiology of PC is not completely understood, numerous risk factors have been identified in association with this cancer, among them diet. However, little is known about the association between the Mediterranean Diet (MedDiet) and the risk of PC. For this reason, we conducted a systematic review with meta-analysis according to the PRISMA guidelines, searching on three databases (PubMed/MEDLINE, Scopus, and EMBASE). The protocol was registered in PROSPERO. Both fixed and random effect models were performed. The Effect size was reported as a hazard ratio (HR) with a 95% Confidence Interval (CI). A total of eight articles were included. The methodological quality of the included meta-analyses was high. Our results show that a higher adherence to the MedDiet is associated with a lower risk of PC [HR:0.82 (0.76–0.88) p < 0.001, based on 1,301,320 subjects]. The results were also confirmed in sensitivity and subgroups analyses (avoidance of potential overlapping effects, type of tools used to assess dietary intake and the diagnosis of PC, prevalence and incidence of PC risk, country where the studies took place, sex, and cancer site). Promoting a higher adherence to the MedDiet could be an effective approach to reduce the risk of PC.
... 121 ITC and some isothiocyanates, compounds found in cruciferous vegetables, such as sulforaphane, benzyl isothiocyanate, and phenethyl isothiocyanate, have been shown in animal studies to have a suppressive effect on cancer cells of the pancreas. 125 Despite that many case-control studies have found an inverse association between the consumption of fruits and vegetables and the risk of PC, A meta-analysis of prospective studies found that consuming large amounts of fruits and vegetables citrus fruit or cruciferous vegetables was not associated with a lower risk of PC. 10 It was also stated that, while increased consumption of fruits is associated with a lower PC risk in men but not in women, the studies included are limited. 10 Furthermore, dose-response analyses revealed no significant dose-response relationships between an increase in fruit and vegetable consumption of 100 g/d and PC risk. ...
Article
Full-text available
This narrative review summarizes the main findings of observational studies (case-control and cohort) as well as systematic reviews and meta-analyses on the role of nutrients and dietary patterns on pancreatic cancer (PC) risk and elucidates possible mechanisms for the association between nutrients or specific food components and the risk of PC. A literature search of MEDLINE (PubMed), Google Scholar, ScienceDirect, and Scopus was performed. An extensive search of related articles published in the English language from 1985 to 2022 was carried out. Our search included macro- and micronutrient intake as well as dietary patterns associated with PC. In conclusion, the consumption of a diet high in nutrients such as sugar, fats, and red and processed meats can increase the risk of PC. Conversely, a high dietary intake of fresh fruit and vegetables and their associated nutrients like fiber, antioxidants, and polyphenols may prevent PC. Dietary patterns loaded with red and processed meats were also linked to an increased risk of PC, whereas dietary patterns rich in plant-based foods like vegetables, fruits, whole grains, and legumes were associated with a reduced risk of PC. Dietary fiber, fat-soluble vitamins, water-soluble vitamins, and minerals might also play a protective role against PC.
Article
Full-text available
Cancer is a crucial global health problem, and prevention is an important strategy to reduce the burden of the disease. Daily diet is the key modifiable risk factor for cancer, and an increasing body of evidence suggests that specific nutrients in foods may have a preventive effect against cancer. This review summarizes the current evidence on the role of nutrients from foods in cancer intervention. It discusses the potential mechanisms of action of various dietary components, including phytochemicals, vitamins, minerals, and fiber. The findings of epidemiological and clinical studies on their association with cancer risk are highlighted. The foods are rich in bioactive compounds such as carotenoids, flavonoids, and ω-3 fatty acids, which have been proven to have anticancer properties. The effects of steady-state delivery and chemical modification of these food’s bioactive components on anticancer and intervention are summarized. Future research should focus on identifying the specific bioactive compounds in foods responsible for their intervention effects and exploring the potential synergistic effects of combining different nutrients in foods. Dietary interventions that incorporate multiple nutrients and whole foods may hold promise for reducing the risk of cancer and improving overall health.
