Journal of the Pancreas Open Access

  • ISSN: 1590-8577
  • Journal h-index: 80
  • Journal CiteScore: 29.12
  • Journal Impact Factor: 19.45*
  • Average acceptance to publication time (5-7 days)
  • Average article processing time (30-45 days) Less than 5 volumes 30 days
    8 - 9 volumes 40 days
    10 and more volumes 45 days
Reach us +44 7460731551

- (2016) Volume 17, Issue 2

Possible Involvement of Pancreatic Fatty Infiltration in Pancreatic Carcinogenesis

Mika Hori1, Michihiro Mutoh2,3, Toshio Imai4, Hitoshi Nakagama3 and Mami Takahashi4*

1Department of Molecular Innovation in Lipidology, National Cerebral and Cardiovascular Center Research Institute, 5-7-1 Fujishirodai, Suita, Osaka 565-8565, Japan

2Epidemiology and Prevention Division, Research Center for Cancer Prevention and Screening, Japan

3Division of Carcinogenesis and Cancer Prevention, Japan

4Central Animal Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan

*Corresponding Author:

Mami Takahashi
Central Animal Division
National Cancer Center Research Institute
5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045
Japan
Phone +81-3-3542-2511
Fax +81-3-3543-9305
E-mail mtakahas@ncc.go.jp

Received: November 20th, 2015 Accepted: December 30th, 2015

Visit for more related articles at Journal of the Pancreas

Abstract

Pancreatic cancer is difficult to diagnose in its early stage and is one of the most lethal human cancers. Thus, it is important to clarify its major risk factors, predictive factors and etiology. Here, we focus on fatty infiltration of the pancreas and suggest that it could be a risk factor for pancreatic cancer. Fatty infiltration of the pancreas is observed as ectopic adipocytes infiltrating the pancreatic tissue and is positively correlated with obesity and the prevalence of diabetes mellitus, which are risk factors for pancreatic cancer. However, whether fatty infiltration is a major risk factor for pancreatic cancer has not been established. Recent clinical studies show there is a positive correlation between fatty infiltration of the pancreas and pancreatic precancerous lesions or ductal adenocarcinomas. Animal experimental studies also show an association between fatty infiltration of the pancreas and pancreatic precancerous lesions or ductal adenocarcinomas development. Syrian golden hamsters, which are sensitive to chemical carcinogens in the pancreas, develop fatty infiltration of the pancreas with age. The combination of a high-fat diet and a chemical carcinogen that induces a K-ras mutation increases the severity of fatty infiltration of the pancreas. Thus, fatty infiltration of the pancreas is suggested to promote pancreatic carcinogenesis via a K-ras activating mutation. It is assumed that increased expression of adipokines and of inflammatory and proliferation-associated factors elicited by fatty infiltration of the pancreas may contribute to pancreatic precancerous lesions or ductal adenocarcinomas development. Accumulating evidence suggests that in addition to suppression of Ras activation, methods to modulate fatty infiltration in the pancreas can be considered as a strategy for preventing pancreatic cancer.

Keywords

Diabetes Mellitus; Obesity; Pancreatic Neoplasms

Abbreviations

FI fatty infiltration; NAFLD non-alcoholic fatty liver diseases; NAFPD nonalcoholic fatty pancreas disease; NASH non-alcoholic steoatohepatitis; PDAC pancreatic ductal adenocarcinoma

INTRODUCTION

Obesity causes accumulation of visceral and ectopic fats. Recently, ectopic fats, which are defined as excess adipose tissue in locations not classically associated with adipose tissue storage [1], have been gathering attention. Ectopic fats are commonly observed in the liver, heart, muscle and pancreas and directly exhibit lipotoxicity or indirectly secrete cytokines when accompanied by inflammation in these organs. Of note, fat deposition into hepatocytes is found in hepatic steatosis. The pathophysiology, diagnostic criteria and clinical implications of hepatic steatosis have already been established [2]. Obesity, diabetes and alcohol consumption are known to be strongly associated with hepatic steatosis [3, 4]. Obesity and diabetes are the leading causes of non-alcoholic fatty liver diseases (NAFLD) / non-alcoholic steoatohepatitis (NASH), and NASH is known to promote hepatocellular carcinoma development [5]. Ectopic fat deposition in the pancreas is synonymous with “fatty pancreas”, “nonalcoholic fatty pancreas disease (NAFPD)”, “fatty infiltration (FI)” or “fatty replacement”. In FI of the pancreas, which is distinct from hepatic steatosis, triglycerides accumulate in adipocytes in the pancreatic tissue.

In this review, we focus on pancreatic FI and its possible contribution to carcinogenesis and development of pancreatic ductal adenocarcinoma (PDAC) in humans and animal models. In addition, we review genetic background of metabolic syndrome and carcinogenesis of PDAC, and discuss the mechanisms behind the pathological consequences of FI of the pancreas.

FATTY INFILTRATION OF THE PANCREAS

Pathology

FI of the pancreas has been observed pathologically for many years. This phenomenon is characterized by adipocyte infiltration of the area between pancreatic lobules, accumulating around great vessels (interlobular fat), and the lobules themselves, accumulating with a scattered pattern (intralobular fat). In humans, pancreatic FI appears mainly in the interlobular fat rather than in intralobular fat [6-8]. Histopathologically, only pancreatic islet cells are observed in massive adipocyte depots, and thus, pancreatic islet cells may be resistant to FI [9].

Apart from obesity or age-related pancreatic FI, extreme degrees of pancreatic adiposity have been traditionally observed in infants [10], young adults [11] and elderly adults [12], and have been described as lipomatous pseudohypertrophy of the pancreas. Lipomatous pseudohypertrophy of the pancreas is benign and characterized by an enlarged pancreas with a massive replacement of pancreatic exocrine tissue by adipose tissue; however, the shape of the pancreas and the islets of Langerhans are preserved.

Detection and Assessment

FI of the pancreas is relatively common and is identified based on magnetic resonance imaging (MRI) [13], magnetic resonance spectroscopy [14], endoscopic ultrasound (comparison the echogenicity of the pancreas with that of the normal liver) [6, 15] or computed tomography (CT; a decrease in the density of the circumscribed areas) [16]. Marks et al. reported that increased pancreatic echogenicity on abdominal ultrasonography, which they termed “bright pancreas”, might represent a fatty change of the pancreas [6].

As quantitative methods to measure FI of the pancreas, the use of MRI (percentage decreases in pancreatic signal intensity on opposed phase images relative to those on in-phase images) or CT (mean Hounsfield unit (HU) of three pancreatic regions (head, body and tail), a threshold of -190 to -30 HU) has been reported to correlate well with histopathological findings [17, 18]. However, these methods did not show sufficient resolution for quantitative measurement of FI of the pancreas. To date, Kim et al. have reported an association between histologic pancreatic fat and CT attenuation indices, such as the difference between pancreatic and splenic attenuation (P-S) and the pancreas-to-spleen attenuation ratio (P/S) on unenhanced CT images [19]. The conventional P-S and P/S measurement could be used to evaluate FI of the pancreas on unenhanced CT images. Recently, pancreatic volume and fat deposition have also been quantitatively assessed using a multidetector CT (histogram analysis, CT attenuation, a threshold of -190 to -30 HU) [20].

A quantitative MRI study revealed a correlation between pancreatic fat and obesity [21]. In addition, FI of the pancreas measured by sonographic analysis was associated with higher levels of total cholesterol, LDL-cholesterol (LDL-C), triglycerides, FFAs, insulin and HOMA-IR [22, 23]. Kim et al. reported that the degree of pancreatic FI was significantly associated with a clinical presence of impaired glucose metabolism measured with the CT attenuation index [19].

