Abstract
The incidence of obesity and type 2 diabetes (T2DM) in the Western world has increased dramatically during the recent decades. According to the American Cancer Society, pancreatic cancer (PC) is the fourth leading cause of cancer‐related death in the United States. The relationship among obesity, T2DM and PC is complex. Due to increase in obesity, diabetes, alcohol consumption and sedentary lifestyle, the mortality due to PC is expected to rise significantly by year 2040. The underlying mechanisms by which diabetes and obesity contribute to pancreatic tumorigenesis are not well understood. Furthermore, metabolism and microenvironment within the pancreas can also modulate pancreatic carcinogenesis. The risk of PC on a population level may be reduced by modifiable lifestyle risk factors. In this review, the interactions of diabetes and obesity to PC development were summarized, and novel strategies for the prevention and treatment of diabetes and PC were discussed.
Keywords: alcoholism, diabetes mellitus, obesity, pancreatic cancers, prevention
1. INTRODUCTION
Pancreatic cancer (PC) is one of the ten most common cancers in human. Most of the cases are pancreatic exocrine cancer, only 1%‐2% of cases of PC are neuroendocrine tumours. According to the American Cancer Society, the incidence of PC was 53 770 in 2019, with a concomitant mortality of 45 750 (23 800 men and 21 950 women). 1 It is the fourth cause of cancer‐related death in both men and women in the United States each year. 1 In the United States, the number of new cases of PC was 12.4 per 100 000 men and women per year based on 2009‐2013 cases. In spite of massive effort on diagnosis and treatment, the 5‐year survival rate has been increased to a mere 8%. 1 By 2030, the number of deaths from PC will surpass breast, prostate and colon cancer and become the second leading cause of cancer‐related death in the United States. 2 Due to unclear symptoms and no screening recommendations, a vast majority of PC patients are diagnosed at late stages, with already advanced disease and no opportunity for surgical intervention. The risk factors for PC include tobacco products, obesity, diabetes, chronic pancreatitis, alcohol abuse, malnutrition, hereditary conditions and family history (Figure 1). 3 , 4 Diabetes mellitus (DM), or impaired glucose tolerance, is concurrently present in 50%‐80% of patients with PC. DM is a known risk factor for PC, 5 , 6 and new‐onset DM could be an early manifestation of PC, 7 resulting from insulin resistance induced by a paraneoplastic syndrome 8 or pancreatic β‐cell dysfunction. 9 , 10 In addition, it has been demonstrated that moderate alcohol consumption had insignificant impact, while high alcohol intake was associated with an increased risk of PC. 11 , 12 Although the effects of DM and alcohol abuse on the development of PC have been studied for the last few decades, their molecular mechanisms of action are not well understood. We conducted this review to update and summarize the mechanisms of association among diabetes mellitus, obesity, alcoholism, other factors and cancerous pancreas. In addition, prevention and treatment strategies are also critically discussed in this review paper.
Figure 1.

