Vous êtes sur la page 1sur 13

N U TR IT ION RE S EA R CH 5 2 ( 20 1 8 ) 1 –1 3

Available online at www.sciencedirect.com

ScienceDirect
www.nrjournal.com

Review Article

Pancreatic cancer: A critical review of dietary risk

Asmaa A. Salem, Gerardo G. Mackenzie⁎


Department of Nutrition, University of California, Davis, One Shields Ave, Davis, CA, 95616

ARTI CLE I NFO A BS TRACT

Article history: Pancreatic cancer is a deadly disease. It is estimated that about 90% of pancreatic cancer
Received 5 August 2017 cases are due to environmental risk factors. Among these, approximately 50% of pancreatic
Revised 31 October 2017 cancer cases may be attributed to diet, which is largely modifiable. Given this large
Accepted 8 December 2017 attribution to diet, there have been numerous epidemiological studies assessing the risk of
various dietary factors on the incidence of pancreatic cancer. However, many of these
Keywords: studies present conflicting and/or inconclusive findings. The objective of this review is two-
Diet fold: (a) to summarize the current evidence on the association between various dietary
Nutrients factors and the risk of developing pancreatic cancer and (b) to discuss what additional
Foods studies are needed to better elucidate the role of diet as a potential risk factor for pancreatic
Pancreatic cancer cancer. We summarized the evidence by using data primarily from meta-analyses and pooled
Pancreatic cancer risk analysis when available, focusing on the most studied nutrients, foods, and dietary patterns.
We observed that, while the association between individual nutrients and pancreatic cancer
risk have been heavily studied, the evidence is mostly conflicting and inconclusive. In
contrast, the evidence of certain associations among dietary patterns and pancreatic cancer
risk is clearer, has more power, and is less conflicting. Therefore, we propose a shift in the
focus of nutritional epidemiological research with regards to pancreatic cancer risk. We
discourage further epidemiological research studies that focus on single nutrients, whereas
we strongly encourage additional studies that investigate how a combination of diet and
other lifestyle factors may promote or prevent pancreatic carcinogenesis.
© 2017 Elsevier Inc. All rights reserved.

Article Outline

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Approach . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3. Findings. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3.1. Nutrients and pancreatic cancer risk . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3.1.1. Vitamin C . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3.1.2. Folate (vitamin B9) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3.1.3. Vitamin E . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.1.4. Retinoids and carotenoids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4

Abbreviations: HCAs, Heterocyclic amines; AGE, Advanced glycation end products; NOCs, N-nitroso compounds; MD, Mediterranean diet.
⁎ Corresponding author at: Department of Nutrition, University of California, Davis, One Shields Ave., Davis, CA, 95616. Tel.: +1 530 752
2140; fax: +1 530 752 8966.
E-mail address: ggmackenzie@ucdavis.edu (G.G. Mackenzie).

https://doi.org/10.1016/j.nutres.2017.12.001
0271-5317/© 2017 Elsevier Inc. All rights reserved.
2 N U TR IT ION RE S EA R CH 52 ( 20 1 8 ) 1 –1 3

3.1.5. Vitamin D . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3.1.6. Selenium . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3.1.7. Lipids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
3.1.8. Discussion: nutrients and pancreatic cancer risk . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.2. Foods and pancreatic cancer risk . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.2.1. Red and processed meat. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.2.2. Dairy products . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.2.3. Fruits and vegetables . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.2.4. Whole grains . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.2.5. High glycemic foods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.2.6. Alcoholic beverages . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.2.7. Discussion: foods and pancreatic cancer risk . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.3. Dietary patterns and pancreatic cancer risk . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.3.1. Western-style dietary pattern. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.3.2. Prudent or healthy dietary pattern . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.3.3. Healthy Eating Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.3.4. Mediterranean Diet . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
3.3.5. Discussion: dietary patterns and pancreatic cancer risk . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5. Knowledge gaps and future research directions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
References. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10

1. Introduction to summarize the current evidence on the association


between various dietary factors and the risk of developing
pancreatic cancer and (b) to discuss what additional studies
Pancreatic cancer, currently the fourth leading cause of
are needed to better elucidate the role of diet as a potential
cancer death worldwide [1], is projected to become the 2nd
risk factor for pancreatic cancer. Moreover, we point out some
by 2030 [2]. With a 5-year survival rate of approximately 8% [1]
of the challenges and limitations of the research and
and an average survival time of 6 months for those with
emphasize the need for future research to include well-
advanced metastasized disease [3], pancreatic cancer is one of
designed and carefully carried out clinical trials on dietary
the most fatal malignancies with an extremely dismal
patterns to better guide us to elucidate how diet may
prognosis. This exceedingly short survival time, however, is
influence the incidence of pancreatic cancer.
counterbalanced with what research shows to be a very slow
evolution lasting up to 18 years from the time of initiating
mutations in pre-cancerous lesions to metastatic disease [4].
2. Approach
This dichotomy between a slow progression and rapid fatality
strongly suggests that prevention should be a key emphasis in
Using PubMed, we searched for meta-analyses or pooled
reducing pancreatic cancer burden.
analyses of epidemiological studies assessing the association
Like many cancers, pancreatic cancer is considered to have a
between nutrients, diets, or dietary patterns and pancreatic
large environmental attributable risk [5]. It is estimated that
cancer between 2010 and August 2017. When a large cohort
only 5% to 10% of pancreatic cancer cases are hereditary, and
study conducted after a meta-analysis was found, this was
the remaining 90% to 95% are due to environmental risk factors,
referenced as well. Search parameters used were “nutrients”
which are largely modifiable [6]. Of those risk factors, smoking,
or “vitamin” or “vitamin C” or “vitamin D” or “vitamin E” or
alcohol, obesity, diet, and physical inactivity have been the
“selenium” or “folate” or “cholesterol” or “fat” or “vitamin A”
most studied, and numerous studies have shown them to be
or “diet” or “dietary pattern” or “red meat” or “fruits and
associated with increased risk. Of these, smoking and obesity
vegetables” or “whole grains” or “high glycemic” or “dairy” or
are the most well established to have a causal relationship, with
“Mediterranean diet” or “alcohol” and “pancreatic cancer.”
estimates that these factors are significant triggers in approx-
imately 33% and 16% percent of pancreatic cancer cases,
respectively [7]. However, diet may have the largest attribution
of cases; some have estimated that approximately 50% of 3. Findings
pancreatic cancer cases may be attributed to diet [8,9].
Given this large attribution to diet, there have been 3.1. Nutrients and pancreatic cancer risk
numerous epidemiological studies assessing the risk of
various dietary factors on the incidence of pancreatic cancer. Multiple micro and macro nutrients have been assessed
However, many of these studies showed conflicting and/or epidemiologically for their association with the incidence of
inconclusive findings. The objective of this review is 2-fold: (a) pancreatic cancer (Table 1). Among the vitamins, those with a
N U TR IT ION RE S EA R CH 5 2 ( 20 1 8 ) 1 –1 3 3

significant and robust amount of data are the water-soluble Given the limitations of assessing dietary intake and that
vitamins C and B9 and the fat-soluble vitamins D, E, and A serum measures of vitamin C may more accurately indicate
(along with the carotenoids). Among the minerals, selenium intake, a few studies have investigated this relationship.
has been researched more extensively. With respect to the Interestingly, one small nested case control study in the EPIC
macronutrients, fat, including select fatty acids (eg, polyun- prospective cohort which followed participants for 11 years
saturated fatty acids) and cholesterol, have garnered much found no association between pre-diagnostic levels of serum
research interest. In the sections below, we summarize the vitamin C (measured once at the beginning of the study) and
most relevant findings for each of these selected nutrients pancreatic cancer risk [18]. On the other hand, another large
and discuss the limitations of the evidence. cohort study showed a statistically significant inverse rela-
tionship between serum vitamin C and pancreatic cancer risk,
3.1.1. Vitamin C despite finding no association for dietary intake as measured
Vitamin C is known for its antioxidant, immune boosting through a 7-day food diary [19]. Although still premature,
properties [10]. Given that inflammation and oxidative stress these findings suggest the need to determine serum vitamin C
may play a role in pancreatic carcinogenesis [11,12], it is levels in future studies to better correlate the levels of this
biologically plausible that vitamin C may mitigate this vitamin to the incidence of pancreatic cancer.
process. In addition, vitamin C can induce apoptosis and
inhibit neoplastic lesions in the pancreas [13,14]. 3.1.2. Folate (vitamin B9)
With regards to human epidemiological data, three recent Folate has been extensively studied for its link to cancer, due
meta-analyses (2015-2016) have been conducted, with some to its function as a methyl donor and its role in the synthesis
but not complete overlap in the studies included [15-17]. of DNA nucleotides [20]. Thus, it is hypothesized that folate
Ranging from 15 to 20 studies, including both case-control may prevent DNA double strand breaks, whereas a folate
and cohort designs, the pooled results of these meta-analyses deficiency may lead to aberrant methylation patterns
consistently show a significantly reduced risk of pancreatic resulting in the altered expression of tumor-suppressor or
cancer for those with the highest dietary intake of vitamin C oncogenes [21]. In contrast, it has been hypothesized that in
compared to those with the lowest. The combined relative the case of pre-existing neoplastic lesions, folate may allow
risk in these meta-analyses ranges from 0.66 to 0.70. When for more frequent DNA replication and cell division, poten-
stratifying results by study design, however, this significant tially leading to further growth of cancerous lesions [20].
inverse association remains only for case-control studies, but The current epidemiological evidence on folate and its
does not persist in cohort studies. association with the incidence of pancreatic cancer has been