Etiology

The etiology of FI of the pancreas is complicated and multifactorial. FI of the pancreas is positively correlated with age, BMI, a history of diabetic mellitus (DM) and fatty liver [9, 22, 24-27] (Table 1). The severity of FI in the pancreas increases with age or obesity [9, 28]. It is known that obesity-related FI of the pancreas could be improved after weight reduction. Van der Zijl et al. described that pancreatic fat increases with impaired glucose metabolism independent of aging and obesity [29]. However, they could not establish a direct relationship between pancreatic fat and diminished β-cell function in subjects with impaired glucose metabolism [29]. Thus, whether lipid accumulation occurs in pancreatic islets and whether this contributes to β-cell dysfunction in humans is still under debate [30, 31].

Ischemia from impaired pancreatic arteries promotes FI [32]. Pancreatic ischemia is spontaneously induced by atherosclerosis or blood clots in the pancreatic artery. Risk factors for atherosclerosis include smoking, high LDL-C levels, insulin resistance, DM [33], obesity, lack of physical activity [34] and aging. These risk factors are also associated with FI of the pancreas and pancreatic cancer. Thus, FI of the pancreas and pancreatic cancer may be associated with ischemia.

FI represents replacement of acinar cells by adipocytes, which is induced by acinar cell death or by fat accumulation promoted by obesity and metabolic syndrome [35]. Loss of acinar cells resulting from various types of damage is a more significant contributor to pancreatic FI than obesity. Induction of pancreatic FI after ligation of the pancreatic duct along with parenchymal loss has been observed in experimental animal models [36, 37]. In a genetic mouse model of pancreatic duct malformation in which the Notch signaling gene Jagged1 is knocked out, abnormal pancreatic ducts are accompanied by acinar cell death, pancreatitis, FI and/or fibrosis in the pancreatic parenchyma [38]. Thus, FI of the pancreas can be induced secondarily by tumor occupation of pancreatic ducts or by obesity and metabolic syndrome. Although the genomic changes responsible for the formation of FI of the pancreas are not clearly known, some genes are addressed in the next section.

Possible Genetic Backgrounds

FI of the pancreas has been reported in several monogenic conditions: cystic fibrosis [39], Shwachman- Diamond syndrome [40] and Johanson-Blizzard syndrome [41]. In addition, pancreatic lipomatosis has been observed in nondiabetic children possessing mutations of the carboxyl-ester lipase gene [42].

The prevalence of obesity and diabetes is partially due to a westernized lifestyle. In addition, obesity- or diabetes- susceptibility loci or single nucleotide polymorphisms (SNPs) have been discovered through genome-wide association studies [43]. SNPs near the melanocortin 4 receptor (MC4R) gene are associated with increased risks of obesity and diabetes [44, 45]. One SNP, MC4R rs17782313, has been reported to correlate with an increased risk for breast [46], colorectal [47] and endometrial [48] cancers, and these associations persist after adjustment for BMI [46-48]. MC4R plays an important role in energy metabolism, and MC4R-deficient mice develop steatohepatitis in addition to obesity, insulin resistance and dyslipidemia when fed a high-fat diet (HFD) [49]. Homozygous null mutations in proopiomelanocortin (POMC) and heterozygous loss-of-function mutations in α- and β-melanocyte-stimulating hormone (α- and β-MSH) also increase obesity risk in human [50, 51]. A common variant in the fat mass and obesity associated (FTO) gene is associated with a predisposition for diabetes by affecting BMI [52]. Variants in the patatin-like phospholipase containing domain 3 gene (PNPLA3) or glucokinase regulatory protein (GCKR) have been reported to be associated with NAFLD [53, 54]. Both NAFLD and fatty pancreas were also associated with diabetes independently of age, gender, adiposity, and other cardiometabolic risk factors [55]. To date, there is no information about associations between these genetic backgrounds and susceptibility to FI of the pancreas; however, some genetic backgrounds related to obesity/metabolic syndrome may be involved in occurrence of severe FI.

ASSOCIATION OF FI OF THE PANCREAS WITH PANCREATIC CARCINOGENESIS IN HUMANS

Pancreatic cancer occurs with a high incidence and mortality rate in developed countries [56]. In Japan, the age-adjusted mortality rate of pancreatic cancer has increased by approximately 9-fold in both males and females from 1950 to 1995 [57]. This phenomenon may be explained by an increase of obesity and DM resulting from a westernization of the common life style including HFDs and low physical activity. Indeed, from 1946 to 1996, dietary fat intake has increased more than 4-fold [58]. In addition, becoming an aging society may also contribute to increase the total number of pancreatic cancer patients.

In addition to age [59], obesity [60, 61] and DM [62], epidemiological studies have shown that smoking[63, 64] and chronic pancreatitis[65, 66], which are reported as etiologies of FI of the pancreas, are also major risk factors for pancreatic cancer. Thus, an association between FI of the pancreas and pancreatic cancer is possible, but to date it is not evident whether FI in the pancreas itself is a risk factor.

Implication of K-ras Mutation

The top two common smoking-associated cancers are adenocarcinomas of the lungs and pancreas [67]. Cigarette smoke contains toxic chemicals such as 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). NNK is known as the most potent carcinogen and is formed under reactions with metabolites of nicotine. In mice and rats, O6-methyguanine formation is associated with a point mutation in the K-ras gene in NNK-induced pulmonary adenocarcinomas [68, 69]. Westra et al. reported more K-ras gene mutations were found in lung adenocarcinomas obtained from current and former smokers (approximately 30%, respectively) than in nonsmokers (7%; P = 0.015) [70]. Activating mutations of the K-ras gene are the most common genetic abnormality in PDAC and are present in approximately 90% of cases [71] even in low-grade PanIN [72]. Thus, they are the earliest genetic abnormalities observed in the progression of PDAC. In addition, K-ras mutations in mucous and in ductal / desquamated cells involved in pancreatic juice production are detected in patients with chronic and/or autoimmune pancreatitis [73- 75]. However, an association between smoking and K-ras gene mutations in pancreatic cancer is controversial [76, 77]. Statistically, it may be impossible to detect an effect of smoking on the K-ras gene because most pancreatic cancer patients harbor the K-ras mutation. Thus, smoking remains one of the likely causes of K-ras mutation.

Data from Clinical Studies

We found and reported on a case in which PDAC developed in the head of the pancreas markedly infiltrated by adipose tissues [78]. This patient underwent distal gastrectomy against early gastric cancer 9 years ago, and 5 years later, CT scans showed that most of the pancreas had been replaced by fat tissue. Namely, fatty pancreas was observed 4 years before PDAC development. Subsequently, we performed a case-control study of FI of the pancreas prevalence between controls (patients with distal bile duct cancer, cancer of the ampulla of Vater, gallbladder cancer, or duodenal cancer) and cases (patients with pancreatic head ductal carcinoma)[79]. The degrees of FI of the pancreas were measured histopathologically from the area occupied by adipocytes in pancreatic sections in the study. The results showed that the degree of FI of the pancreas was significantly higher in PDAC cases than in non-PDAC controls (median 26 vs. 15%, P<0.001). It is also suggested that FI of the pancreas may increase the risk of pancreatic cancer beyond the effect of obesity itself (OR=6.1; adjusted with BMI, DM and other confounding factors, P<0.001, Table 2). Rebours et al. have also reported that PanINs were associated with the degree of intralobular fibrosis and the intralobular fat from surgical specimens of normal pancreatic tissue [80]. The odds ratio (OR) in intralobular fat (OR=17.86) was higher than that in intralobular fibrosis (OR=5.61). The number of PanIN lesions also increased with the severity of hepatic steatosis, but not with the percentage of subcutaneous fat area or BMI [80].

ASSOCIATION OF FI OF THE PANCREAS WITH PANCREATIC CARCINOGENESIS IN ANIMAL MODELS

The initiation of pancreatic carcinogenesis results from K-ras activation, which induces precancerous lesions, PanINs. In addition to activation of the K-ras gene, either nongenetic events involving tissue damage and/or an inflammatory response or genetic events causing loss of p16/Ink4a or p53 are necessary for the formation of PDAC [81]. Pancreatitis has been shown to contribute to PDAC progression by abrogating the senescence barrier characteristic of low-grade mPanIN expansion [82]. Pathologically, development of PDACs would be promoted by adipocytes during FI after the K-ras activation and by loss of acinar cells by carcinogens.