Article
Objective: Most previous studies investigated the associations between intake of individual nutrients and risk of disease, which failed to consider the potential interactions and correlations between various nutrients contained in food. Although dietary quality scores provide a comprehensive evaluation of the entire diet, it remains elusive whether they are associated with the risk of pancreatic cancer. Methods: Dietary intake data collected with the Dietary Questionnaire (DQX) and Diet History Questionnaire (DHQ) were used to calculate HEI-2015 and DQI-R scores for participants in the Prostate, Lung, Colorectal and Ovarian (PLCO) Cancer Screening Trial. A high score indicates an increased intake of adequacy components and a decreased intake of moderation components. This study included 252 cases of pancreatic cancer documented from 58,477 persons during a median follow-up of 12.2 years in the DQX cohort and 372 cases of pancreatic cancer ascertained from 101,721 persons during a median follow-up of 8.9 years in the DHQ cohort. Cox proportional hazards regression analysis was performed to calculate hazard ratios (HR) and 95% confidence intervals (CI) for the associations between the two dietary quality scores and pancreatic cancer risk. Results: After adjustment for confounders, HEI-2015 and DQI-R scores were not significantly associated with pancreatic cancer risk. However, a significantly lower risk was observed for overweight persons with a higher HEI-2015 score in the DQX cohort (HR [95% CI] comparing the highest with lowest tertile: 0.52 [0.32, 0.85], p for trend = 0.009) and those with higher scores of some individual components. Conclusion: Collectively, overall dietary quality is not associated with an altered risk of pancreatic cancer in this US population.
Chapter
Our understanding of the molecular bases of carcinogenesis has come a long way in the past 20 years, with consensus that nearly all cancers result from the accumulation of genetic abnormalities, largely influenced by environmental and lifestyle factors. As such, most cancer-causing genetic mutations are not inherited but rather emerge over the course of a person’s lifetime because of environmental exposures that are not yet fully understood. The field known as “environmental oncology” investigates the complex interplay between environmental factors and the vulnerability of living organisms to the transformative effects of carcinogenic substances. In fact, up to 60% of all cancers can be linked with environmental factors. This chapter discusses salient aspects of gene-environment-lifestyle interactions that define cancer outcomes including factors such as diet, tobacco use, alcohol, obesity, and physical activity, ultraviolet light, radon gas, pathogenic organisms, radiation, and pollution. The study of environmental oncology can help identify and reduce exposures to biological, chemical, physical agents linked to cancer outcomes with the goal of ultimately lowering the cancer burden by focusing “upstream” on controllable conditions and strategies for prevention rather than traditional “downstream” consequences.KeywordsCancer etiologyCancer typesCancer risk factorsCarcinogensGenetic causes of cancerEnvironmental causes of cancerLifestyle causes of cancer
Article
Full-text available
Significance Pancreatic ductal adenocarcinoma (PDAC) is projected to become the second deadliest cancer by 2030. Advances in therapeutic treatments are urgently required to fight against this fatal disease. Here, elucidation of the metabolic signature of PDAC has identified the low-density lipoprotein receptor (LDLR), which facilitates cholesterol uptake, as a promising therapeutic target. Blocking of LDLR reduces the proliferative and clonogenic potential of PDAC cells and decreases activation of the ERK1/2 survival pathway. Moreover, LDLR silencing sensitizes PDAC cells to chemotherapeutic drugs and potentiates the tumoral regression promoted by chemotherapy. Finally, Ldlr is highly expressed at all stages of human PDAC and expression is associated with an increased risk of PDAC recurrence.
Article
Full-text available
Autophagy, a lysosomal degradation pathway for cellular constituents and organelles, is an adaptive and essential process required for cellular homeostasis. Although autophagy functions as a survival mechanism in response to cellular stressors such as nutrient or growth factor deprivation, it can also lead to a non-apoptotic form of programmed cell death (PCD) called autophagy-induced cell death or autophagy-associated cell death (type II PCD). Current evidence suggests that cell death through autophagy can be induced as an alternative to apoptosis (type I PCD), with therapeutic purpose in cancer cells that are resistant to apoptosis. Thus, modulating autophagy is of great interest in cancer research and therapy. Natural polyphenolic compounds that are present in our diet, such as rottlerin, genistein, quercetin, curcumin, and resveratrol, can trigger type II PCD via various mechanisms through the canonical (Beclin-1 dependent) and non-canonical (Beclin-1 independent) routes of autophagy. The capacity of these compounds to provide a means of cancer cell death that enhances the effects of standard therapies should be taken into consideration for designing novel therapeutic strategies. This review focuses on the autophagy- and cell death-inducing effects of these polyphenolic compounds in cancer.