Schema of risk factors of pancreatic cancer. Pancreatic cancer (PC) can be induced by several factors. Smoking is known as the strong carcinogen for PC. Obesity and T2DM synergistically act to induce the development of PC. Other factors such as alcohol abuse, pancreatitis, nutrition, genetics, family history and chemical carcinogen can contribute to the development of PC
2. DIABETES MELLITUS (DM), A RISK FACTOR OF PANCREATIC CANCER
There are three major types of diabetes mellitus (DM): type 1, type 2 and gestational diabetes. Type 1 diabetes (T1DM) may account for about 5% of all cases of diabetes. Type 2 diabetes account for about 90% to 95% of diabetes. In obese individuals with euglycaemia, peripheral insulin resistance is present but compensated by increased insulin secretion. 13 , 14 Increase in insulin resistance and β‐cell dysfunction, and reduction in β‐cell mass occur over time, finally leading to T2DM. 13 , 14 Unlike T2DM, studies conducted on small on number of patients have not shown that T1DM is risk factor of PC. 15 , 16 Patients with T2DM possess a threefold risk of developing PC. 17 Consistently, duration of the diabetes increases the risk of developing PC in a prospective study with hazard risk 2.0 in both men and women. 18 Although HbA1C is used as a marker of metabolic control, some studies have suggested its use as a predictor and prognostic factor in PC. Moreover, hyperglycaemia in diabetic patients promotes the growth of solid tumours and metastasis to distant organs in cancers. 19
3. POTENTIAL MECHANISMS OF DIABETES ON THE DEVELOPMENT OF PANCREATIC CANCER
The pathogenesis of PC in DM or hyperglycaemia has been substantially studied (Figure 2). High glucose‐activated p38 MAPK induced the proliferation and invasion of PC cells. 20 In response to cellular stress and inflammatory conditions, P38 MAPK is activated. 21 In addition, the proliferation, apoptosis and metastasis can also be regulated by the p38 MAPK. 22 , 23 P38 MAPK enhanced inflammatory cytokine (IL‐6)‐ and VEGF‐mediated paracrine effects, resulting in PC cell growth and development. 24 Moreover, proliferation and invasiveness in PC cells were promoted by high glucose via RET [proto‐oncogene encodes a receptor tyrosine kinase for members of the glial cell line‐derived neurotrophic factor (GDNF) family of extracellular signalling molecules]. 25 DM stimulates PC growth, epithelial‐mesenchymal transition and metastasis through generation of inflammatory cytokines (IL‐6 and TNFα) and activation of kinases (p38 MAPK and NFκB). 20 NFкB is not only activated in cancer cells, but also in immune cells where it regulates the production of inflammatory cytokines (IL‐6, IL‐8, IL‐1β and TNFα) triggering PC cell growth.
Figure 2.

Pathogenesis of pancreatic cancer. Pancreatic cancer can be initiated and accelerated by several factors. Hyperglycaemia with or without insulin resistance is a risk factors of PC. Inflammation, oxidative stress and certain immune cells can initiate the development of PC. Angiogenesis can enhance the development of PC
In type 2 DM (T2DM), elevated insulin levels result in increased bioavailability of insulin‐like growth factor‐1 (IGF‐1) by reducing hepatic production of IGF‐binding proteins (IGFBPs) and PC cells highly express receptors of high‐affinity insulin and IGF‐1. 26 , 27 The interaction of insulin, IGF‐1 and their receptors plausibly induces more activated PC cells. Insulin regulates glucose uptakes in target tissues and also acts as a mitogen for PC cells. 28 , 29 IGF‐1 besides its mitogenic effects, induces angiogenesis and increases EMT and metastasis, and blocks apoptosis, thereby enhancing PC growth. 30 Higher insulin and IGF‐1 levels in T2DM patients were associated with larger pancreas tumours than in non‐diabetic controls. 31 , 32 The prevalence of PC in T1DM is lower than in T2DM. It is not clear whether hyperglycaemia alone or the combination of hyperglycaemia with insulin resistance in T2DM act as an initiating factor for PC. Hyperglycaemia has been shown to accelerate the development of PC by providing glucose as a fuel to cancer cells. Some other studies have indicated hyperglycaemia enhanced proliferation via the induction of EGF/EGFR, 33 local invasiveness and metastatic potential in PC via neural and perineural environment. 34 Hyperglycaemia has been shown to induce endothelial dysfunction and neo‐angiogenesis, 35 resulting in endothelial discontinuities, large blood‐filled spaces and decreased vessel density via angiopoietin‐2. 36