BOP-Treated Hamster Model for Pancreatic Carcinogenesis

The hamster is a unique model animal that can develop PDAC by the subcutaneous injections of N-nitrosobis(2- oxopropyl)amine (BOP)[83]. Moreover, similar to human PDACs [84, 85], point mutations in codon 12 of the K-ras gene are frequently observed and expression of the fragile histidine triad gene is aberrant in BOP-treated hamsters [86]. The p16 gene is one of the most frequently inactivated tumor-suppressor genes in human PDACs [87], and loss of p16 expression has also been found in hamster PDAC lesions [88]. Syrian golden hamsters have hyperlipidemia. The effect of a HFD on hyperlipidemia and pancreatic carcinogenesis was examined in BOP-treated hamsters, and a HFD was shown to enhance PDAC formation with a corresponding increase of FI of the pancreas, BW, and serum lipid and leptin levels [89].

Mouse and Rat Models for Pancreatic Carcinogenesis

Unlike hamsters, mice and rats are not susceptible to chemically induced pancreatic carcinogenesis. They do not develop PDAC upon treatment with BOP [90]. The Syrian golden hamster shows hyperlipidemia because hepatic lipoprotein lipase activity is low compared with that of mice and rats. Our previous study has shown that the OLETF rat, a model of T2DM accompanied by hypertriglyceridemia, failed to develop BOP-induced PDAC [91]. It was notable that FI of the pancreas of OLETF rats did not develop to the same degree as that observed in Syrian golden hamsters, and the data indicated that hypertriglyceridemia and/or hyperinsulinemia in T2DM are not sufficient to increase susceptibility to pancreatic carcinogenesis. It is assumed that the enzymatic activation and detoxification of BOP as well as the ability of BOP metabolites to induce formation of DNA adducts is different between hamsters and mice/ rats. Another difference is the basal potential to develop FI. Hamsters have more scattered FI in the pancreas than mice/rats. Thus, it is possible that sensitivity to pancreatic carcinogens depends on the degree of pancreatic FI.

Despite the fact that mice are less susceptible to PDAC, many genetically engineered mouse models of PDAC have been developed [92]. A mouse strain carrying a conditional knocked-in of a KRasG12D allele silenced by a floxed STOP transcriptional cassette (LSL-KRasG12D) was crossed to transgenic strains that expressed the bacterial Cre recombinase under the control of gene promoters of pancreas-specific factors, such as Pdx1, Ptf1a/P48 [93] or elastase. These transgenic mice develop acinar-to-ductal metaplasia and PanINs via K-ras activation [93, 94]. A small proportion of these mice develop PDAC after a prolonged latency period (>12 months) because additional alterations, including p16 [95], p53 [96] and dpc4 [97, 98], are necessary for development of PDAC in LSL-KRasG12D mice.

Pigment epithelium-derived factor (PEDF) is a multifunctional secreted protein, for example, it has antiangiogenic, antiproliferative and neurotrophic activities [99, 100]. It has been reported that Elastase- KrasG12D/PEDF deficient mice developed invasive PDAC and increased FI of the pancreas accompanied by stromal expression of two lipid droplet associated proteins, tail-interacting protein 47 (TIP47, perilipin 3) and adipose differentiation-related protein (ADRP, perilipin 2) [101]. Ras oncogenes themselves are sufficient to induce differentiation of 3T3-L1 fibroblasts into adipocytes [102], and additional PEDF deficiency may promote adipogenesis by cancelling its suppressive role on master adipogenic transcription factors such as CCAAT-enhancer-binding protein α (C/EBP-α), and peroxisome proliferatoractivated receptor γ (PPARγ) that are upregulated through the Ras/MAPK pathway[103].

It is known that HF and high-calorie diets promote pancreatic neoplasia in Pdx-1-KRasG12D mice [104]. The pancreas of these mice have an increased number of infiltrating inflammatory cells, increased stromal fibrosis and more advanced PanIN lesions; however, FI of the pancreas has not been described in these mice.

ADIPOCYTE-ASSOCIATED FACTORS CONTRIBUTING TO TUMOR PROMOTION

Possible mechanisms of tumor promotion by FI of the pancreas include an increase of inflammatory- and proliferation-associated factors derived from infiltrated adipocytes.

Inflammatory Factors

Adipose tissue has been characterized as an organ that secretes cytokines such as TNF-α, IL-6, and monocyte chemotactic protein-1 (MCP-1). In turn, macrophages accumulate around adipose tissue and produce cytokines including IL-1β thereby aggravating a state of inflammation. NAFPD presents with inflammation accompanied with FI of the pancreas, and inflammatory cells such as macrophages can accumulate around adipose cells. In BOP-treated hamsters, a HFD increased or tended to increase the pancreatic expression of MCP-1, IL-1β and COX-2 as well as aggravate FI and PDAC [89]. These cytokines could promote inflammation and contribute to tumor progression. Overproduction of PGE2 by COX-2 has been suggested to activate cell proliferation, angiogenesis and anti-apoptotic mechanisms [105, 106]. PGE2 has the ability to differentiate 3T3-L1 cells to adipocytes [107]. Additionally, COX-2 expression is regulated by Wnt and Ras signaling [108]. Oncogenic K-ras increases COX- 2 levels in intestinal epithelial cells [109] and NF-κBmediated positive feedback involving COX-2 expression could increase in pancreatic duct cells with oncogenic K-ras [110].

Adipokines

Adipose tissue has been described as an endocrine organ that produces adipokines such as leptin and adiponectin. The serum levels of adipokines such as leptin and adiponectin in BOP-treated hamsters fed HFDs were higher than those in the BOP-treated hamsters fed a standard diet (STD) [89]. Leptin is a hormone whose level increases in the serum with the onset of obesity. There are reports about allografted tumor growth in obese mice. To determine whether diet-induced obesity (DIO) could result in FI of the pancreas in mice, C57BL/6 mice were fed HFDs. In addition to insulin resistance and/or increased serum leptin levels, FI of the pancreas was observed in the mice with DIO [111-113]. DIO increased the proliferation of orthotopically implanted pancreatic tumor cells in vivo via an activation of the PI3K/Akt pathway by leptin signaling [111]. In vitro, leptin directly stimulates the phosphorylation of Akt, with increased pancreatic cancer cell proliferation and migration. When lean (C57BL/6J) and obese (Lepob and Lepdb) mice were inoculated with murine pancreatic cancer cells, obese mice developed larger tumors, metastases, intratumoral adipocyte masses, and greater mortality than lean mice [114]. These findings suggest that both insulin resistance and an altered adipokine milieu could lead directly to changes in the microenvironment.

Free Fatty Acids

Fatty tissue that accumulates during FI of the pancreas contains high levels of triglycerides and FFAs. Serum FFA levels are correlated with the degree of FI of the pancreas in BOP-treated hamsters. Moreover, the expression of fatty acid synthase, representing de novo FFA synthesis, is enhanced in the pancreas of BOP+HFD-treated hamsters compared with BOP+STD-treated hamsters [89]. A high level of FFAs is toxic to cells due to its peroxidation [115, 116], suggesting that FFAs could damage pancreatic acinar cells and induce FI.

Growth Factors

The mRNA levels of insulin, IGF-I and cyclin D1 were elevated in the pancreas of BOP-treated hamsters with HFD [89]. Insulin and IGF-I promote the differentiation of 3T3- L1 cells to adipocytes [117], and thus, FI may increase in the pancreas. IGF-I is known to stimulate cell proliferation [118]. In KrasInk4a+/- mice, IGF-I levels clearly represent the diet-dependent changes in body fat and PDAC development compared with other markers, such as adiponectin, leptin and insulin [119]. Leptin, insulin and IGF-I could promote cancer development thorough induction of inflammation-and growth–related gene expression.