Table 1 – Associations between nutrients and pancreatic cancer risk


Nutrient Potential link to pancreatic cancer Association with pancreatic cancer risk Reference(s)

Vitamin C Boosts immune function Significant reduced risk (remained significant only [15-17]
Antioxidant in case control studies after stratification)
Prevents DNA damage [10] Mixed results for serum vitamin C
Folate Methyl donor Inverse association for dietary folate and total folate [22,23,26,27]
Prevent DNA double strand breaks [20] (remained significant only in case-control studies)
Deficiency may lead to aberrant No association for supplemental folate
methylation of tumor suppressor or Non-significant U-shaped association for serum folate
oncogenes [21].
Vitamin E Chain breaking antioxidant [28] Reduced risk (borderline significant association after [15,18,31]
Growth inhibitory effect of pancreatic stratification for cohort studies)
cancer cells in vitro [29,30] Non-significant inverse association for plasma vitamin E
Vitamin A Vitamin A may mechanically reprogram Inverse association for Vitamin A and carotenoids in [15,18,39,40]
(Retinoids and pancreatic cancer stellate cells [36] general (no association after stratification for cohort studies)
Carotenoids) β-carotene has pro-vitamin A capacity. Possible inverse association for lycopene, β-carotene,
Other carotenoids have antioxidant and and β-cryptoxanthin after stratification by carotenoid type
anti-inflammatory properties [37]. Inverse association for the highest plasma levels of
β-carotene and zeaxanthin
Vitamin D Activates genes involved in cell No association for dietary intake or serum [43-47]
transformation: proliferation, levels of 25-OH D3
apoptosis, differentiation [41].
Modulates the immune system [42].
Selenium Component of antioxidant enzymes [52]. Significant inverse association [15,19,53]
(only significant in case control studies)
Lipids Chronic and excess dietary fat may No association for total fat, saturated fat, [60-65]
lead to pancreatic hypertrophy [55]. monounsaturated fat
Saturated fat associated with Possible inverse association for
insulin resistance [57]. polyunsaturated fat, particularly DHA
Higher Cholesterol increases Increased association for cholesterol
pro-inflammatory cytokines [59]. (only significant in case control studies)
4 N U TR IT ION RE S EA R CH 52 ( 20 1 8 ) 1 –1 3

largely inconsistent. A meta-analysis of 13 studies, including Thus, the data, so far, suggest that there may be a small
7 case-controls and 6 cohorts, found an inverse association inverse relationship between vitamin E and pancreatic cancer
between dietary folate and total folate for those in the highest risk. However, additional larger studies are needed to confirm
level of folate intake, but no association for supplemental whether this inverse relationship is significant and/or wheth-
folate [22]. A potential limitation of this meta-analysis, er it occurs only with individuals at the highest vitamin E
however, is the significant heterogeneity found between the levels.
studies. On the other hand, a pooled analysis of 14 cohort
studies found no association for dietary, supplemental, or 3.1.4. Retinoids and carotenoids
total folate intake [23]. Among the 14 cohort studies pooled in Vitamin A is essential for cell differentiation and growth, and
this analysis, only two found an association between folate therefore this represents a plausible link through which it
intake and pancreatic cancer risk. The ATB Cancer Prevention may modulate carcinogenesis [32]. In vitro studies have
Study conducted in smokers reported the lowest levels of shown that the retinoids can induce apoptosis in pancreatic
folate intake but found a reduced risk for those in the highest cancer cells [33], as well as inhibit cell adhesion [34] and
quartile [24], whereas the Cancer Prevention Study Nutrition migration [35]. In addition, vitamin A may also modulate the
Cohort II, as reported in this meta-analysis, found an pancreatic cancer microenvironment, by restoring mechani-
increased risk [25]. Additionally, The Singapore Chinese cal quiescence in pancreatic stellate cells, through a mecha-
Health Study, a prospective cohort not included in these nism involving the retinoic acid receptor beta (RAR-β) [36].
earlier analyses, found no association between dietary folate Besides the ability of some of the carotenoids to metabolize
intake and the risk of pancreatic cancer [26]. When analyzing to vitamin A, many of them have been ascribed antioxidant
plasma folate levels, a nested case-control study in the EPIC and anti-inflammatory functions. For example, lycopene has
cohort found a U-shaped relationship between plasma folate been found to inhibit pancreatic cancer cell proliferation
and pancreatic risk, with an increased, although non- [37], and this may be mediated by the suppression of NF-κB
significant, risk for those in the highest and lowest quintiles signaling [38].
of plasma folate status [27]. Based on these mixed findings, A meta-analysis including 11 studies showed an inverse
we cannot currently conclude with any degree of confidence association between the highest levels of dietary vitamin A
whether folate is associated with pancreatic cancer risk. intake and pancreatic cancer risk [39]. A limitation of this
meta-analysis was that the majority of the studies in this
3.1.3. Vitamin E analysis were case-control and this inverse association did
Vitamin E, a well-known chain-breaking antioxidant, pre- not remain significant in the one cohort study included [39]. A
vents the oxidation of plasma membrane lipids, thereby larger meta-analysis including 17 studies investigating vita-
maintaining the integrity of the cell membrane [28]. Vitamin min A, retinol, and the various carotenoids found a signifi-
E may prevent tumor growth by preventing DNA and cell cantly inverse association for the highest level of vitamin A
damage caused by the free radicals produced from oxidized intake and pancreatic cancer risk, but no association for
lipids [28]. Besides its role as an antioxidant, there is in vitro retinol intake [40]. The significant inverse association found
evidence indicating that vitamin E can suppress the growth of for vitamin A, however, was only observed in case-control
pancreatic cancer cells, [29] and this may be potentially studies after stratification by study design. Similarly, a
mediated by the increased expression of the p27 tumor significant inverse association was found for the carotenoids
suppressor gene [30]. in general, but this also did not persist in cohort studies after
Two recent meta-analyses with significant overlap in stratification [40].
the studies included found a significant inverse association After stratifying by carotenoid type, an inverse association
between the highest levels of dietary intake of vitamin E was only found for β-carotene (pooled OR, 0.78; 95% CI, 0.66-
and pancreatic cancer risk [15,31]. After stratifying by study 0.92; P = .003) and lycopene (pooled OR, 0.84; 95% CI, 0.73-0.97;
design, both meta-analyses found a significant inverse P = .020) [40]. A previously mentioned meta-analysis found an
relationship persist in case-control studies, but differed in inverse association between highest levels of dietary
their statistical outcome for cohort studies, even though β-carotene, but this did not remain significant after stratifi-
they included the same four cohort studies [15,31]. One cation in cohort studies [15]. Three studies in this
study concluded that the inverse relationship remained meta-analysis yielded a pooled inverse association between
significant for cohort studies: pooled RR 0.88 (0.79, 0.99) β-cryptoxanthin and the risk of pancreatic cancer, and a
[31]. The second meta-analysis concluded that the inverse borderline significant inverse association for lycopene. No
association was no longer significant in cohort studies: association was found for the other carotenoids. When
pooled OR, 0.85; 95% CI, 0.68-1.06 [15]. This slight difference measuring plasma levels of carotenoids and retinoids, the
appears to be dependent on the type of summary risk EPIC nested case-control study found an inverse association
calculated in each study. with pancreatic cancer risk for the highest plasma levels of
When assessing plasma α-tocopherol levels, the afore- β-carotene and zeaxanthin [18]. No association was found
mentioned nested case-control study in the EPIC cohort found for plasma levels of β- cryptoxanthin, lycopene, lutein, or α-
an inverse, but non-significant (P = .08), association for the carotene. In summary, the most consistent findings of the
highest levels of plasma vitamin E and pancreatic cancer [18]. mentioned studies point toward a potential inverse associ-
However, for every doubling of plasma α-tocopherol levels, a ation between β-carotene and pancreatic cancer risk, while
40% reduction in pancreatic cancer risk was found after no definitive conclusions can be made for the other
analyzing data by a log-transformation. retinoids/carotenoids.
N U TR IT ION RE S EA R CH 5 2 ( 20 1 8 ) 1 –1 3 5