In another aspect, receptors of advanced glycation end products (RAGE), another cancer‐related factor, are constitutively expressed on the epithelial, immune, endothelial and vascular smooth muscle cells. 37 There is still a paradox about the role of AGE/RAGE in cancerous pancreas. The RAGE has been shown to enhance pancreatic carcinogenesis by amplifying IL‐6‐induced autophagic translocation of STAT3 to the mitochondria in mice. 38 However, human studies failed to show any association between AGE/RAGE and PC risk. 39 This controversy is probably explained by the fact that AGE/RAGE could be only functional in the earlier stages of pancreatic carcinogenesis; and smoking, another risk factor, in the latter stages could affect to the role of AGE/RAGE in tumorigenesis. Alternatively, the disparity is simply due to the difference between human and mouse models. However, other studies showed a positive correlationship between the AGE/RAGE interaction and development of pancreatic and gastric cancers, and melanoma. 40 , 41 , 42 AGE/RAGE induces pro‐inflammatory cytokines and generates oxidative stress and reactive oxygen species, resulting in activation of NFкB and its target genes. 43
Thrombospondin‐1 (TSP‐1), adhesive glycoprotein, mediates cell‐to‐cell and cell‐to‐matrix interactions and play crucial roles in platelet aggregation. TSP‐1 has been described as anti‐carcinogenic due to its potent anti‐angiogenic properties. 44 In addition, a significant reduction in TSP‐1 was reported up to 24 months prior to diagnosis of PC with DM compared to non‐cancer DM. 45 These findings suggest that TSP‐1 could be a novel target in lowering the risk of PC. Aside from that, miRNAs are being evaluated as novel therapeutic targets and biomarkers for PC. A combination of serum miRNAs (miR‐483‐5p, miR‐19a, miR‐29a, miR‐20a, miR‐24, miR‐25) is potential biomarker for the diagnosis and discrimination of PC‐DM from non‐cancer new‐onset DM. 46 , 47 Future studies are needed to validate the function of these miRNA in PC.
The bromodomain and extraterminal domain (BET) proteins are a bromodomain subfamily that includes BRD2, BRD3, BRD4 and BRDT. 48 , 49 , 50 , 51 , 52 The BET proteins utilize bromodomains to interact with acetylated histone tails and mediate downstream functions, such as histone acetylation recognition, chromatin remodelling and transcription regulation. They have been implicated to play roles in inflammation, apoptosis, cell proliferation and cell cycle and cancer. 50 , 53 Bromodomain containing protein 4 (BRD4) is a coactivator of NFkB, enhances the transcription of NFkB‐dependent pro‐inflammatory cytokine genes. 54 In addition to activating NFkB, BET proteins also regulate STAT signalling pathway. 55 The inhibition of BET protein attenuates cytokine production (such as IL‐6, IL‐17 and IL‐1β), which have been implicated in diabetogenic inflammation. 56 , 57 As a linkage of diabetes and inflammation, Hsp60, a mitochondrial stress protein, regulate immune inflammation by acting as a ligand for innate immune receptors and adaptive immune receptors. 58
Immune cells, including macrophages, B cells and T cells, play key roles in the progression of T2DM. They contribute to cytokine driven diabetogenic inflammation and the development of T2DM. 59 , 60 , 61 The most common pro‐inflammatory T‐cell subsets implicated in diabetogenic inflammation are Th1 cells, Th17 cells and CD8+T cells. 62 , 63 , 64 , 65 It was shown that T cells from humans with T2DM only produce levels of pathogenic IL‐17 in the presence of B cells 66 which was found in visceral adipose tissue of obese people. 67 IL‐17 produced by γδ T cells drives neutrophil polarization into a myeloid‐derived suppressor cell (MDSC)‐like phenotype, resulting in inhibition of cytotoxic CD8+T cells and promotion of metastasis. 