Angiogenesis Factors

Serum levels of VEGF, VEGF-C and VEGF-D are elevated in overweight/obese subjects [120]. Leptin secreted by adipocytes increases VEGF levels by activating STAT3. VEGF induces angiogenesis and promotes tumor growth [121]. Thus, FI has the potential to promote tumor angiogenesis in the pancreas. Angiotensin II, which is secreted by adipocytes, also increases the production of VEGF. VEGF-C and VEGF-D are reported to enhance lymphatic metastasis of PDAC [122].

CHALLENGES FOR THE FUTURE

Prospective Study

We cannot distinguish whether FI is a risk factor or a consequence of pancreatic cancer by case-control studies or retrospective clinical studies. The only way to demonstrate that FI is a risk factor for PDAC is to perform a prospective cohort study that examines whether individuals with severe FI in the pancreas could develop PDAC. The implementation of a large cohort is needed because the incidence of pancreatic cancer is low. As described above, the conventional CT attenuation index on unenhanced CT images has been recently established for the detection of fatty pancreas [19]. Mass screening of individuals with severe FI of the pancreas (so-called “fatty pancreas”) using such conventional methods may be useful for a future prospective study to assess fatty pancreas, as a high-risk marker for pancreatic cancer. In addition to the image diagnosis, the establishment of useful biomarkers for fatty pancreas is also desired to support the screening.

Prevention Study

If fatty pancreas is determined to be a risk for pancreatic cancer, therapeutic intervention of FI of the pancreas could increase prevention of pancreatic cancer development. Indeed, weight reduction, high physical activity, and reducing intake of HF foods are possible ways to lower the risk of both fatty pancreas and pancreatic cancer.

Obora et al. detected a “bright pancreas” in 776 subjects of 1,993 participants (38.6%) who were followed up for a mean of 8.7 years. This study found that the “bright pancreas” disappeared in approximately 30% of both sexes from baseline “bright pancreas” detection (detected at the follow-up) [123], indicating that “bright pancreas” is reversible and could be improved by changes of lifestyle. It is known that obesity-related FI of the pancreas could be improved after weight reduction [124].

Obesity and DM are associated with both FI and increased risk of pancreatic cancer. The use of metformin, an insulin-lowering agent for T2DM, has been shown to be associated with a decreased risk of pancreatic cancer in epidemiological studies [125, 126]. Metformin has also been shown to decrease the growth of PDAC in a xenograft model [127] and to lower the number of PDACs in BOP-treated hamsters with HFD [128]; however, there are currently no data on the effect of metformin on FI of the pancreas.

One anti-diabetic drug, pioglitazone, which is a ligand for PPARγ, improved hyperlipidemia and suppressed the incidence of pancreatic tumors in BOP-treated hamsters [129]. Another PPARγ ligand, rosiglitazone, also reduced fasting glucose and insulin levels in mice fed a HF plus highsucrose diet, however, these PPARγ ligands induced weight gain and remarkable FI of the intralobular space [113]. This finding seems to be inconsistent with the expected roles of FI of the pancreas in pancreatic carcinogenesis. Recently, it has been reported that pioglitazone use in people with diabetes increased their risk for developing pancreatic and prostate cancers [130], though the increased risk for pancreatic cancer was observed in short time users (≤ 12 months since initiation). Therefore, further studies are required to clarify the effects of pioglitazone on fatty pancreas and pancreatic cancer.

FI, especially NAFPD, is accompanied by inflammation in the pancreas. Clinical and animal experimental data show that COX-2 inhibitors have the potential to prevent pancreatic cancer prolongation [131, 132].

CONCLUSION

FI of the pancreas, which is associated with age, obesity and DM, is fundamentally caused by acinar cell damage. A K-ras mutation is essential for pancreatic cancer development in humans and in animal models. FI of the pancreas combined with K-ras mutation is strongly correlated with pancreatic cancer development in humans and animal models (Figure 1). Hamsters are likely to have a high sensitivity to pancreatic carcinogens, and their acinar cells can be easily damaged compared with those of mice and rats. The difference of such sensitivity may be explained in part by occurrence of severe FI of the pancreas. In human cases, severe FI of the pancreas is associated with PDAC. Severe FI of the pancreas presumes acinar cell damage, while proliferation- and inflammation-related factors secreted from adipocytes are considered to promote pancreatic carcinogenesis. Therefore, FI of the pancreas is a possible pancreatic risk factor and its modulation could be a useful method for preventing pancreatic cancer. Further prospective studies on these points are warranted.

Author contributions

Hori M, Mutoh M, Imai T, Nakagama H and Takahashi M solely contributed to this paper.

Support

National Cancer Center Research and Development Fund (21-2-1); a grant of the Research Grant of the Princess Takamatsu Cancer Research Fund; and a Grantin- Aid for Scientific Research from the Japan Society for the Promotion of Science (JSPS KAKENHI Grant number 25290049).

Conflict of Interest

The authors have no conflict of interest.