3.1.5. Vitamin D lead to hypertrophy and hyperplasia in pancreatic tissue [55].


The mechanisms linking vitamin D and cancer are related to As dietary fat enters the duodenum, cholecystokinin is
its role as a regulator of genes involved in cell transformation: released stimulating the release of lipase enzymes from the
growth, differentiation, proliferation, and apoptosis [41]. For pancreas. An excess, however, may lead to hypertrophy or
example, Vitamin D regulates gate-keeper genes which prevent hyperplasia in the pancreas in order to keep up with the
damaged cells from proliferating and care-taker genes that demand for digestive enzymes [55]. This, in turn, may render
promote DNA repair. Vitamin D has been shown to interact pancreatic tissue more susceptible to carcinogens. Addition-
with cyclin D1 to inhibit cell proliferation, and has been ally, a high intake of fat may promote excess bile acid
implicated in the modulation of the immune system [42]. secretion, which has been shown to stimulate the release of
Given these mechanisms for Vitamin D and therefore the COX-2, an eicosanoid producing enzyme that has been found
biological plausibility associated with its potentially chemo- to be over-expressed in pancreatic tumors [56]. High intake of
preventive functions, it is surprising that the epidemiological saturated fat has also been associated with insulin resistance
evidence for its association with pancreatic cancer remains [57], which may lead to diabetes, a condition shown to
highly inconsistent and conflicting. A meta-analysis of 9 increase the risk of pancreatic cancer [58]. Evidence for a
studies with over 1 million participants found no association biological connection between cholesterol and pancreatic
between dietary intake of vitamin D or circulating levels of 25- carcinoma is more sparse, but some research indicates that
hydroxyvitamin D (25-OH D3) and the risk of pancreatic higher total cholesterol levels may be associated with
cancer [43]. This is consistent with the fact that some pooled increased pro-inflammatory cytokines [59], and inflamma-
analyses show an increased risk with higher dietary intake tion, as previously mentioned, plays a role in pancreatic
[44] or with highest levels of plasma 25-OH D3 [45], whereas carcinogenesis [11].
another pooled analyses of case-control studies assessing Despite some possible mechanisms that may confer
pre-diagnostic 25-OH D3 plasma levels [46] and two large biological plausibility to the association of fat intake and
prospective cohort studies calculating a predicted vitamin D pancreatic cancer, the epidemiological evidence fails to
status score [47] found a reduced risk of pancreatic cancer for support a strong link. One large meta-analysis of 19 studies
those with sufficient measured or predicted levels of vitamin D, reported no association between total fat intake and pancre-
respectively. These conflicting results may be attributed to the atic cancer risk, as well as no association for both fat from
different methods of assessing vitamin D status used in the animal sources or fat from vegetable sources [60]. Another
various studies, as well as geographic location or ethnicity. For large meta-analyses of 20 studies that looked at the associa-
instance, African Americans have a higher incidence of pancre- tion of various types of fatty acids with pancreatic cancer
atic cancer compared to Caucasian Americans [48-50]. incidence, found a non-significant positive association (RR =
Even though there is ongoing research on the potential 1.13 (95% CI, 0.94-1.35) for saturated fatty acids, no association
role of vitamin D and vitamin D receptor as a putative for monounsaturated fatty acids, and an inverse association
treatment for pancreatic cancer [51], epidemiological data on for polyunsaturated fatty acids (RR = 0.87 (95% CI, 0.75-1.00)
vitamin D status and pancreatic cancer risk report mixed [61]. These associations however only remain significant after
results, with no clear conclusion currently. stratification in case-control studies. Given the intriguing
potential inverse association between polyunsaturated fatty
3.1.6. Selenium acids and pancreatic cancer, many studies have investigated
It is currently still debated whether selenium can reduce the this relationship. Using fish intake as proxy for long-chain
risk of pancreatic cancer. Selenium is a mineral that functions polyunsaturated fatty acid (LC-PUFA) intake, a meta-analysis
as an antioxidant. It reduces oxidative stress and DNA of 19 studies found no association between increased LC-
damage, and is a component of some antioxidant enzymes PUFA intake and pancreatic cancer risk [62]. It is important to
such as glutathione peroxidase [52]. note that most of these studies were conducted in Western
Two meta-analyses, with significant overlap in the studies countries, where fish intake may be relatively limited. A large
included, revealed an inverse association for higher dietary prospective population-based cohort study conducted in
selenium intake and pancreatic cancer risk, but again, this Japan after this meta-analysis found a non-significant inverse
inverse association was only significant in case-control association for the highest intake of marine omega-3 polyun-
studies after stratification by study design [15,53]. Two of the saturated fatty acids. However, this inverse relationship was
large cohort studies included in these meta-analyses did significant (P = .03) specifically for docosahexaenoic acid
independently find significant inverse associations, for high (DHA) [63]. Thus, the data currently available shows no
versus low selenium intake in the VITAL cohort study [54] and convincing association between fat intake and pancreatic
for a summation of the three highest quartiles compared to cancer risk, with larger more robust trials needed to conclu-
the lowest in the EPIC-Norfolk cohort [19]. Thus, based on sively determine whether an inverse association between
these studies, it is still too early to make any assumptions on omega-3 fatty acids and pancreatic cancer risk exists.
whether selenium can reduce the risk of pancreatic cancer. With respect to cholesterol, one meta-analysis found a
significantly positive association between highest dietary
3.1.7. Lipids cholesterol intake and pancreatic cancer risk. However, this
Two lipid compounds, fatty acids and cholesterol, have relationship only remained significant in case-control studies
received much research attention for their potential impact after stratification [64]. Another meta-analysis, which includ-
on a variety of disease processes. With respect to pancreatic ed most but not all of the studies in the previously mentioned
cancer, chronic and excessive consumption of dietary fat may meta-analysis, also found a significant positive association
6 N U TR IT ION RE S EA R CH 52 ( 20 1 8 ) 1 –1 3

for dietary cholesterol intake, but again this relationship only only highly correlated, but they also appear to act synergis-
remained significant in case-control studies, and when tically, having additive and potentially compounding effects
stratifying for geographic location, was only significant for on health [71]. The milieu in which a nutrient exists within a
studies conducted in North America [65]. A dose–response food, known as the food matrix, may also impact the
analysis indicated that for every 100 mg/day increase in physiological effect of that nutrient, suggesting that the unit
dietary cholesterol, there was a significant 8% increase in risk of interest in nutritional research should be the food, and not
for pancreatic cancer [65]. a specific nutrient [72].
A related consideration that may affect the analysis of the
3.1.8. Discussion: nutrients and pancreatic cancer risk studies and contribute to the confusion in the literature is
This survey of the literature on the association between that in some cases there is a lack of clear distinction on
various nutrients and pancreatic cancer risk brings up some whether the select nutrient measured included intake
important points for consideration. First, it is common to find through food, supplementation, or a combination of the two.
studies on the same nutrient reporting different, and at times, This distinction is critical since, as just mentioned, nutrients
completely conflicting outcomes. Second, a common theme ingested as part of a food matrix can be absorbed differently
found in the analysis is that strength of the evidence varies than a nutrient given as a supplement.
depending on study design. While case-control studies are
more likely to find a significant association between the 3.2. Foods and pancreatic cancer risk
nutrient of interest and the outcome of pancreatic cancer,
these significant associations rarely ever hold in cohort We next reviewed the evidence on some of the most
studies. It is important to note that cohort studies are commonly studied foods and their impact on the risk of
considered to have a stronger and more robust design, free pancreatic cancer. Many studies have been conducted on
of some of the serious limitations that accompany the case- various foods, including red and processed meat, dairy
control studies. Because dietary intake is measured after products, fruits and vegetables, whole grains, alcoholic
disease diagnosis, case-control studies suffer from limitations beverages, and high glycemic foods (Table 2). In the sections
such as recall bias, reverse causality, and surrogate reporting. below, we summarize the most relevant findings for each of
Although study design is a significant source of conflicting, the foods mentioned and discuss the limitations of the
inconclusive results, measurement error and confounding evidence.
variables are serious limitations of epidemiological studies,
regardless of design. Both case-control and cohort studies 3.2.1. Red and processed meat
generally assess dietary intake using food frequency ques- Red meat, barbequed or cooked at high temperatures,
tionnaires, which have long been shown to be replete with contains heterocyclic amines (HCAs) which have been
inaccuracies and limitations [66]. In fact, some epidemiol- shown to be mutagens associated with pancreatic cancer
ogists have called into serious question the validity of [73]. Additionally, advanced glycation end products (AGEs)
these questionnaires [67], arguing that it is time to abandon found in red meat cooked at high temperatures have been
their use as a means of assessing the intake of specific hypothesized to cause chronic inflammation, explaining
nutrients, especially when attempting to assess their their possible association with pancreatic cancer [74]. Also,
association with diseases as complex as cancer [68]. N-nitroso compounds (NOCs), found in processed meats, are
Although these scientists purport that 7 day food diaries known carcinogens that increase the risk of pancreatic
may potentially circumvent the limitations of the food cancer [75], as may be the nitrite preservative [76].
frequency questionnaire, even these may not adequately The most recent and inclusive meta-analysis of 28 studies
reflect long-term intake [69]. Even in the one study cited involving red and processed meat consumption reports a
that measured dietary intake using a 7-day food diary, significant positive association for red meat in case-control
results of serum levels of vitamin C in the same study still studies, but not cohort studies [77]. When stratifying by sex,
conflicted with measures of dietary intake using the 7-day however, a significant positive association persists in men for
food diary, with the former indicating an inverse associa- red and processed meat consumption in cohort studies as
tion with pancreatic cancer, and the latter showing no well. A dose–response analysis indicates an 11% increased
association [19]. This suggests that assessing dietary intake risk of pancreatic cancer for every 100 g/day of red meat, but
through the currently available methods may not be no association for every 50 g/day of processed meat con-
appropriate for measuring intakes of specific nutrients, sumption [77]. An earlier meta-analysis of only prospective
and that future research on specific nutrients should opt studies did reveal, however, a 19% increase in pancreatic risk
for more objective biomarkers of nutritional status. for every 100 g of processed meat consumed daily [78]. It is
Another important issue for consideration with regards to important to carefully interpret these data, based on possible
attempting to associate the dietary intake of select nutrients confounders. For instance, high red meat eaters tend to also
with pancreatic cancer risk is the fact that these nutrients are have high alcohol intake and as mentioned in the introduc-
not commonly consumed (nor do they naturally exist) in tion, high alcohol intake is associated with higher pancreatic
isolation. Given the high correlation of various nutrients cancer risk. Thus, it remains unclear whether the observed
within a particular food, attempting to identify the specific associations are fully attributable to red meat intake itself,
nutrient responsible for a reduced risk of disease through because of potential residual confounding by unadjusted
epidemiological studies assessing dietary intake is rather dietary factors, such as alcohol consumption, that co-vary
questionable [70]. Nutrients present in a given food are not with red meat intake.
N U TR IT ION RE S EA R CH 5 2 ( 20 1 8 ) 1 –1 3 7