68 CD4+T cells tend to polarize to pro‐inflammatory Th1 and Th17 cells in peripheral blood and adipose tissue of T2DM patients. In contrast, polarization of anti‐inflammatory Th2 cells was decreased. M1 pro‐inflammatory macrophages induced the secretion of IL‐1β, IL‐6, IL‐12, TNF‐α, CCL5 and CCL2 in adipose tissue during obesity. 69 , 70 During islet inflammation, macrophage transformation shifts towards the pro‐inflammatory activated M1‐like phenotype, which contributes to β cell dysfunction in T2DM. 71 , 72 Consumption of foods high in saturated fatty acids (palmitate) is known as one of the risk factors of T2DM. There was palmitate‐dependent recruitment of CD11b+Ly‐6C+classically activated M1‐like monocytes/macrophages to the islets. 73 TLR4 in islet cells and Myd88 were required for M1‐like macrophage recruitment, leading the β cell dysfunction via secretion of IL‐1β. 73
4. OBESITY AND PANCREATIC CANCER
The prevalence of obesity is on the rise worldwide and is considered a serious pandemic today. It is now well accepted that the obesity is one of the leading risk factors for PC. 3 , 74 , 75 , 76 Although obese people possess an increased risk of developing PC, patients with increased pancreatic fat have a poorer outcome than those who develop cancer in a lean pancreas. 77 The mechanisms by which obesity mediates the risk for PC are not well understood. Studies have suggested the involvement of inflammation and hormonal imbalance. Emerging evidence demonstrates that the increase of certain hormones in obese patients, such as insulin, adipokines and resistin, and oxidative stress are responsible for the initiation and progression of PC. 78 , 79 Adipocytes in obesity secreted high IL‐1β, recruiting tumour‐associated neutrophils (TAN) which induces activation of pancreatic stellate cells (PSC). IL‐1β, TAN and PSC induced the aggravation of desmoplasia which is modulated by angiotensin‐II type‐1 receptor, and promoted PC progression. 80 Obesity has been found to be associated with increased systemic levels of placental growth factor (PLGF). 81 , 82 PLGF/VEGFR‐1 system modulates angiogenesis and promotes tumour‐associated macrophage (TAM) recruitment and activity in PC. 83 Therefore, targeting PLGF/VEGFR‐1 signalling may be an attractive strategy to reprogramme the tumour immune microenvironment and inhibit obesity‐induced acceleration of PC progression. 84
Recent studies have shown a link between obesity and obesity‐related PC through adipokines. 85 , 86 Epidemiologic studies reported low adiponectin levels in human obesity and have been associated with increased PC risk. As adiponectin can suppress PC growth by inhibiting the β‐Catenin signalling pathway, 87 the activation of adiponectin signalling could be a novel therapeutic strategy for obesity‐related PC.
Obesity is the leading risk factor associated with the development of T2DM. Obesity causes the activation of multiple inflammatory signalling pathways. Some of these pathways are associated with adipose tissue hypoxia, 88 , 89 leptin secretion and unfolded protein responses that are activated by endoplasmic reticulum stress. 90 Secretion of inflammatory cytokines (eg TNFα, IL‐6) and increased lipolysis can also occur during obesity‐related complications. 91 Visceral adipose tissue (VAT) is one of the major sites for the inflammation that is associated with DM. 59 , 92 Immune cells infiltrate VAT, resulting in low‐grade chronic inflammation and secretion of pro‐inflammatory cytokines that can induce insulin resistance, both locally and systemically. 93
The abdominal adiposity is one of the modifiable risk factors for PC onset, suggesting weight loss could manifest an effective preventive measure. Lifestyle modifications (eg exercise and healthy diets) for the purpose of reducing obesity could also reduce PC rates. In cases when these measures alone were found to be ineffective, bariatric (metabolic) surgery was a useful alternative. 86 , 94 , 95 Bariatric surgery in extremely obese patients may produce many health benefits along with weight loss, consequently reducing the chances of PC, especially when additionally incorporating healthy lifestyle habits.