References

  1. Britton KA, Fox CS. Ectopic fat depots and cardiovascular disease. Circulation 2011; 124:e837-41. [PMID: 22156000]
  2. Angulo P. Nonalcoholic Fatty Liver Disease.N Engl J Med 2002; 346:1221-31. [PMID: 11961152]
  3. Scaglioni F, Ciccia S, Marino M, Bedogni G, Bellentani S. ASH and NASH.Dig Dis 2011; 29:202-10. [PMID: 21734385]
  4. Clark JM, Brancati FL, Diehl AM. Nonalcoholic fatty liver disease.Gastroenterology 2002; 122:1649-57. [PMID: 12016429]
  5. McCullough AJ. Pathophysiology of nonalcoholic steatohepatitis.J Clin Gastroenterol 2006; 40 Suppl 1:S17-29. [PMID: 16540762]
  6. Marks WM, Filly RA, Callen PW. Ultrasonic evaluation of normal pancreatic echogenicity and its relationship to fat deposition. Radiology 1980; 137:475-9. [PMID: 7433680]
  7. Nghiem DD, Olson PR, Ormond D. The "fatty pancreas allograft":anatomopathologic findings and clinical experience.Transplant Proc 2004; 36:1045-7. [PMID: 15194363]
  8. Tham RT, Heyerman HG, Falke TH, Zwinderman AH, Bloem JL, Bakker W, et al. Cystic fibrosis:MR imaging of the pancreas. Radiology 1991; 179:183-6. [PMID: 2006275]
  9. Walters MN. Adipose atrophy of the exocrine pancreas. J Pathol Bacteriol 1966; 92:547-57. [PMID: 5964381]
  10. Høyer A. Lipomatous pseudohypertrophy of the pancreas with complete absence of exocrine tissue. The Journal of Pathology and Bacteriology 1949; 61:93-100.
  11. Beresford OD, Owen TK. Lipomatous pseudohypertrophy of the pancreas. J Clin Pathol 1957; 10:63-6. [PMID: 13406076]
  12. Bralet MP, Terris B, Bregeaud L, Ruszniewski P, Bernades P, Belghiti J, et al. Squamous cell carcinoma and lipomatous pseudohypertrophy of the pancreas. Virchows Arch 1999; 434:569-72. [PMID: 10394894]
  13. Isserow JA, Siegelman ES, Mammone J. Focal fatty infiltration of the pancreas:MR characterization with chemical shift imaging. AJR Am J Roentgenol 1999; 173:1263-5. [PMID: 10541101]
  14. Lingvay I, Esser V, Legendre JL, Price AL, Wertz KM, Adams-Huet B, et al. Noninvasive quantification of pancreatic fat in humans. J Clin Endocrinol Metab 2009; 94:4070-6. [PMID: 19773401]
  15. Glaser J, Stienecker K. Pancreas and aging:a study using ultrasonography.Gerontology 2000; 46:93-6. [PMID: 10671806]
  16. Heuck A, Maubach PA, Reiser M, Feuerbach S, Allgayer B, Lukas P, et al. Age-related morphology of the normal pancreas on computed tomography. Gastrointest Radiol 1987; 12:18-22. [PMID: 3792751]
  17. Lee SE, Jang JY, Lim CS, Kang MJ, Kim SH, Kim MA, et al. Measurement of pancreatic fat by magnetic resonance imaging: predicting the occurrence of pancreatic fistula after pancreatoduodenectomy. Ann Surg 2010; 251:932-6. [PMID: 20395858]
  18. Kim MK, Chun HJ, Park JH, Yeo DM, Baek KH, Song KH, et al. The association between ectopic fat in the pancreas and subclinical atherosclerosis in type 2 diabetes. Diabetes Res Clin Pract 2014; 106:590-6. [PMID: 25444353]
  19. Kim SY, Kim H, Cho JY, Lim S, Cha K, Lee KH, et al. Quantitative Assessment of Pancreatic Fat by Using Unenhanced CT: Pathologic Correlation and Clinical Implications. Radiology 2014; 271:104-12. [PMID: 24475851]
  20. Lim S, Bae JH, Chun EJ, Kim H, Kim SY, Kim KM, et al. Differences in pancreatic volume, fat content, and fat density measured by multidetector-row computed tomography according to the duration of diabetes. Acta Diabetol 2014; 51:739-48. [PMID: 24671510]
  21. Kovanlikaya A, Mittelman SD, Ward A, Geffner ME, Dorey F, Gilsanz V. Obesity and fat quantification in lean tissues using three-point Dixon MR imaging. Pediatr Radiol 2005; 35:601-7. [PMID: 15785930]
  22. Lee JS, Kim SH, Jun DW, Han JH, Jang EC, Park JY, et al. Clinical implications of fatty pancreas:correlations between fatty pancreas and metabolic syndrome. World J Gastroenterol 2009; 15:1869-75. [PMID: 19370785]
  23. Wu WC, Wang CY. Association between non-alcoholic fatty pancreatic disease (NAFPD) and the metabolic syndrome: case-control retrospective study. Cardiovasc Diabetol 2013; 12:77. [PMID: 23688357]
  24. Rosso E, Casnedi S, Pessaux P, Oussoultzoglou E, Panaro F, Mahfud M, et al. The role of "fatty pancreas" and of BMI in the occurrence of pancreatic fistula after pancreaticoduodenectomy. J Gastrointest Surg 2009; 13:1845-51. [PMID: 19639369]
  25. Choi CW, Kim GH, Kang DH, Kim HW, Kim DU, Heo J, et al. Associated factors for a hyperechogenic pancreas on endoscopic ultrasound. World J Gastroenterol 2010; 16:4329-34. [PMID: 208188 17]
  26. Al-Haddad M, Khashab M, Zyromski N, Pungpapong S, Wallace MB, Scolapio J, et al. Risk factors for hyperechogenic pancreas on endoscopic ultrasound: a case-control study. Pancreas 2009; 38:672-5. [PMID: 19506531]
  27. Sepe PS, Ohri A, Sanaka S, Berzin TM, Sekhon S, Bennett G, et al. A prospective evaluation of fatty pancreas by using EUS. Gastrointest Endosc 2011; 73:987-93. [PMID: 21521567]
  28. Olsen TS. Lipomatosis of the pancreas in autopsy material and its relation to age and overweight. Acta Pathol Microbiol Scand A 1978; 86A:367-73. [PMID: 716899]
  29. van der Zijl NJ, Goossens GH, Moors CC, van Raalte DH, Muskiet MH, Pouwels PJ, et al. Ectopic fat storage in the pancreas, liver, and abdominal fat depots: impact on beta-cell function in individuals with impaired glucose metabolism. J Clin Endocrinol Metab 2011; 96:459-67. [PMID: 21084401]
  30. Tushuizen ME, Bunck MC, Pouwels PJ, Bontemps S, van Waesberghe JH, Schindhelm RK, et al. Pancreatic fat content and beta-cell function in men with and without type 2 diabetes. Diabetes Care 2007; 30:2916-21. [PMID: 17666465]
  31. Heni M, Machann J, Staiger H, Schwenzer NF, Peter A, Schick F, et al. Pancreatic fat is negatively associated with insulin secretion in individuals with impaired fasting glucose and/or impaired glucose tolerance: a nuclear magnetic resonance study. Diabetes Metab Res Rev 2010; 26:200-5. [PMID: 20225188]
  32. Maeda H. [Ductal and vascular factors in the etiology of experimentally induced pancreatic fibrosis and fat replacement in dogs] Nihon Shokakibyo Gakkai Zasshi 1986; 83:2580-7. [PMID: 3560492]
  33. Sarwar N, Gao P, Seshasai SR, Gobin R, Kaptoge S, Di Angelantonio E, et al. Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease:a collaborative meta-analysis of 102 prospective studies. Lancet 2010; 375:2215-22. [PMID: 20609967]
  34. Stein RA, Rockman CB, Guo Y, Adelman MA, Riles T, Hiatt WR, et al. Association between physical activity and peripheral artery disease and carotid artery stenosis in a self-referred population of 3 million adults.Arterioscler Thromb Vasc Biol 2015; 35:206-12. [PMID: 25359858]
  35. Smits MM, van Geenen EJ. The clinical significance of pancreatic steatosis. Nat Rev Gastroenterol Hepatol 2011; 8:169-77. [PMID: 21304475]
  36. Watanabe S, Abe K, Anbo Y, Katoh H. Changes in the mouse exocrine pancreas after pancreatic duct ligation: a qualitative and quantitative histological study. Arch Histol Cytol 1995; 58:365-74. [PMID: 8527243]