3.2.2. Dairy products 3.2.4. Whole grains


Dairy products contain nutrients that have been hypothe- Whole grains share some of the beneficial nutrients found in
sized to either promote or prevent carcinogenesis. Dairy fruits and vegetables previously discussed: mainly fiber,
products naturally contain calcium and have been fortified vitamin E, and folate, and for these reasons, there are
with vitamin D, which has been hypothesized to play a role in biologically plausible mechanisms by which they may reduce
preventing carcinogenesis [41,42]. On the other hand, dairy the risk of pancreatic cancer. Additionally, whole grain intake
products may also contain hormones and growth factors, has been associated with a reduced risk of diabetes [90], an
which can promote tumor growth [79-81]. established risk factor for pancreatic cancer [58].
Given this potential dual nature of dairy foods, it is not Given this association with diabetes, it is surprising that
surprising that epidemiological evidence is inconsistent and the relationship of whole grains to pancreatic cancer has only
conflicting. A pooled analysis of 14 cohort studies found no been assessed in a little more than a handful of case-control
association between pancreatic cancer risk and the intake of studies, and 1 cohort study to date. A meta-analysis of these 8
total milk, whole milk, low fat milk, cheese, cottage cheese, studies indicates an inverse association for the highest levels
ice cream, or yogurt [82]. of whole grain consumption, which is statistically significant
only for the case-control studies [91]. No association was
3.2.3. Fruits and vegetables found in the cohort study.
Fruits and vegetables contain many nutrients and phyto-
chemicals that have antioxidant, anti-mutagenic and anti- 3.2.5. High glycemic foods
carcinogenic properties [83-85]. The aforementioned vitamins As previously discussed, insulin resistance and type II diabetes
C, E, and folate as well as the carotenoids can be found in rich have been associated with an increased risk of pancreatic
quantities in many to most fruits and/or vegetables. In cancer [58]. There also have been associations made between
addition, isothiocyanates, phytochemicals found in crucifer- diabetes and foods high in glycemic index or glycemic load, as
ous vegetables are powerful inducers of the detoxification well as the post-prandial glycaemia that results after con-
enzymes, which assist in the removal of potentially carcino- sumption of such foods [92]. Moreover, some studies have
genic substances. Similarly, fiber present in fruits and found an association between high blood glucose levels and
vegetables can bind carcinogens, lower inflammatory the incidence of pancreatic cancer, particularly in women [93].
markers in circulation [86], and improve insulin metabolism Fructose-sweetened beverages have also been found to induce
[87], dysregulation of which is associated with pancreatic insulin resistance in overweight or obese individuals [94].
cancer [47,79,81]. Despite the biological plausibility of this association, a
The most recent and inclusive meta-analysis of 24 studies comprehensive meta-analysis of prospective studies found
assessing the association between fruit and vegetable intake no association between pancreatic cancer and glycemic
and pancreatic cancer risk found an inverse association, index, glycemic load, total carbohydrate intake, as well as
which was significant in case-control studies and borderline sucrose in both their high versus low and dose–response
significant in cohort studies [88]. When stratifying by geo- analyses [95].
graphic location, this correlation was only significant for Aune et al observed, however, a significant positive
vegetables in North America, whereas it was significant for association for highest levels of fructose consumption, as
both fruit and vegetables in Asia. A meta-analysis of 9 studies well as a 22% increased risk for every 25 g/day of fructose
assessing specifically cruciferous vegetable intake found a consumed [95]. There are some speculations regarding the
significant inverse association for the highest intake of mechanism on how fructose may increase pancreatic cancer
cruciferous vegetables, which remained significant in case- risk. These include: (a) the contribution of fructose to nucleic
control studies, but did not persist in cohort studies [89]. This acid synthesis through the pentose phosphate pathway
relationship was also most significant in studies conducted in (catalyzed by an activation of the transketolase enzyme),
the United States. which is greater than glucose [96]; and as previously

Table 2 – Associations between foods and pancreatic cancer risk


Food Association with pancreatic cancer risk Reference(s)

Red and processed meat Positive association (remains significant after [77,78]
stratification only in case control studies for
both sexes; remains significant in cohort studies for men)
Dairy products No association [82]
Fruits and vegetables Inverse association (only borderline significant in cohort studies) [88,89]
Whole grains Inverse association (only significant in case control studies) [91]
High glycemic foods No association for glycemic load, glycemic index, [95]
total carbohydrate intake, and sucrose.
Positive association for highest levels of fructose consumption
Alcoholic beverages Positive association for heavy drinkers [102,103]
8 N U TR IT ION RE S EA R CH 52 ( 20 1 8 ) 1 –1 3

mentioned (b) chronic fructose feeding to mice leads to an frequency questionnaires used by these studies, and their
increase in insulin resistance and obesity [97]. Thus, fructose, subsequent questionable validity [66-68]. This could also be
which is mainly ingested in the form of high fructose corn due to the numerous confounding variables present in such
syrup in processed foods and drinks, may increase pancreatic studies and the difficulty in adequately controlling for them
cancer risk. However, this finding of an increased risk with [106]. Potential confounders in these nutritional studies
fructose intake warrants further investigation in studies with include other foods consumed by the individual [107]. For
better adjustment for confounding factors. example, with respect to foods, an individual who consumes
a greater amount of red meat may also consume more alcohol
3.2.6. Alcoholic beverages and/or fructose-sweetened beverages and less fruits, vegeta-
Although alcohol itself has been classified as a Group 1 bles, and whole grains, rendering it difficult to pinpoint which
carcinogen [98], it is thought that its metabolites, such as food (whose presence or absence) may be contributing to the
acetaldehyde, may be more directly involved in carcinogen- association. Moreover, foods are also often consumed in
esis. With regards to pancreatic carcinogenesis, long-term combination with other foods, if not at a given meal, at least
alcohol consumption may be associated with chronic pancre- throughout a given day or week. This observation points,
atitis, a known risk factor for pancreatic cancer [99,100]. again, to the idea of food synergy, in which different foods
Additionally, acetaldehyde and other fatty acid ethyl esters (like nutrients) may interact to impact physiology in a way
produced from ethanol in pancreatic acinar cells may result in that is different than when consumed in isolation.
inflammation of the pancreas [100].
Several meta-analyses have been conducted to assess the 3.3. Dietary patterns and pancreatic cancer risk
association between alcohol consumption and the risk of
pancreatic cancer. One meta-analysis included a total of 32 The above findings and analysis suggest that one's dietary
studies and found an increased risk of pancreatic cancer for pattern may be more indicative of disease risk, as the effect of
those who consumed 3 or more drinks a day [1.22 (95% CI, a dietary pattern may be greater than the sum of its
1.12-1.34)], and no increase in risk for those who consumed constituent foods [108]. Of the variety of dietary patterns
less than 3 drinks a day [101]. The increased risk for heavy characteristic of an individual's eating behavior, nutritionists
drinkers remained, and was in fact stronger, in the cohort often divide them into two broad categories, known as the
studies. Similar results of increased association for heavy Western-style dietary pattern and the prudent/healthy die-
drinking were reported in two more recent (2016-2017) meta- tary pattern (Table 3). Similar in composition to the prudent
analyses [102,103]. On the other hand, a smaller meta- pattern, the Mediterranean diet has also garnered much
analysis of five case-control studies conducted in China failed research interest for its favorable associations with reduction
to find an association between alcohol consumption and in disease risk. In the sections below, we summarize the most
pancreatic cancer risk [104]. Limitations of this study, relevant findings for each of the dietary patterns mentioned
however, include the small number of studies analyzed, as and discuss the implications and limitations of the evidence.
well as the type of study included (case controls only) and the
specific study population. Finally, a large cohort study (not 3.3.1. Western-style dietary pattern
included in any of the meta-analyses) found that alcohol A Western-style dietary pattern is characterized by high
consumption was associated with earlier onset of pancreatic intake of red or processed meats, refined grains, sweets,
cancer, and this effect was seen most strongly in heavy high fat dairy products, potatoes, and low intake of fruits and
drinkers [105]. Thus, based on the above, there appears to be a vegetables [102]. A meta-analysis which included 18 studies
link between heavy alcohol consumption and the risk of that considered dietary intake consistent with a Western-
pancreatic cancer, implying that a threshold effect may be at style diet found a significant positive association for those in
play, where an effect is only seen once a certain level of the highest category of intake of foods in the Western-type
alcohol consumption is reached. dietary pattern compared to those in the lowest category of
intake (OR, 1.24; 95% CI, 1.06-1.45; P = .008) [102].
3.2.7. Discussion: foods and pancreatic cancer risk
The current evidence on foods and their association with 3.3.2. Prudent or healthy dietary pattern
pancreatic risk yields a few more consistent results than the A prudent or healthy dietary pattern is characterized by high
literature on individual nutrients, but still leaves much to be intakes of fruits, vegetables, whole grains, olive oil, fish, soy,
desired in terms of clear, unequivocal associations. Granted, poultry, and low fat dairy [102]. The aforementioned meta-
there does appear to be a positive association with the intake analysis included 17 studies that assessed a prudent or
of red and processed meat, for men in particular [77], a healthy dietary pattern and found a reduced risk of pancreatic
positive association with alcohol in heavy drinkers, and a cancer (OR, 0.85; 95% CI, 0.77-0.95; P = .004) for those in the
possible inverse association with the consumption of fruits highest category of intake of these foods as compared to those
and vegetables, particularly in Asian countries [88]. However, in the lowest category of intake [102].
the associations for other foods with biologically plausible
mechanisms for increasing or decreasing the risk of pancre- 3.3.3. Healthy Eating Index
atic cancer are overwhelmingly weak or null, particularly One of the cohort studies included in the previous meta-
when looking at prospective cohort studies [82,91,95]. analysis [102] assessed adherence to the 2005 Dietary Guide-
In agreement with our previous discussion, this could be lines for Americans [109]. Using the Healthy Eating Index to
largely due to the measurement error inherent in the food score adherence to a high quality diet composed of fruit,
N U TR IT ION RE S EA R CH 5 2 ( 20 1 8 ) 1 –1 3 9