5. GENETIC MUTATIONS IN PANCREATIC CANCER
Genome‐wide association studies have shown that the pancreatic development genes, HNF1A, HNF1B, HNF4G, PDX1 and NR5A2 contributed to the significant association of PC. 85 , 96 , 97 HNF1A and HNF1B are also associated with T2DM. 85 , 96 In addition, the glucokinase regulator single‐nucleotide polymorphism (SNP) rs780094 with CC genotype was associated with PC risk in T2DM patients. 98 The DM risk alleles of two SNPs, FTO rs8050136 and MTNR1B rs1387153, showed slight but significant associations with increased PC risk, 99 , 100 supporting with the notion that DM or obesity (which is influenced by the FTO locus) 101 increases PC risk. 102 The KRAS mutations are among the earliest genetic alterations which have been associated with the initiation of PC. 4 , 103 , 104
Apart from that, the role of BRCA2 alterations has been used in early diagnosis and prediction of familial PC patients. 105 BRCA2 mutation was a late event in sporadic pancreatic tumorigenesis 106 preceded by KRAS mutation (G12D) or loss of TP53. 107 PALB2 was found to be the second most commonly mutated gene for hereditary PC. 108 Interestingly, the most commonly mutated gene was BRCA2, whose protein product was capable of binding with the PALB2 protein. 109 The germline mutations of STK11 (a tumour suppressor gene) are associated with Peutz–Jeghers syndrome which increases the risk of PC. 110 Moreover, mutations in STK11 gene have also been reported in sporadic PC. 111 , 112
6. MICROENVIRONMENT, IMMUNE CELLS AND PANCREATIC CANCER
Microenvironment plays a major role in cancer initiation, progression and metastasis. 113 Stroma consists of various cell types including fibroblasts and immune cells, and the extracellular matrix (ECM) secreted by the cellular components. In mouse model of PC, targeting the hyaluronan in the ECM of stroma relieved the pressure on the blood vessels resulting in enhancing the survival of tumour‐bearing mice. 114 In contrast, PEGylated hyaluronidase failed to improve the survival of patients. 115 Signalling pathways in stroma environment play significant roles in chemotherapy resistance. For example, inhibition of the Sonic Hedgehog (Shh) pathway depleted tumour‐associated stromal tissue, increased micro‐vessel density and drug delivery in animals 116 but failed to offer the benefit to patients. The tumour‐associated stroma (TAS) is responsible for deposition of collagen and various ECM components that stimulate cancer cell proliferation and angiogenesis. 117 MyD88‐dependent TAS responded to PC cell–secreted factors resulting in loss of Th1 function and inhibition of cytotoxicity by the CD8+T cells. 117 , 118 Pancreatic cancer cells express prostate stem cell antigen (PSCA). Immunosuppressive function of IL‐4 in microenvironment was investigated by using the fusion of IL‐4 receptor exodomain with immunostimulatory IL‐7 receptor endodomain in PSCA‐targeting T cells, resulting in potent and sustained anti‐tumour effects. 119 Shh/Gli signalling pathway plays a significant role in both epithelial and stromal cells of PDAC tumours. 120 In PDAC, the inhibition of bromodomain and extraterminal domain (BET) protein resulted in down‐regulation of Shh. 120 Shh ablation resulted in diminished stroma formation, reduced survival due to formation of more aggressive, dedifferentiated tumours and increased metastases. 121 Inconsistently, stromal depletion resulted in decreased survival with similarly aggressive tumours. 122 The increased tumour vascularity with stromal depletion correlated with disease progression but also increased responsiveness to anti‐angiogenic agents. Elevated pro‐angiogenic vascular endothelial growth factor A (VEGF‐A) levels in patients have been found to correlate with increased vascular density of PDAC and greater disease progression.