  37. Uchida T, Tsuchiya R, Harada N, Tsunoda T, Yamaguchi T, Eto T, et al. Ischemic changes in the pancreas of Watanabe heritable hyper-lipidemic (WHHL) rabbits. Int J Pancreatol 1988; 3:261-71. [PMID: 3385220]
  38. Golson ML, Loomes KM, Oakey R, Kaestner KH. Ductal malformation and pancreatitis in mice caused by conditional Jag1 deletion. Gastroenterology 2009; 136:1761-71.e1. [PMID: 19208348]
  39. Wilschanski M, Novak I. The cystic fibrosis of exocrine pancreas. Cold Spring Harb Perspect Med 2013; 3:a009746. [PMID: 23637307]
  40. Toiviainen-Salo S, Raade M, Durie PR, Ip W, Marttinen E, Savilahti E, et al. Magnetic resonance imaging findings of the pancreas in patients with Shwachman-Diamond syndrome and mutations in the SBDS gene. J Pediatr 2008; 152:434-6. [PMID: 18280855]
  41. Alkhouri N, Kaplan B, Kay M, Shealy A, Crowe C, Bauhuber S, et al. Johanson-Blizzard syndrome with mild phenotypic features confirmed by UBR1 gene testing. World J Gastroenterol 2008; 14:6863-6. [PMID: 19058315]
  42. Raeder H, Haldorsen IS, Ersland L, Gruner R, Taxt T, Sovik O, et al. Pancreatic lipomatosis is a structural marker in nondiabetic children with mutations in carboxyl-ester lipase. Diabetes 2007; 56:444-9. [PMID: 17259390]
  43. Lu Y, Loos RJ. Obesity genomics: assessing the transferability of susceptibility loci across diverse populations. Genome Med 2013; 5:55. [PMID: 23806069]
  44. Xi B, Chandak GR, Shen Y, Wang Q, Zhou D. Association between common polymorphism near the MC4R gene and obesity risk: a systematic review and meta-analysis. PLoS One 2012; 7:e45731. [PMID: 23049848]
  45. Xi B, Takeuchi F, Chandak GR, Kato N, Pan HW, Zhou DH, et al. Common polymorphism near the MC4R gene is associated with type 2 diabetes:data from a meta-analysis of 123,373 individuals. Diabetologia 2012; 55:2660-6. [PMID: 22869321]
  46. da Cunha PA, de Carlos Back LK, Sereia AF, Kubelka C, Ribeiro MC, Fernandes BL, et al. Interaction between obesity-related genes, FTO and MC4R, associated to an increase of breast cancer risk. Mol Biol Rep 2013; 40:6657-64. [PMID: 24091943]
  47. Lim U, Wilkens LR, Monroe KR, Caberto C, Tiirikainen M, Cheng I, et al. Susceptibility variants for obesity and colorectal cancer risk: the multiethnic cohort and PAGE studies. Int J Cancer 2012; 131:E1038-43. [PMID: 22511254]
  48. Delahanty RJ, Beeghly-Fadiel A, Xiang YB, Long J, Cai Q, Wen W, et al. Association of obesity-related genetic variants with endometrial cancer risk: a report from the Shanghai Endometrial Cancer Genetics Study. Am J Epidemiol 2011; 174:1115-26. [PMID: 21976109]
  49. Itoh M, Suganami T, Nakagawa N, Tanaka M, Yamamoto Y, Kamei Y, et al. Melanocortin 4 receptor-deficient mice as a novel mouse model of nonalcoholic steatohepatitis. Am J Pathol 2011; 179:2454-63. [PMID: 21906580]
  50. Krude H, Biebermann H, Luck W, Horn R, Brabant G, Gruters A. Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nat Genet 1998; 19:155-7. [PMID: 9620771]
  51. Lee YS, Challis BG, Thompson DA, Yeo GS, Keogh JM, Madonna ME, et al. A POMC variant implicates beta-melanocyte-stimulating hormone in the control of human energy balance. Cell Metab 2006; 3:135-40. [PMID: 16459314]
  52. Frayling TM, Timpson NJ, Weedon MN, Zeggini E, Freathy RM, Lindgren CM, et al. A common variant in the FTO gene is associated with body mass index and predisposes to childhood and adult obesity. Science 2007; 316:889-94. [PMID: 17434869]
  53. Romeo S, Kozlitina J, Xing C, Pertsemlidis A, Cox D, Pennacchio LA, et al. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nat Genet 2008; 40:1461-5. [PMID: 18820647]
  54. Santoro N, Zhang CK, Zhao H, Pakstis AJ, Kim G, Kursawe R, et al. Variant in the glucokinase regulatory protein (GCKR) gene is associated with fatty liver in obese children and adolescents. Hepatology 2012; 55:781-9. [PMID: 22105854]
  55. Ou HY, Wang CY, Yang YC, Chen MF, Chang CJ. The association between nonalcoholic fatty pancreas disease and diabetes. PLoS One 2013; 8:e62561. [PMID: 23671610]
  56. Michaud DS. Epidemiology of pancreatic cancer. Minerva Chir 2004; 59:99-111. [PMID: 15238885]
  57. Lin Y, Tamakoshi A, Wakai K, Kawamura T, Aoki R, Kojima M, et al. Descriptive epidemiology of pancreatic cancer in Japan. J Epidemiol 1998; 8:52-9. [PMID: 9575696]
  58. Yoshiike N, Matsumura Y, Iwaya M, Sugiyama M, Yamaguchi M. National Nutrition Survey in Japan. J Epidemiol 1996; 6:S189-200. [PMID: 8800293]
  59. Li D, Morris JS, Liu J, Hassan MM, Day RS, Bondy ML, et al. Body mass index and risk, age of onset, and survival in patients with pancreatic cancer. Jama 2009; 301:2553-62. [PMID: 19549972]
  60. Patel AV, Rodriguez C, Bernstein L, Chao A, Thun MJ, Calle EE. Obesity, recreational physical activity, and risk of pancreatic cancer in a large U.S. Cohort.Cancer Epidemiol Biomarkers Prev 2005; 14:459-66. [PMID: 15734973]
  61. Luo J, Iwasaki M, Inoue M, Sasazuki S, Otani T, Ye W, et al. Body mass index, physical activity and the risk of pancreatic cancer in relation to smoking status and history of diabetes: a large-scale population-based cohort study in Japan--the JPHC study. Cancer Causes Control 2007; 18:603-12. [PMID: 17401636]
  62. Huxley R, Ansary-Moghaddam A, Berrington de Gonzalez A, Barzi F, Woodward M. Type-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer 2005; 92:2076-83. [PMID: 15886696]
  63. Schenk M, Schwartz AG, O'Neal E, Kinnard M, Greenson JK, Fryzek JP, et al. Familial risk of pancreatic cancer. J Natl Cancer Inst 2001; 93:640-4. [PMID: 11309441]
  64. Lin Y, Tamakoshi A, Kawamura T, Inaba Y, Kikuchi S, Motohashi Y, et al. A prospective cohort study of cigarette smoking and pancreatic cancer in Japan. Cancer Causes & Control 2002; 13:249-54.
  65. Lowenfels AB, Maisonneuve P, Cavallini G, Ammann RW, Lankisch PG, Andersen JR, et al. Pancreatitis and the risk of pancreatic cancer. International Pancreatitis Study Group. N Engl J Med 1993; 328:1433-7. [PMID: 8479461]
  66. Risch HA, Yu H, Lu L, Kidd MS. Detectable Symptomatology Preceding the Diagnosis of Pancreatic Cancer and Absolute Risk of Pancreatic Cancer Diagnosis. Am J Epidemiol 2015 [PMID: 26049860]
  67. Schuller HM. Mechanisms of smoking-related lung and pancreatic adenocarcinoma development. Nat Rev Cancer 2002; 2:455-63. [PMID: 12189387]
  68. Belinsky SA, Devereux TR, Maronpot RR, Stoner GD, Anderson MW. Relationship between the formation of promutagenic adducts and the activation of the K-ras protooncogene in lung tumors from A/J mice treated with nitrosamines. Cancer Res 1989; 49:5305-11. [PMID: 2670201]
  69. Belinsky SA, Foley JF, White CM, Anderson MW, Maronpot RR. Dose-response relationship between O6-methylguanine formation in Clara cells and induction of pulmonary neoplasia in the rat by 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone. Cancer Res 1990; 50:3772-80. [PMID: 2340522]