vegetables, grains, legumes, and low fat dairy and meat, this have been possible in the Southern European cohort in the
study found a reduced risk of pancreatic cancer for those in EPIC study or in the Swedish study [111], associations are
the highest categories of compliance (HR, 0.85; 95% CI, 0.74- more likely to become evident.
0.97) [109]. Additionally, as it has been pointed out, although food
frequency questionnaires are rather crude measures, they
3.3.4. Mediterranean Diet may be adequate enough to detect broader measures of
Although one of the case-control studies in the meta-analysis consumption as in these dietary pattern analyses [68]. These
of dietary patterns [102] assessed adherence to the Mediter- dietary patterns, reflecting more realistically an individual's
ranean diet (MD), very few prospective studies have exclu- dietary intake and the complexity of the synergies between
sively examined adherence to a MD diet and its association nutrients in foods, and foods in combination with each other,
with pancreatic cancer risk. One cohort study calculated a are also more significantly associated with disease risk
non-alcohol MD score (arMED) based on 8 dietary components [71,108].
with high scores indicating a high consumption of fruits, It is interesting to note that the associations seen in these
vegetables, legumes, fish, olive oil, and cereals (both white meta-analyses of dietary patterns are not as dramatic as one
and non-white) and low consumption of meat and dairy might expect, especially when considering the large estimat-
products [110]. This prospective study conducted in 10 ed attribution of pancreatic cancer cases to dietary causes
European countries with over 500 000 participants failed to [6,8]. As pointed out above, this may be due to the weakness of
find a significant association between arMED score and current dietary assessment tools to accurately detect the
pancreatic cancer risk, but did find a borderline significant magnitude of associations as they truly are [68]. This could
association in Southern European cohorts, who tended to also be due to the difficulty in controlling for confounding
have higher arMED scores [110]. Another prospective study variables, as previously discussed [106]. Although many
conducted in North Sweden calculated a MD score based on studies attempt to statistically control for other risk factors
similar criteria with a few slight differences [111]. This study, for pancreatic cancer such as smoking, obesity, and diabetes,
which lasted for 18 years, included alcohol intake, only the statistical controls applied are far from perfect and may
considered whole-grain intake versus refined grains, and leave behind a residual confounding. Moreover, many studies
took into consideration the ratio of MUFA and PUFA to SFA. do not adequately control for all confounding variables, nor
A significant reduction in mortality due to pancreatic cancer can it be assumed that all possible confounders have been
was found [HR, 0.82 (95% CI, 0.68-0.99)] in men with highest recognized.
level of adherence to the MD diet according to these criteria as
compared to those with the lowest level of adherence.
4. Conclusion
3.3.5. Discussion: dietary patterns and pancreatic cancer risk
Although the number of studies are fewer, the evidence
Given that there are no current screening recommendations
analyzed indicates that dietary patterns are more significant-
for pancreatic cancer, primary prevention is of utmost impor-
ly associated with pancreatic cancer risk, with a healthy or
tance. Therefore, a better understanding of the etiology and
prudent pattern decreasing risk and a Western-style diet
risk factors, including dietary risk factors, is essential for the
increasing risk. The evidence for a link between the MD diet
primary prevention of this disease. Following our thorough
and pancreatic cancer is sparser, with insufficient studies to
analysis of the evidence on this topic, a key trend emerges. As
conduct a large meta-analysis. Despite this, analysis of the
we advance from individual nutrients to foods to dietary
cohort study that failed to detect an association brings up an
patterns, the evidence of certain associations with pancreatic
interesting point worth commenting on [110]. Although no
cancer risk becomes clearer, has more power, and is less
significant association was detected overall, a borderline
conflicting. This supports the notion of food synergy and
significant association was detected in populations that
prompts a more holistic approach to studying dietary risk.
tended to score higher with respect to adherence to a MD
diet. It is conceivable that with more extreme variation in the
adherence to a MD diet in the studied population, as may
5. Knowledge gaps and future research directions

Our analysis of the data revealed a number of limitations and


Table 3 – Associations between dietary patterns and
gaps in our current knowledge of dietary factors and
pancreatic cancer risk
pancreatic cancer risk. Below, we describe some of these
Dietary patterns Association with References(s) limitations and suggest avenues for future research to further
pancreatic cancer risk
our understanding in this critical area. First, in the meta-
Western-style Positive association [102] analyses reviewed assessing the association of nutrients and
(OR, 1.24; 95% CI, foods to pancreatic cancer risk, it was common to find that
1.06-1.45; P = .008) statistically significant results only remain significant after
Prudent or healthy Reduced risk of [102]
stratification in case control studies. Due to this, we empha-
pancreatic cancer
(OR, 0.85; 95% CI,
size the need to focus future research on epidemiological
0.77-0.95; P = .004) associations by using the more robust prospective cohort
design. This will lend more credence to any associations
10 N U TR IT ION RE S EA R CH 52 ( 20 1 8 ) 1 –1 3