Article
Evidence of a protective effect of several antioxidants and other nutrients on pancreatic cancer risk is inconsistent. The aim of this study was to investigate the association for prediagnostic plasma levels of carotenoids, vitamin C, retinol and tocopherols with risk of pancreatic cancer in a case–control study nested within the European Prospective Investigation into Cancer and Nutrition (EPIC). 446 incident exocrine pancreatic cancer cases were matched to 446 controls by age at blood collection, study center, sex, date and time of blood collection, fasting status and hormone use. Plasma carotenoids (a- and ß-carotene, lycopene, ß-cryptoxanthin, canthaxanthin, zeaxanthin and lutein), a- and ¿-tocopherol and retinol were measured by reverse phase high-performance liquid chromatography and plasma vitamin C by a colorimetric assay. Incidence rate ratios (IRRs) with 95% confidence intervals (95%CIs) for pancreatic cancer risk were estimated using a conditional logistic regression analysis, adjusted for smoking status, smoking duration and intensity, waist circumference, cotinine levels and diabetes status. Inverse associations with pancreatic cancer risk were found for plasma ß-carotene (IRR highest vs. lowest quartile 0.52, 95%CI 0.31–0.88, p for trend¿=¿0.02), zeaxanthin (IRR highest vs. lowest quartile 0.53, 95%CI 0.30–0.94, p for trend¿=¿0.06) and a-tocopherol (IRR highest vs. lowest quartile 0.62, 95%CI 0.39–0.99, p for trend¿=¿0.08. For a- and ß-carotene, lutein, sum of carotenoids and ¿-tocopherol, heterogeneity between geographical regions was observed. In conclusion, our results show that higher plasma concentrations of ß-carotene, zeaxanthin and a-tocopherol may be inversely associated with risk of pancreatic cancer, but further studies are warranted.
Conference Paper
Epidemiological studies have consistently shown that regular consumption of fruits and vegetables is strongly associated with reduced risk of developing chronic diseases, such as cancer and cardiovascular disease. It is now widely believed that the actions of the antioxidant nutrients alone do not explain the observed health benefits of diets rich in fruits and vegetables, because taken alone, the individual antioxidants studied in clinical trials do not appear to have consistent preventive effects. Work performed by our group and others has shown that fruits and vegetable phytochemical extracts exhibit strong antioxidant and anti proliferative activities and that the major part of total antioxidant activity is from the combination of phytochemicals. We proposed that the additive and synergistic effects of phytochemicals in fruits and vegetables are responsible for these potent antioxidant and anticancer activities and that the benefit of a diet rich in fruits and vegetables is attributed to the complex mixture of phytochemicals present in whole foods. This explains why no single antioxidant can replace the combination of natural phytochemicals in fruits and vegetables to achieve the health benefits. The evidence suggests that antioxidants or bioactive compounds are best acquired through whole-food consumption, not from expensive dietary supplements. We believe that a recommendation that consumers eat 5 to 10 servings of a wide variety of fruits and vegetables daily is an appropriate strategy for significantly reducing the risk of chronic diseases and to meet their nutrient requirements for optimum health.
Article
Pancreatic cancer is currently the fourth leading cause of cancer-related death, and it is projected that within the next two decades it will become the second most common cause of death due to cancer. Few patients are diagnosed when surgical resection is feasible and the efficacy of existing chemotherapeutic agents for advanced/metastatic cancer is limited. Thus, there is a need to identify agents that can prevent pancreatic cancer or improve survival in those affected. Vitamin D and its analogues, with their ability to regulate cell growth, differentiation, apoptosis and angiogenesis, may be promising agents. This review explores the published literature about the potential role of vitamin D and its analogues in preventing or treating pancreatic cancer. The vitamin D system is altered in pancreatic cancer. Pancreatic cancer tissue expresses vitamin D receptors, but the calcitriol analogues may affect pancreatic cancer tissue by mechanisms that do not involve interaction with its receptors. Experimental evidence postulates multiple potential mechanisms by which calcitriol analogues may exert their anti-cancer effect, the most common being by action on cyclin-dependent kinases p21 and p27. Use of calcitriol analogues in pancreatic cancer remains largely underexplored and warrants further clinical trials. Copyright © 2015. Published by Elsevier Ireland Ltd.
Article
Obesity and intrapancreatic fatty infiltration are associated with increased risk of pancreatic cancer and its precursor lesions. The interplay amongst obesity, inflammation, and oncogenic Kras signaling promotes pancreatic tumorigenesis. Targeting the interaction between obesity-associated inflammation and Kras signaling may provide new strategies for prevention and therapy of pancreatic cancer. Copyright © 2015, American Association for Cancer Research.
Article
Pancreatic cancer is expected to be the second deadliest malignancy in the USA by 2020. The survival rates for patients with other gastrointestinal malignancies have increased consistently during the past 30 years; unfortunately, however, the outcomes of patients with pancreatic cancer have not changed significantly. Although surgery remains the only curative treatment for pancreatic cancer, therapeutic strategies based on initial resection have not substantially improved the survival of patients with resectable disease over the past 25 years; presently, more than 80% of patients suffer disease relapse after resection. Preclinical evidence that pancreatic cancer is a systemic disease suggests a possible benefit for early administration of systemic therapy in these patients. In locally advanced disease, the role of chemoradiotherapy is increasingly being questioned, particularly considering the results of the LAP-07 trial. Novel biomarkers are clearly needed to identify subsets of patients likely to benefit from chemoradiotherapy. In the metastatic setting, FOLFIRINOX (folinic acid, 5-fluorouracil, irinotecan, and oxaliplatin), and nab-paclitaxel plus gemcitabine have yielded only modest improvements in survival. Thus, new treatments are urgently needed for patients with pancreatic cancer. Herein, we review the state-of-the-art of pancreatic cancer treatment, and the upcoming novel therapeutics that hold promise in this disease are also discussed.