  70. Westra WH, Slebos RJ, Offerhaus GJ, Goodman SN, Evers SG, Kensler TW, et al. K-ras oncogene activation in lung adenocarcinomas from former smokers. Evidence that K-ras mutations are an early and irreversible event in the development of adenocarcinoma of the lung. Cancer 1993; 72:432-8. [PMID: 8319174]
  71. Smit VT, Boot AJ, Smits AM, Fleuren GJ, Cornelisse CJ, Bos JL. KRAS codon 12 mutations occur very frequently in pancreatic adenocarcinomas. Nucleic Acids Res 1988; 16:7773-82. [PMID: 3047672]
  72. Kanda M, Matthaei H, Wu J, Hong SM, Yu J, Borges M, et al. Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology 2012; 142:730-3 e9. [PMID: 22226782]
  73. Kamisawa T, Tsuruta K, Okamoto A, Horiguchi S, Hayashi Y, Yun X, et al. Frequent and significant K-ras mutation in the pancreas, the bile duct, and the gallbladder in autoimmune pancreatitis. Pancreas 2009; 38:890-5. [PMID: 19752775]
  74. Yanagisawa A, Ohtake K, Ohashi K, Hori M, Kitagawa T, Sugano H, et al. Frequent c-Ki-ras oncogene activation in mucous cell hyperplasias of pancreas suffering from chronic inflammation. Cancer Res 1993; 53:953-6. [PMID: 8439969]
  75. Popovic Hadzija M, Korolija M, Jakic Razumovic J, Pavkovic P, Hadzija M, Kapitanovic S. K-ras and Dpc4 mutations in chronic pancreatitis: case series. Croat Med J 2007; 48:218-24. [PMID: 17436386]
  76. Blackford A, Parmigiani G, Kensler TW, Wolfgang C, Jones S, Zhang X, et al. Genetic mutations associated with cigarette smoking in pancreatic cancer. Cancer Res 2009; 69:3681-8. [PMID: 19351817]
  77. Jiao L, Zhu J, Hassan MM, Evans DB, Abbruzzese JL, Li D. K-ras mutation and p16 and preproenkephalin promoter hypermethylation in plasma DNA of pancreatic cancer patients:in relation to cigarette smoking. Pancreas 2007; 34:55-62. [PMID: 17198183]
  78. Hori M, Onaya H, Takahashi M, Hiraoka N, Mutoh M, Kosuge T, et al. Invasive ductal carcinoma developing in pancreas with severe Fatty infiltration. Pancreas 2012; 41:1137-9. [PMID: 22940831]
  79. Hori M, Takahashi M, Hiraoka N, Yamaji T, Mutoh M, Ishigamori R, et al. Association of pancreatic Fatty infiltration with pancreatic ductal adenocarcinoma. Clin Transl Gastroenterol 2014; 5:e53. [PMID: 24622469]
  80. Rebours V, Gaujoux S, d'Assignies G, Sauvanet A, Ruszniewski P, Levy P, et al. Obesity and fatty pancreatic infiltration are risk factors for pancreatic precancerous lesions (PanIN). Clin Cancer Res 2015 [PMID: 25700304]
  81. Guerra C, Schuhmacher AJ, Canamero M, Grippo PJ, Verdaguer L, Perez-Gallego L, et al. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell 2007; 11:291-302. [PMID: 17349585]
  82. Guerra C, Collado M, Navas C, Schuhmacher AJ, Hernandez-Porras I, Canamero M, et al. Pancreatitis-induced inflammation contributes to pancreatic cancer by inhibiting oncogene-induced senescence. Cancer Cell 2011; 19:728-39. [PMID: 21665147]
  83. Pour P, Althoff J, Kruger FW, Mohr U. A potent pancreatic carcinogen in Syrian hamsters:N-nitrosobis(2-oxopropyl)amine. J Natl Cancer Inst 1977; 58:1449-53. [PMID: 857032]
  84. Sorio C, Baron A, Orlandini S, Zamboni G, Pederzoli P, Huebner K, et al. The FHIT gene is expressed in pancreatic ductular cells and is altered in pancreatic cancers. Cancer Res 1999; 59:1308-14. [PMID: 10096564]
  85. Grünewald K, Lyons J, Fröhlich A, Feichtinger H, Weger RA, Schwab G, et al. High frequency of Ki-ras codon 12 mutations in pancreatic adenocarcinomas. International Journal of Cancer 1989; 43:1037-41.
  86. Tsujiuchi T, Sasaki Y, Kubozoe T, Konishi Y, Tsutsumi M. Alterations in the Fhit gene in pancreatic duct adenocarcinomas induced by N-nitrosobis(2-oxopropyl)amine in hamsters. Mol Carcinog 2003; 36:60-6. [PMID: 12557261]
  87. Caldas C, Hahn SA, da Costa LT, Redston MS, Schutte M, Seymour AB, et al. Frequent somatic mutations and homozygous deletions of the p16 (MTS1) gene in pancreatic adenocarcinoma.Nat Genet1994; 8:27-32. [PMID: 7726912]
  88. Hanaoka M, Shimizu K, Shigemura M, Kato A, Fujii H, Honoki K, et al. Cloning of the hamster p16 gene 5' upstream region and its aberrant methylation patterns in pancreatic cancer. Biochem Biophys Res Commun 2005; 333:1249-53. [PMID: 15979573]
  89. Hori M, Kitahashi T, Imai T, Ishigamori R, Takasu S, Mutoh M, et al. Enhancement of carcinogenesis and fatty infiltration in the pancreas in N-nitrosobis (2-oxopropyl) amine-treated hamsters by high-fat diet. Pancreas 2011; 40:1234-40. [PMID: 21989024]
  90. Fujii K, Hayakawa T, Kikuchi M. Tumor induction in mice administered neonatally with bis(2-oxopropyl)nitrosamine. Tohoku J Exp Med 1994; 174:361-8. [PMID: 7732518]
  91. Sakano K, Takahashi M, Mutoh M, Niho N, Komiya M, Sato H, et al. Enhanced thyroid carcinogenicity of N-nitrosobis(2-oxopropyl)amine in Otsuka Long-Evans Tokushima Fatty rats, a model of type II diabetes mellitus. Carcinogenesis 2007; 28:2193-8. [PMID: 17510084]
  92. Colvin EK, Scarlett CJ. A historical perspective of pancreatic cancer mouse models. Semin Cell Dev Biol 2014; 27:96-105. [PMID: 24685616]
  93. Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, Jacobetz MA, et al. Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell 2003; 4:437-50. [PMID: 14706336]
  94. Grippo PJ, Nowlin PS, Demeure MJ, Longnecker DS, Sandgren EP. Preinvasive pancreatic neoplasia of ductal phenotype induced by acinar cell targeting of mutant Kras in transgenic mice. Cancer Res 2003; 63:2016-9. [PMID: 12727811]
  95. Aguirre AJ, Bardeesy N, Sinha M, Lopez L, Tuveson DA, Horner J, et al. Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic pancreatic ductal adenocarcinoma. Genes Dev 2003; 17:3112-26. [PMID: 14681207]
  96. Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell 2005; 7:469-83. [PMID: 15894267]
  97. Kojima K, Vickers SM, Adsay NV, Jhala NC, Kim HG, Schoeb TR, et al. Inactivation of Smad4 accelerates Kras(G12D)-mediated pancreatic neoplasia. Cancer Res 2007; 67:8121-30. [PMID: 17804724]
  98. Izeradjene K, Combs C, Best M, Gopinathan A, Wagner A, Grady WM, et al. Kras(G12D) and Smad4/Dpc4 haploinsufficiency cooperate to induce mucinous cystic neoplasms and invasive adenocarcinoma of the pancreas. Cancer Cell 2007; 11:229-43. [PMID: 17349581]
  99. Tombran-Tink J, Barnstable CJ. PEDF:a multifaceted neurotrophic factor. Nat Rev Neurosci 2003; 4:628-36. [PMID: 12894238]
  100. Becerra SP, Notario V. The effects of PEDF on cancer biology:mechanisms of action and therapeutic potential. Nat Rev Cancer 2013; 13:258-71. [PMID: 23486238]
  101. Grippo PJ, Fitchev PS, Bentrem DJ, Melstrom LG, Dangi-Garimella S, Krantz SB, et al. Concurrent PEDF deficiency and Kras mutation induce invasive pancreatic cancer and adipose-rich stroma in mice. Gut 2012; 61:1454-64. [PMID: 22234980]
  102. Benito M, Porras A, Nebreda AR, Santos E. Differentiation of 3T3-L1 fibroblasts to adipocytes induced by transfection of ras oncogenes. Science 1991; 253:565-8. [PMID: 1857988]