found. Second, with respect to nutrients, many studies report propose that future epidemiological research studies, espe-
conflicting results, and this may be due in part to the cially prospective cohort studies, should focus on how a
inadequacy of food frequency questionnaires to accurately combination of these various factors may interact with
estimate dietary intake of the nutrients of interest. We dietary patterns to promote carcinogenesis of the pancreas.
therefore propose that food frequency questionnaires no Finally, and perhaps even more importantly, we see the
longer be used as tools to assess nutrient intake, and that need for future research to determine the most effective
more accurate and objective markers of nutritional status be evidence-based strategies for the implementation of what is
developed, validated, and utilized. Third, given the role of the already known. We call for an increase in nutrition and
food matrix and the concept of food synergy, we generally disease prevention education in our health science programs,
discourage a heavy focus on assessing the association of so physicians and nurses can encourage dietary and lifestyle
specific nutrients with pancreatic cancer risk. This does not practices that are known to reduce the risk of disease. Efforts
suggest that it is not important to study individual nutrients to assist and make accessible to individuals and communities
or specific foods. In fact, experimental studies, whether in the practice of the known healthy lifestyle choices are
vitro or in vivo, are valuable in elucidating the mechanisms by paramount [118]. Further research should be devoted to
which certain nutrients affect physiological or pathophysio- determining the best evidence-based practices in this area.
logical states, and may assist in developing therapeutic Not only does this have the potential to reduce the incidence
approaches. Thus, there is an urgent need for further of pancreatic cancer specifically, but numerous other chronic
experimental studies to research the role of nutrients in this diseases as well.
context.
In other words, it appears unwise to continue devoting
time, effort, and funds into determining specific nutrient-
Acknowledgment
disease associations through epidemiology for the purposes
of public health recommendations. It is highly unlikely that
This work was supported by funds from the University of
one or two specific nutrient deficiencies are the culprits
California, Davis to G.G.M. The authors declare no conflict of
behind the pandemic of such a highly complex and multifac-
interest.
torial disease as pancreatic cancer. Rather, it is likely a
combination of a number of nutrient deficiencies/excesses
along with oxidative stress or inflammation imposed by other
dietary choices, better reflected in the dietary pattern, that are REFERENCES
contributing to this disease process.
Given that diet is only one aspect of a lifestyle that [1] Siegel RL, Miller KD, Jemal A. Cancer Statistics, 2017. CA
contributes to the risk of pancreatic cancer, it appears critical Cancer J Clin 2017;67:7–30.
to consider diet within the context of other lifestyle and [2] Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman
established risk factors, such as smoking, obesity, physical JM, Matrisian LM. Projecting cancer incidence and deaths to
inactivity, and excessive alcohol consumption [9]. For exam- 2030: the unexpected burden of thyroid, liver, and pancreas
cancers in the United States. Cancer Res 2014;74:2913–21.
ple, the NIH-AARP Diet and Health study assessed the risk of
[3] Poplin E, Wasan H, Rolfe L, Raponi M, Ikdahl T, Bondarenko
pancreatic cancer based on lifestyle score and prospectively
I, et al. Randomized, multicenter, phase II study of CO-101
followed close to 500 000 participants for approximately 7 versus gemcitabine in patients with metastatic pancreatic
years [112]. The highest lifestyle score possible in their study ductal adenocarcinoma: including a prospective evaluation
was 5, which indicated that the individual never smoked or of the role of hENT1 in gemcitabine or CO-101 sensitivity. J
ceased at least 10 years ago, engaged in at least 20 minutes of Clin Orthod 2013;31:4453–61.
vigorous activity 3 to 4 times per week, had limited alcohol [4] Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al.
Distant metastasis occurs late during the genetic evolution
consumption, a healthy BMI, and adhered to a MD type diet. A
of pancreatic cancer. Nature 2010;467:1114–7.
58% reduction in pancreatic cancer risk was observed when [5] Wu S, Powers S, Zhu W, Hannun YA. Substantial
comparing those with the highest score (5) to those with the contribution of extrinsic risk factors to cancer development.
lowest (0). Similarly, a 46% reduction in risk was noted for Nature 2016;529:43–7.
those in the two highest score categories (4-5) as compared to [6] Anand P, Kunnumakkara AB, Sundaram C, Harikumar KB,
those in the lower categories (0-1). These dramatic and Tharakan ST, Lai OS, et al. Cancer is a preventable disease
significant associations represent compelling evidence that that requires major lifestyle changes. Pharm Res 2008;25:
2097–116.
we cannot truly separate diet alone as a causative factor in
[7] Maisonneuve P, Lowenfels AB. Risk factors for pancreatic
the etiology of pancreatic cancer. cancer: a summary review of meta-analytical studies. Int J
In fact, as previously mentioned, pancreatic cancer Epidemiol 2015;44:186–98.
presents multiple risk factors, which may have an additive [8] Willett WC. Diet and cancer. Oncologist 2000;5:393–404.
or compounding effect or interaction with each other. [9] Ilic M, Ilic I. Epidemiology of pancreatic cancer. World J
Established risk factors include diabetes, pancreatitis, Gastroenterol 2016;22:9694–705.
[10] Lee KW, Lee HJ, Surh YJ, Lee CY. Vitamin C and cancer
smoking, heavy drinking, and Helicobacter pylori infection [9],
chemoprevention: reappraisal. Am J Clin Nutr 2003;78:1074–8.
whereas others may be yet to be officially recognized or even
[11] Garcea G, Dennison AR, Steward WP, Berry DP. Role of
discovered. These may include exposure to environmental inflammation in pancreatic carcinogenesis and the
toxins [113,114], chronic stress [115], or even the effects of our implications for future therapy. Pancreatology 2005;5:
food system, both locally and globally [116,117]. Therefore, we 514–29.
N U TR IT ION RE S EA R CH 5 2 ( 20 1 8 ) 1 –1 3 11

[12] Leung PS, Chan YC. Role of oxidative stress in pancreatic [31] Peng L, Liu X, Lu Q, Tang T, Yang Z. Vitamin E Intake and
inflammation. Antioxid Redox Signal 2009;11:135–65. Pancreatic Cancer Risk: A Meta-Analysis of Observational
[13] McCullough ML, Giovannucci EL. Diet and cancer Studies. Med Sci Monit 2015;21:1249–55.
prevention. Oncogene 2004;23:6349–64. [32] Mongan NP, Gudas LJ. Diverse actions of retinoid receptors
[14] Woutersen RA, Appel MJ, Van Garderen-Hoetmer A. in cancer prevention and treatment. Differentiation 2007;75:
Modulation of pancreatic carcinogenesis by antioxidants. 853–70.
Food Chem Toxicol 1999;37:981–4. [33] Pettersson F, Dalgleish AG, Bissonnette RP, Colston KW.
[15] Chen J, Jiang W, Shao L, Zhong D, Wu Y, Cai J. Association Retinoids cause apoptosis in pancreatic cancer cells via
between intake of antioxidants and pancreatic cancer risk: a activation of RAR-gamma and altered expression of Bcl-2/
meta-analysis. Int J Food Sci Nutr 2016;67:744–53. Bax. Br J Cancer 2002;87:555–61.
[16] Fan H, Kou J, Han D, Li P, Zhang D, Wu Q, et al. Association [34] Rosewicz S, Wollbergs K, Von Lampe B, Matthes H, Kaiser A,
between vitamin C intake and the risk of pancreatic cancer: Riecken EO. Retinoids inhibit adhesion to laminin in human
a meta-analysis of observational studies. Sci Rep 2015;5: pancreatic carcinoma cells via the alpha 6 beta 1-integrin
13973. receptor. Gastroenterology 1997;112:532–42.
[17] Hua Y-F, Wang G-Q, Jiang W, Huang J, Chen G-C, Lu C-D. [35] Guan J, Zhang H, Wen Z, Gu Y, Cheng Y, Sun Y, et al. Retinoic
Vitamin C Intake and Pancreatic Cancer Risk: A Meta- acid inhibits pancreatic cancer cell migration and EMT
Analysis of Published Case-Control and Cohort Studies. through the downregulation of IL-6 in cancer associated
PLoS ONE 2016;11:e0148816. fibroblast cells. Cancer Lett 2014;345:132–9.
[18] Jeurnink SM, Ros MM, Leenders M, van Duijnhoven FJ, [36] Chronopoulos A, Robinson B, Sarper M, Cortes E,
Siersema PD, Jansen EH, et al. Plasma carotenoids, vitamin Auernheimer V, Lachowski D, et al. ATRA mechanically
C, retinol and tocopherols levels and pancreatic cancer risk reprograms pancreatic stellate cells to suppress matrix
within the European Prospective Investigation into Cancer remodelling and inhibit cancer cell invasion. Nat Commun
and Nutrition: a nested case-control study: plasma 2016;7:12630.
micronutrients and pancreatic cancer risk. Int J Cancer 2015; [37] Levy J, Bosin E, Feldman B, Giat Y, Miinster A, Danilenko M,
136:E665-6. et al. Lycopene is a more potent inhibitor of human cancer
[19] Banim PJ, Luben R, McTaggart A, Welch A, Wareham N, cell proliferation than either alpha-carotene or beta-
Khaw KT, et al. Dietary antioxidants and the aetiology of carotene. Nutr Cancer 1995;24:257–66.
pancreatic cancer: a cohort study using data from food [38] Assar EA, Vidalle MC, Chopra M, Hafizi S. Lycopene acts
diaries and biomarkers. Gut 2013;62:1489–96. through inhibition of IkappaB kinase to suppress NF-kappaB
[20] Sanchez GV, Weinstein SJ, Stolzenberg-Solomon RZ. Is signaling in human prostate and breast cancer cells.
dietary fat, vitamin D, or folate associated with pancreatic Tumour Biol 2016;37:9375–85.
cancer? Mol Carcinog 2012;51:119–27. [39] Zhang T, Chen H, Qin S, Wang M, Wang X, Zhang X, et al.
[21] Kim YI. Folate and DNA methylation: a mechanistic link The association between dietary vitamin A intake and
between folate deficiency and colorectal cancer? Cancer pancreatic cancer risk: a meta-analysis of 11 studies. Biosci
Epidemiol Biomarkers Prev 2004;13:511–9. Rep 2016;36.
[22] Lin HL, An QZ, Wang QZ, Liu CX. Folate intake and [40] Huang X, Gao Y, Zhi X, Ta N, Jiang H, Zheng J. Association
pancreatic cancer risk: an overall and dose–response meta- between vitamin A, retinol and carotenoid intake and
analysis. Public Health 2013;127:607–13. pancreatic cancer risk: Evidence from epidemiologic
[23] Bao Y, Michaud DS, Spiegelman D, Albanes D, Anderson KE, studies. Sci Rep 2016;6:38936.
Bernstein L, et al. Folate intake and risk of pancreatic cancer: [41] Ingraham BA, Bragdon B, Nohe A. Molecular basis of the
pooled analysis of prospective cohort studies. J Natl Cancer potential of vitamin D to prevent cancer. Curr Med Res Opin
Inst 2011;103:1840–50. 2008;24:139–49.
[24] Stolzenberg-Solomon RZ, Pietinen P, Barrett MJ, Taylor PR, [42] Barreto SG, Neale RE. Vitamin D and pancreatic cancer.
Virtamo J, Albanes D. Dietary and other methyl-group Cancer Lett 2015;368:1–6.
availability factors and pancreatic cancer risk in a cohort of [43] Liu SL, Zhao YP, Dai MH, You L, Wen Z, Xu JW. Vitamin D
male smokers. Am J Epidemiol 2001;153:680–7. status and the risk of pancreatic cancer: a meta-analysis.
[25] Calle EE, Rodriguez C, Jacobs EJ, Almon ML, Chao A, Chin Med J 2013;126:3356–9.
McCullough ML, et al. The American Cancer Society Cancer [44] Waterhouse M, Risch HA, Bosetti C, et al. Vitamin D and
Prevention Study II Nutrition Cohort. Cancer 2002;94: pancreatic cancer: a pooled analysis from the Pancreatic
2490–501. Cancer Case–Control Consortium. Ann Oncol 2015;26:
[26] Huang JY, Butler LM, Wang R, Jin A, Koh WP, Yuan JM. 1776–83. https://doi.org/10.1093/annonc/mdv236.
Dietary Intake of One-Carbon Metabolism-Related [45] Stolzenberg-Solomon RZ, Jacobs EJ, Arslan AA, Qi D, Patel
Nutrients and Pancreatic Cancer Risk: The Singapore AV, Helzlsouer KJ, et al. Circulating 25-hydroxyvitamin d
Chinese Health Study. Cancer Epidemiol Biomarkers Prev and risk of pancreatic cancer: cohort consortium vitamin D
2016;25:417–24. pooling project of rarer cancers. Am J Epidemiol 2010;172:
[27] Chuang S-C, Stolzenberg-Solomon R, Ueland PM, Vollset SE, 81–93.
Middtun Ø, Olsen A, et al. A U-shaped association between [46] Wolpin BM, Ng K, Bao Y, Kraft P, Stampfer MJ, Michaud
plasma folate and pancreatic cancer risk in the European DS, et al. Plasma 25-Hydroxyvitamin D and risk of
Prospective Investigation into Cancer and Nutrition. Eur J pancreatic cancer. Cancer Epidemiol Biomarkers Prev
Cancer 2011;47:1808–16. 2012;21:82–91.
[28] Traber MG, Atkinson J. Vitamin E, antioxidant and nothing [47] Bao Y, Ng K, Wolpin BM, Michaud DS, Giovannucci E, Fuchs
more. Free Radic Biol Med 2007;43:4–15. CS. Predicted vitamin D status and pancreatic cancer risk
[29] Heisler T, Towfigh S, Simon N, Liu C, McFadden DW. Peptide in two prospective cohort studies. Br J Cancer 2010;102:
YY augments gross inhibition by vitamin E succinate of 1422–7.
human pancreatic cancer cell growth. J Surg Res 2000;88:23–5. [48] SEER. Surveillance, Epidemiology, and End Results (SEER)
[30] Venkateswaran V, Fleshner NE, Klotz LH. Modulation of cell Program (1973–2011). National Cancer Institute, DCCPS,
proliferation and cell cycle regulators by vitamin E in Surveillance Research Program, Surveillance Systems
human prostate carcinoma cell lines. J Urol 2002;168: Branch; 1973–2011 [p. released April 2014, based on the
1578–82. November 3 submission].
12 N U TR IT ION RE S EA R CH 52 ( 20 1 8 ) 1 –1 3