Article
Purpose: The roles of intravisceral and subcutaneous fat are unknown and the prevalence of precancerous lesions in obese patients was never evaluated. Aims- To assess the frequency and severity of pancreatic intraepithelial neoplasia (PanIN), to correlate pathological findings with metabolic abnormalities, type of fat and fatty pancreatic infiltration. Experimental Design: Normal pancreatic tissue from surgical specimens was analyzed. Fatty infiltration and fibrosis in intra- and extralobular locations, PanIN were assessed. General characteristics were collected: body mass index (BMI), diabetes and tobacco intake. Liver steatosis, subcutaneous and intravisceral fat were assessed by CT scan (ImageJ software). Results- 110 patients were included (median age: 53.8 [17-85] years). Arterial hypertension, diabetes, tobacco intake were found in 19, 9 and 23%, respectively. Median BMI was 24 [16-37], (BMI<25: 45%, 25-<30: 24%, ≥30: 11%). Overall, PanIN lesions were found in 65% (Type 1, 2 and 3 PanIN in 62, 38 and 1%, respectively). Fibrosis and fatty pancreas (intra- and extralobular locations) were found in 1% and 24% and in 30% and 51%, respectively. A correlation was observed between PanIN lesions and fatty pancreas [extra- (0.01) and intralobular (<0.0001)], intralobular fibrosis (0.003), high BMI (p=0.02), subcutaneous (p=0.02) and intravisceral fat (p=0.02). The number of PanIN lesions was correlated with the intravisceral fat (r=0,22, p=0,04) but not with the subcutaneous fat (r=0.14, p=0.22). In multivariate analysis, PanIN were associated with intralobular fibrosis (OR=5.61 [1.18-42.99]) and the intralobular fat (OR=17.86 [4.935-88.12]). Conclusions: Obesity (android obesity, especially) and pancreatic fatty infiltration are risk factors for pancreatic precancerous lesions. Copyright © 2015, American Association for Cancer Research.
Article
Pancreatic cancer is one of the deadliest cancers, with exceptionally high mortality. Despite the relatively low incidence rate (10th), it is the fourth leading cause of cancer-related deaths in most developed countries. To improve the early diagnosis of pancreatic cancer and strengthen the standardized comprehensive treatment are still the main focus of pancreatic cancer research. Here, we summarized the rapid developments in the diagnosis and treatments of pancreatic cancer. Regarding diagnosis, we reviewed advances in medical imaging technology, tumor markers, molecular biology (e.g., gene mutation), and proteomics. Moreover, great progress has also been made in the treatments of this disease, including surgical resection, chemotherapy, targeted radiotherapy, targeted minimally invasive treatment, and molecular targeted therapy. Therefore, we also recapitulated the development, advantages, and disadvantages of each of the treatment methods in this review.
Article
Pancreatic cancer is one of the most aggressive malignancies in US adults. Experimental studies have found that antioxidant nutrients could reduce oxidative DNA damage, suggesting that these antioxidants may protect against pancreatic carcinogenesis. Several epidemiologic studies showed that dietary intake of antioxidants was inversely associated with the risk for pancreatic cancer, demonstrating the inhibitory effects of antioxidants on pancreatic carcinogenesis. Moreover, nutraceuticals, the anticancer agents from diet or natural plants, have been found to inhibit the development and progression of pancreatic cancer through the regulation of cellular signaling pathways. Importantly, nutraceuticals also up-regulate the expression of tumor-suppressive microRNAs (miRNAs) and down-regulate the expression of oncogenic miRNAs, leading to the inhibition of pancreatic cancer cell growth and pancreatic cancer stem cell self-renewal through modulation of cellular signaling network. Furthermore, nutraceuticals also regulate epigenetically deregulated DNAs and miRNAs, leading to the normalization of altered cellular signaling in pancreatic cancer cells. Therefore, nutraceuticals could have much broader use in the prevention and/or treatment of pancreatic cancer in combination with conventional chemotherapeutics. However, more in vitro mechanistic experiments, in vivo animal studies, and clinical trials are needed to realize the true value of nutraceuticals in the prevention and/or treatment of pancreatic cancer.