  103. Wang M, Wang JJ, Li J, Park K, Qian X, Ma JX, et al. Pigment epithelium-derived factor suppresses adipogenesis via inhibition of the MAPK/ERK pathway in 3T3-L1 preadipocytes. sAm J Physiol Endocrinol Metab 2009; 297:E1378-87. [PMID: 19808909]
  104. Dawson DW, Hertzer K, Moro A, Donald G, Chang HH, Go VL, et al. High-fat, high-calorie diet promotes early pancreatic neoplasia in the conditional KrasG12D mouse model. Cancer Prev Res (Phila) 2013; 6:1064-73. [PMID: 23943783]
  105. Iniguez MA, Rodriguez A, Volpert OV, Fresno M, Redondo JM. Cyclooxygenase-2:a therapeutic target in angiogenesis. Trends Mol Med 2003; 9:73-8. [PMID: 12615041]
  106. Kuwano T, Nakao S, Yamamoto H, Tsuneyoshi M, Yamamoto T, Kuwano M, et al. Cyclooxygenase 2 is a key enzyme for inflammatory cytokine-induced angiogenesis. FASEB J 2004; 18:300-10. [PMID: 14769824]
  107. Fajas L, Miard S, Briggs MR, Auwerx J. Selective cyclo-oxygenase-2 inhibitors impair adipocyte differentiation through inhibition of the clonal expansion phase. J Lipid Res 2003; 44:1652-9. [PMID: 12837847]
  108. Araki Y, Okamura S, Hussain SP, Nagashima M, He P, Shiseki M, et al. Regulation of cyclooxygenase-2 expression by the Wnt and ras pathways. Cancer Res 2003; 63:728-34. [PMID: 12566320]
  109. Sheng H, Shao J, Dubois RN. K-Ras-mediated increase in cyclooxygenase 2 mRNA stability involves activation of the protein kinase B1. Cancer Res 2001; 61:2670-5. [PMID: 11289146]
  110. Daniluk J, Liu Y, Deng D, Chu J, Huang H, Gaiser S, et al. An NF-kappaB pathway-mediated positive feedback loop amplifies Ras activity to pathological levels in mice. J Clin Invest 2012; 122:1519-28. [PMID: 22406536]
  111. Mendonsa AM, Chalfant MC, Gorden LD, VanSaun MN. Modulation of the leptin receptor mediates tumor growth and migration of pancreatic cancer cells. PLoS One 2015; 10:e0126686. [PMID: 25919692]
  112. Fraulob JC, Ogg-Diamantino R, Fernandes-Santos C, Aguila MB, Mandarim-de-Lacerda CA. A Mouse Model of Metabolic Syndrome: Insulin Resistance, Fatty Liver and Non-Alcoholic Fatty Pancreas Disease (NAFPD) in C57BL/6 Mice Fed a High Fat Diet. J Clin Biochem Nutr 2010; 46:212-23. [PMID: 20490316]
  113. Fernandes-Santos C, Evangelista Carneiro R, de Souza Mendonca L, Barbosa Aguila M, Mandarim-de-Lacerda CA. Rosiglitazone aggravates nonalcoholic Fatty pancreatic disease in C57BL/6 mice fed high-fat and high-sucrose diet. Pancreas 2009; 38:e80-6. [PMID: 19214135]
  114. Zyromski NJ, Mathur A, Pitt HA, Wade TE, Wang S, Nakshatri P, et al. Obesity potentiates the growth and dissemination of pancreatic cancer. Surgery 2009; 146:258-63. [PMID: 19628082]
  115. Morita Y, Yoshikawa T, Takeda S, Matsuyama K, Takahashi S, Yoshida N, et al. Involvement of lipid peroxidation in free fatty acid-induced isolated rat pancreatic acinar cell injury. Pancreas 1998; 17:383-9. [PMID: 9821180]
  116. Mylonas C, Kouretas D. Lipid peroxidation and tissue damage. In Vivo 1999; 13:295-309. [PMID: 10459507]
  117. Spooner PM, Chernick SS, Garrison MM, Scow RO. Development of lipoprotein lipase activity and accumulation of triacylglycerol in differentiating 3T3-L1 adipocytes. Effects of prostaglandin F2alpha, 1-methyl-3-isobutylxanthine, prolactin, and insulin. J Biol Chem 1979; 254:1305-11. [PMID: 83996]
  118. Ma J, Sawai H, Matsuo Y, Ochi N, Yasuda A, Takahashi H, et al. IGF-1 mediates PTEN suppression and enhances cell invasion and proliferation via activation of the IGF-1/PI3K/Akt signaling pathway in pancreatic cancer cells. J Surg Res 2010; 160:90-101. [PMID: 19560785]
  119. Lashinger LM, Harrison LM, Rasmussen AJ, Logsdon CD, Fischer SM, McArthur MJ, et al. Dietary energy balance modulation of Kras- and Ink4a/Arf+/--driven pancreatic cancer:the role of insulin-like growth factor-I. Cancer Prev Res (Phila) 2013; 6:1046-55. [PMID: 23980075]
  120. Silha JV, Krsek M, Sucharda P, Murphy LJ. Angiogenic factors are elevated in overweight and obese individuals. Int J Obes (Lond) 2005; 29:1308-14. [PMID: 15953938]
  121. Huang C, Huang R, Chang W, Jiang T, Huang K, Cao J, et al. The expression and clinical significance of pSTAT3, VEGF and VEGF-C in pancreatic adenocarcinoma. Neoplasma 2012; 59:52-61. [PMID: 22082308]
  122. Zhang B, Zhao W-H, Zhou W-Y, Yu W-S, Yu J-M, Li S. Expression of vascular endothelial growth factors-C and -D correlate with evidence of lymphangiogenesis and angiogenesis in pancreatic adenocarcinoma.Cancer Detection and Prevention 2007; 31:436-42.
  123. Obora A, Kojima T, Kato T, Endo M, Miyawaki K, Fukuta N, et al. Associations between Bright Pancreas and Features of Metabolic Syndrome. Ningen Dock 2012; 26:935-43.
  124. Rossi AP, Fantin F, Zamboni GA, Mazzali G, Zoico E, Bambace C, et al. Effect of moderate weight loss on hepatic, pancreatic and visceral lipids in obese subjects. Nutr Diabetes 2012; 2:e32. [PMID: 23449531]
  125. Decensi A, Puntoni M, Goodwin P, Cazzaniga M, Gennari A, Bonanni B, et al. Metformin and cancer risk in diabetic patients: a systematic review and meta-analysis. Cancer Prev Res (Phila) 2010; 3:1451-61. [PMID: 20947488]
  126. Wang Z, Lai ST, Xie L, Zhao JD, Ma NY, Zhu J, et al. Metformin is associated with reduced risk of pancreatic cancer in patients with type 2 diabetes mellitus: a systematic review and meta-analysis. Diabetes Res Clin Pract 2014; 106:19-26. [PMID: 24837144]
  127. Kisfalvi K, Eibl G, Sinnett-Smith J, Rozengurt E. Metformin disrupts crosstalk between G protein-coupled receptor and insulin receptor signaling systems and inhibits pancreatic cancer growth. Cancer Res 2009; 69:6539-45. [PMID: 19679549]
  128. Schneider MB, Matsuzaki H, Haorah J, Ulrich A, Standop J, Ding XZ, et al. Prevention of pancreatic cancer induction in hamsters by metformin. Gastroenterology 2001; 120:1263-70. [PMID: 11266389]
  129. Takeuchi Y, Takahashi M, Sakano K, Mutoh M, Niho N, Yamamoto M, et al. Suppression of N-nitrosobis(2-oxopropyl)amine-induced pancreatic carcinogenesis in hamsters by pioglitazone, a ligand of peroxisome proliferator-activated receptor gamma. Carcinogenesis 2007; 28:1692-6. [PMID: 17449904]
  130. Lewis JD, Habel LA, Quesenberry CP, Strom BL, Peng T, Hedderson MM, et al. Pioglitazone Use and Risk of Bladder Cancer and Other Common Cancers in Persons With Diabetes. Jama 2015; 314:265-77. [PMID: 26197187]
  131. Furukawa F, Nishikawa A, Lee IS, Kanki K, Umemura T, Okazaki K, et al. A cyclooxygenase-2 inhibitor, nimesulide, inhibits postinitiation phase of N-nitrosobis (2-oxopropyl) amine-induced pancreatic carcinogenesis in hamsters.Int J Cancer 2003; 104:269-73. [PMID: 12569549]
  132. Kokawa A, Kondo H, Gotoda T, Ono H, Saito D, Nakadaira S, et al. Increased expression of cyclooxygenase-2 in human pancreatic neoplasms and potential for chemoprevention by cyclooxygenase inhibitors. Cancer 2001; 91:333-8. [PMID: 11180079]