[49] Coughlin SS, Calle EE, Patel AV, Thun MJ. Predictors of [71] Jacobs DR, Gross MD, Tapsell LC. Food synergy: an
pancreatic cancer mortality among a large cohort of United operational concept for understanding nutrition. Am J Clin
States adults. Cancer Causes Control 2000;11:915–23. Nutr 2009;89:1543S–8S.
[50] Parkin DM, Muir CS, Whelan SL, Gao YT, Ferlay J, Powell J. [72] Jacobs Jr DR, Tapsell LC. Food, not nutrients, is the
Cancer Incidence in Five Continents, Vol. VI; 1992. fundamental unit in nutrition. Nutr Rev 2007;65:439–50.
[51] Sherman MH, Yu RT, Engle DD, Ding N, Atkins AR, Tiriac H, [73] Stolzenberg-Solomon RZ, Cross AJ, Silverman DT, Schairer
et al. Vitamin D receptor-mediated stromal reprogramming C, Thompson FE, Kipnis V, et al. Meat and meat-mutagen
suppresses pancreatitis and enhances pancreatic cancer intake and pancreatic cancer risk in the NIH-AARP cohort.
therapy. Cell 2014;159:80–93. Cancer Epidemiol Biomarkers Prev 2007;16:2664–75.
[52] Vinceti M, Dennert G, Crespi CM, Zwahlen M, Brinkman M, [74] Jiao L, Stolzenberg-Solomon R, Zimmerman TP, Duan Z,
Zeegers MPA, et al. Selenium for preventing cancer. Cochrane Chen L, Kahle L, et al. Dietary consumption of advanced
Database Syst Rev 2014;3:CD005195 [CD] https://doi.org/10.1002/ glycation end products and pancreatic cancer in the
14651858.CD005195.pub3. prospective NIH-AARP Diet and Health Study. Am J Clin Nutr
[53] Wang L, Wang J, Liu X, Liu Q, Zhang G, Liang L. Association 2015;101:126–34.
between selenium intake and the risk of pancreatic cancer: [75] Risch HA. Etiology of pancreatic cancer, with a hypothesis
a meta-analysis of observational studies. Biosci Rep 2016;36: concerning the role of N-nitroso compounds and excess
e00395. gastric acidity. J Natl Cancer Inst 2003;95:948–60.
[54] Han X, Li J, Brasky TM, Xun P, Stevens J, White E, et al. [76] Aschebrook-Kilfoy B, Cross AJ, Stolzenberg-Solomon RZ,
Antioxidant intake and pancreatic cancer risk: the Vitamins Schatzkin A, Hollenbeck AR, Sinha R, et al. Pancreatic cancer
and Lifestyle (VITAL) Study. Cancer 2013;119:1314–20. and exposure to dietary nitrate and nitrite in the NIH-AARP
[55] Woutersen RA, Appel MJ, van Garderen-Hoetmer A, Diet and Health Study. Am J Epidemiol 2011;174:305–15.
Wijnands MV. Dietary fat and carcinogenesis. Mutat Res [77] Zhao Z, Yin Z, Pu Z, Zhao Q. Association between
1999;443:111–27. consumption of red and processed meat and pancreatic
[56] Tucker ON, Dannenberg AJ, Yang EK, Fahey III TJ. Bile acids cancer risk: a systematic review and meta-analysis. Clin
induce cyclooxygenase-2 expression in human pancreatic Gastroenterol Hepatol 2017;15:486–93 [e10].
cancer cell lines. Carcinogenesis 2004;25:419–23. [78] Larsson SC, Wolk A. Red and processed meat consumption
[57] Vessby B. Dietary fat and insulin action in humans. Br J Nutr and risk of pancreatic cancer: meta-analysis of prospective
2000;83(Suppl. 1):S91–6. studies. Br J Cancer 2012;106:603–7.
[58] Ben Q, Xu M, Ning X, Liu J, Hong S, Huang W, et al. Diabetes [79] Lin Y, Tamakoshi A, Kikuchi S, Yagyu K, Obata Y, Ishibashi
mellitus and risk of pancreatic cancer: A meta-analysis of T, et al. Serum insulin-like growth factor-I, insulin-like
cohort studies. Eur J Cancer 2011;47:1928–37. growth factor binding protein-3, and the risk of pancreatic
[59] Hardardottir I, Grunfeld C, Feingold KR. Effects of endotoxin cancer death. Int J Cancer 2004;110:584–8.
and cytokines on lipid metabolism. Curr Opin Lipidol 1994;5: [80] Maruyama K, Oshima T, Ohyama K. Exposure to exogenous
207–15. estrogen through intake of commercial milk produced from
[60] Shen QW, Yao QY. Total fat consumption and pancreatic pregnant cows. Pediatr Int 2010;52:33–8.
cancer risk: a meta-analysis of epidemiologic studies. Eur J [81] Nakao M, Hosono S, Ito H, Watanabe M, Mizuno N, Yatabe Y,
Cancer Prev 2015;24:278–85. et al. Interaction between IGF-1 polymorphisms and
[61] Yao X, Tian Z. Saturated, Monounsaturated and overweight for the risk of pancreatic cancer in Japanese. Int J
polyunsaturated fatty acids intake and risk of pancreatic Mol Epidemiol Genet 2011;2:354–66.
cancer: evidence from observational studies. PLoS ONE 2015; [82] Genkinger JM, Wang M, Li R, Albanes D, Anderson KE,
10:e0130870. Bernstein L, et al. Dairy products and pancreatic cancer risk:
[62] Qin B, Xun P, He K. Fish or long-chain (n-3) PUFA intake is a pooled analysis of 14 cohort studies. Ann Oncol 2014;25:
not associated with pancreatic cancer risk in a meta- 1106–15.
analysis and systematic review. J Nutr 2012;142:1067–73. [83] Lampe JW. Health effects of vegetables and fruit: assessing
[63] Hidaka A, Shimazu T, Sawada N, Yamaji T, Iwasaki M, mechanisms of action in human experimental studies. Am J
Sasazuki S, et al. Fish, n-3 PUFA consumption, and pancreatic Clin Nutr 1999;70:475s–90s.
cancer risk in Japanese: a large, population-based, prospective [84] Steinmetz KA, Potter JD. Vegetables, fruit, and cancer. II
cohort study. Am J Clin Nutr 2015;102:1490–7. Mechanisms. Cancer Causes Control 1991;2:427–42.
[64] Wang J, Wang W-J, Zhai L, Zhang D-F. Association of [85] Steinmetz KA, Potter JD. Vegetables, fruit, and cancer
cholesterol with risk of pancreatic cancer: A meta-analysis. prevention. J Am Diet Assoc 1996;96:1027–39.
World J Gastroenterol 2015;21:3711–9. [86] Ma Y, Hebert JR, Li W, Bertone-Johnson ER, Olendzki B,
[65] Chen H, Qin S, Wang M, Zhang T, Zhang S. Association Pagoto SL, et al. Association between dietary fiber and
between cholesterol intake and pancreatic cancer risk: markers of systemic inflammation in the Women's Health
evidence from a meta-analysis. Sci Rep 2015;5:8243. Initiative Observational Study. Nutrition 2008;24:941–9.
[66] Byers T. Food frequency dietary assessment: how bad is [87] Jensen MK, Koh-Banerjee P, Franz M, Sampson L, Gronbaek
good enough? Am J Epidemiol 2001;154:1087–8. M, Rimm EB. Whole grains, bran, and germ in relation to
[67] Kristal AR, Potter JD. Not the time to abandon the food homocysteine and markers of glycemic control, lipids, and
frequency questionnaire: counterpoint. Cancer Epidemiol inflammation 1. Am J Clin Nutr 2006;83:275–83.
Biomarkers Prev 2006;15:1759–60. [88] Wu Q-J, Wu L, Zheng L-Q, Xu X, Ji C, Gong T-T. Consumption
[68] Kristal AR, Peters U, Potter JD. Is it time to abandon the food of fruit and vegetables reduces risk of pancreatic cancer:
frequency questionnaire? Cancer Epidemiol Biomarkers evidence from epidemiological studies. Eur J Cancer Prev
Prev 2005;14:2826–8. 2016;25:196–205.
[69] Willett WC, Hu FB. Not the time to abandon the food [89] Li LY, Luo Y, Lu MD, Xu XW, Lin HD, Zheng ZQ. Cruciferous
frequency questionnaire: point. Cancer Epidemiol vegetable consumption and the risk of pancreatic cancer: a
Biomarkers Prev 2006;15:1757–8. meta-analysis. World J Surg Oncol 2015;13:44.
[70] Messina M, Lampe JW, Birt DF, Appel LJ. Reductionism and [90] Aune D, Norat T, Romundstad P, Vatten LJ. Whole grain and
the narrowing nutrition perspective: time for reevaluation refined grain consumption and the risk of type 2 diabetes: a
and emphasis on food synergy. J Acad Nutr Diet 2001;101: systematic review and dose-response meta-analysis of
1416. cohort studies. Eur J Epidemiol 2013;28:845–58.
N U TR IT ION RE S EA R CH 5 2 ( 20 1 8 ) 1 –1 3 13