Accumulation of hyaluronan/ hyaluronic acids (HA) promotes tumour growth in mice and correlates with poor prognosis in patients with PDAC. Inhibiting HA signalling or depleting HA levels in tumour stroma could represent a promising therapeutic strategy against PDAC. Production of HA was used as the primary determinant of elevated intratumoural pressures. In PDAC, targeting the HA in the ECM of stroma significantly reduced the pressure on the blood vessels and enhanced the survival of tumour‐bearing mice. 114 Indeed, treatment with human recombinant hyaluronidase (PEGPH20) relieved intratumoural pressures, increase in tumour vascular perfusion and gemcitabine delivery, improved survival and decreased the metastatic burden. 123
Regulatory T (Treg) cells, myeloid‐derived suppressor cells (MDSCs) and macrophages appears early in PDAC development. T helper 17 (TH17) with few CD8+lymphocytes were also found in the microenvironment of the KRAS PDAC model. 124 Depletion of carcinoma‐associated fibroblasts (CAFs) that express fibroblast activation protein (FAP) resulted in an increased anti‐tumour cytotoxic T‐cell‐mediated inhibition of PDAC. 125 Furthermore, blocking the activity of CAF‐secreted cytokine CXCL12 induced rapid T‐cell accumulation in tumours and acted synergistically with PD‐L1 to greatly deplete cancer cell. 125 Analysis of resected tumour specimens demonstrated that that intratumoural levels of IL‐8, IL‐1β and TNFα were associated with larger tumours and poor differentiation. 126 Inflammation within the pancreatic tumour microenvironment has been linked to tumour progression and chemo‐resistance through IL‐6, Toll‐like receptor, NFkB and TGF‐β signalling pathways. 127 In KrasG12D mouse model of PC, we have demonstrated that Th17, PMN‐MDSC, IL‐6 and IL‐8 (Th2 type) cells were up‐regulated, whereas CTL, NKT, γδT, NK and IFNϒ (Th1 type) cells were suppressed. 128 These studies suggest that microenvironment plays a significant role in the development of PC.
7. NUTRITION AND PANCREATIC CANCER
Malnutrition and consumption of unhealthy diets have been associated with incidence of PC; however, the results have been inconsistent. 4 , 129 The Western diet, containing high red meat, refined grains and sugar‐sweetened beverages, has been associated with increased levels of systemic inflammatory markers 130 and increased risk of PC. 4 In contrast, the Mediterranean‐style diet, containing plant foods, whole grains and fish, has been associated with reduced levels of inflammatory markers 131 , 132 and reduced risk of PC. 133 The higher inflammatory scores of diet increased the risk of PC with odds ratio (OR) of 2.54, 134 confirming a previous study on Italian population in which higher OR was associated with PC risk. 135 Inflammatory conditions created by unhealthy diet may contribute to pancreatic carcinogenesis by increasing blood levels of inflammatory cytokines (eg IL‐6, IL‐8, IL‐1β, TNF‐α and IFN‐ϒ), which can promote excessive levels of reactive oxygen species. 136 As a result, these inflammatory conditions cause DNA damage, mutagenesis and, thus lead to the development of PC. 136 Interesting consumption of red and processed meat was shown to increase risk of PC in men but not in women through unknown mechanism. 137 The consumption of citrus fruits, vitamin B6 and choline has been inversely correlated with the risk of PC. 138 , 139 Intake of N‐3 (ω‐3) polyunsaturated fatty acids and docosahexaenoic acid from fish was associated with a lower risk of PC. 140 Unsaturated fatty acids was associated with decreased risk of developing PC whereas saturated fatty acids found in dairy products increased the risk of PC. 141 Like saturated fatty acids, higher consumption of sugar and high‐sugar foods was associated with a greater risk of PC. 142 The intake of folate also lowered the risk of PC. 143 , 144 The folic acid exerts its effects via reducing RhoA activity mediated by activation of the FR/cSrc/p190RhoGAP signalling pathway. 145 Dietary fat‐induced PC growth and metastasis via endogenous cholecystokinin (CCK). 146
Purified natural products have shown promising results in cell culture and mouse models of diabetes and PC. 4 Epigallocatechin‐3‐gallate (EGCG), a polyphenolic compound from green tea, showed anticancer activities in cell culture and mouse xenograft model of PC. 147 , 148 Several other natural products such as mangostin, curcumin, resveratrol, silibinin, sulforaphane, anthothecol, berberine and embelin have demonstrated anticancer activities against PC. 4 , 128 , 149 , 150 , 151 , 152 , 153 , 154 , 155 , 156 , 157 , 158 , 159 These agents inhibited PC growth, development and metastasis by generally suppressing NFκB, AKT, Shh, Notch or Wnt pathways. 4 , 160 Although effective in mouse models, these agents have not demonstrated satisfactory pharmacokinetic (PK) and pharmacodynamics (PD) profiles in clinical trials. Nanotechnology appears to be promising in improving the PK/PD profiles of natural products in mouse models of PC and diabetes. 4 , 158 , 161 , 162 , 163 Clinical trials are needed to assess the therapeutic potential of nanoparticles containing natural products for the treatment and/or prevention of PC and diabetes.