[91] Lei Q, Zheng H, Bi J, Wang X, Jiang T, Gao X, et al. Whole presentation in sporadic pancreatic cancer in a dose-
grain intake reduces pancreatic cancer risk: a meta-analysis dependent manner: a multicenter study. Am J Gastroenterol
of observational studies. Medicine 2016;95:e2747. 2012;107:1730–9.
[92] Brand-Miller JC. Postprandial glycemia, glycemic index, and [106] Greenland S, Morgenstern H. Confounding in health
the prevention of type 2 diabetes. Am J Clin Nutr 2004;80: research. Annu Rev Public Health 2001;22:189–212.
243–4. [107] Ursin G, Ziegler RG, Subar AF, Graubard BI, Haile RW, Hoover
[93] Stocks T, Rapp K, Bjorge T, Manjer J, Ulmer H, Selmer R, et al. R. Dietary patterns associated with a low-fat diet in the
Blood glucose and risk of incident and fatal cancer in the national health examination follow-up study: identification
metabolic syndrome and cancer project (me-can): analysis of potential confounders for epidemiologic analyses. Am J
of six prospective cohorts. PLoS Med 2009;6:e1000201. Epidemiol 1993;137:916–27.
[94] Stanhope KL, Schwarz JM, Keim NL, Griffen SC, Bremer AA, [108] Hu FB. Dietary pattern analysis: a new direction in
Graham JL, et al. Consuming fructose-sweetened, not nutritional epidemiology. Curr Opin Lipidol 2002;13:3–9.
glucose-sweetened, beverages increases visceral adiposity [109] Arem H, Reedy J, Sampson J, Jiao L, Hollenbeck AR, Risch H,
and lipids and decreases insulin sensitivity in overweight/ et al. The Healthy Eating Index 2005 and Risk for Pancreatic
obese humans. J Clin Invest 2009;119:1322–34. Cancer in the NIH–AARP Study. J Natl Cancer Inst 2013;105:
[95] Aune D, Chan DSM, Vieira AR, Navarro Rosenblatt DA, Vieira 1298–305.
R, Greenwood DC, et al. Dietary fructose, carbohydrates, [110] Molina-Montes E, Sanchez MJ, Buckland G, Bueno-de-
glycemic indices and pancreatic cancer risk: a systematic Mesquita HB, Weiderpass E, Amiano P, et al. Mediterranean
review and meta-analysis of cohort studies. Ann Oncol diet and risk of pancreatic cancer in the European
2012;23:2536–46. Prospective Investigation into Cancer and Nutrition cohort.
[96] Liu H, Huang D, McArthur DL, Boros LG, Nissen N, Heaney Br J Cancer 2017;116:811–20.
AP. Fructose induces transketolase flux to promote [111] Tognon G, Nilsson LM, Lissner L, Johansson I, Hallmans G,
pancreatic cancer growth. Cancer Res 2010;70:6368–76. Lindahl B, et al. The Mediterranean diet score and mortality
[97] Dekker MJ, Su Q, Baker C, Rutledge AC, Adeli K. Fructose: a are inversely associated in adults living in the subarctic
highly lipogenic nutrient implicated in insulin resistance, region. J Nutr 2012;142:1547–53.
hepatic steatosis, and the metabolic syndrome. Am J Physiol [112] Jiao L, Mitrou PN, Reedy J, Graubard BI, Hollenbeck AR,
Endocrinol Metab 2010;299:E685-4. Schatzkin A, et al. A Combined healthy lifestyle score and
[98] Baan R, Grosse Y, Straif K, Secretan B, El Ghissassi F, risk of pancreatic cancer – a large cohort study. Arch Intern
Bouvard V, et al. A review of human carcinogens–Part F: Med 2009;169:764–70.
chemical agents and related occupations. Lancet Oncol [113] Alavanja MC, Hoppin JA, Kamel F. Health effects of chronic
2009;10:1143–4. pesticide exposure: cancer and neurotoxicity* 3. Annu Rev
[99] Duell EJ. Epidemiology and potential mechanisms of Public Health 2004;25:155–97.
tobacco smoking and heavy alcohol consumption in [114] Fucic A, Gamulin M, Ferencic Z, Katic J, Krayer von Krauss M,
pancreatic cancer. Mol Carcinog 2012;51:40–52. Bartonova A, et al. Environmental exposure to
[100] Zakhari S. Chronic alcohol drinking: Liver and pancreatic xenoestrogens and oestrogen related cancers: reproductive
cancer? Clin Res Hepatol Gastroenterol 2015;39:S86–91. system, breast, lung, kidney, pancreas, and brain. Environ
[101] Tramacere I, Scotti L, Jenab M, Bagnardi V, Bellocco R, Rota M, Health 2012;11:S8.
et al. Alcohol drinking and pancreatic cancer risk: a meta- [115] Kim-Fuchs C, Le CP, Pimentel MA, Shackleford D, Ferrari D,
analysis of the dose-risk relation. Int J Cancer 2010;126:1474–86. Angst E, et al. Chronic stress accelerates pancreatic cancer
[102] Lu P-Y, Shu L, Shen S-S, Chen X-J, Zhang X-Y. Dietary growth and invasion: a critical role for beta-adrenergic
patterns and pancreatic cancer risk: a meta-analysis. signaling in the pancreatic microenvironment. Brain Behav
Nutrients 2017;9:38. Immun 2014;40:40–7.
[103] Wang Y-T, Gou Y-W, Jin W-W, Xiao M, Fang H-Y. [116] Popkin BM. Understanding global nutrition dynamics as a
Association between alcohol intake and the risk of pancre- step towards controlling cancer incidence. Nat Rev Cancer
atic cancer: a dose–response meta-analysis of cohort 2007;7:61–7.
studies. BMC Cancer 2016;16:212. [117] Wallinga D. Today's Food System: How Healthy Is It? J
[104] Li Y, Yang H, Cao J. Association between alcohol consumption Hunger Environ Nutr 2009;4:251–81.
and cancers in the Chinese population—a systematic review [118] Amine E, Baba N, Belhadj M, Deurenbery-Yap M, Djazayery
and meta-analysis. PLoS ONE 2011;6:e18776. A, Forrester T, et al. Diet, nutrition and the prevention of
[105] Anderson MA, Zolotarevsky E, Cooper KL, Sherman S, Shats chronic diseases: report of a Joint WHO/FAO Expert
O, Whitcomb DC, et al. Alcohol and tobacco lower the age of Consultation. World Health Organization; 2002.

Vous aimerez peut-être aussi