8. ANTIHYPERGLYCAEMIC THERAPY AND RISK OF PC
There is some evidence that therapies that increase insulin levels such as exogenous insulin may have potential increase PC risk. 164 Metformin (T2DM drug) can decreases insulin levels by decreasing insulin resistance and also reduces the risk of pancreatic malignancy with hazard ratio of 0.15‐0.54. 165 The use of metformin in patients with diabetes and pancreatic malignancy prolonged median survival compared to control (non‐users): 16 vs 11 months. 166 , 167 , 168 , 169 , 170 Although an anti‐tumour effect of metformin has been shown in preclinical and epidemiological studies, other studies have not shown a consistent survival benefit from metformin in PC patients with pre‐existing diabetes. 171 , 172 Metformin is known to activate the LKB1‐AMPK pathway, a cellular energy stress‐sensing mechanism, and blocks proliferation through inhibition of mTOR which also enhances cell proliferation by activation the insulin signalling pathway. 173 , 174 In addition to reducing glucose levels in T2DM, the use of metformin in prevention of PC is very promising and needs further evaluation.
9. PREVENTION OF PANCREATIC CANCER
The first step in preventive strategy is avoiding the exposure to the modifiable risk factors such as smoking, drinking, obesity, carcinogenic chemicals and some specific foods (saturated fats). PC protecting foods (polyphenols or flavonoids, folic acid and fish) should be included in daily dietary. Recent studies have recommended the use of polyphenols rich in specific foods (cloves, peppermint, star anise, cocoa powder and so on) as a chemopreventive approach for PC. 175 , 176 The health‐related properties of a wide range of dietary constituents show potential biological activities against PC. 177 Resveratrol, a red wine polyphenol, can directly bind and inhibit leukotriene A4 hydrolase (LTA‐4H) activity and, therefore, induces the production of LTB4 178 which prevents the development of PC. 154 , 179 Most natural products including curcumin inhibit PC growth and metastasis by suppressing NF‐κB and its targets. 179 , 180 Therefore, resveratrol and curcumin can be used for preventing PC in high‐risk patients. The preliminary data on resveratrol and curcumin indicated potential benefits but needs confirmation in clinical trials. It is also critical to diagnose and start radical treatment for gallstones, cholecystitis and chronic pancreatitis and set up a PC screening in these high‐risk patients. So far, no tumour‐specific markers with high sensitivity and specificity have existed for PC. The carbohydrate antigen 19‐9 (CA19‐9) as a prognostic marker of PDAC has been used with a limited success. Serum CA19‐9 maker has low specificity but high sensitivity. 181 , 182 Using quantitative proteomic analysis, C4b‐binding protein alpha‐chain (C4BPA) was identified as a novel serum biomarker for diagnosis of early stage PDAC. 183 Therefore, CA19‐9 and C4BPA could be used for screening high‐risk individuals with PC.
10. CONCLUSIONS
In spite of enormous research efforts, pancreatic cancer remains a deadly disease with incremental benefits seen with cytotoxic chemotherapy in recent years. Fortunately, the treatment of patients with chemotherapy has nearly doubled median overall survival but still remains very poor (<1 year). Immunotherapy, vaccine and checkpoint inhibition appear to be emerging modalities for PC. Diabetes, obesity and dietary patterns are closely correlated to cancer risk. Thus, avoiding these risks will obviously attenuate the morbidity and mortality of cancers. Furthermore, regular exercise and improving lifestyle can help in reducing obesity and diabetes. In addition, the discovery of new cancer‐related genes is critical for the cancer screening strategy and prevention. Eventually, the better comprehension of pathogenesis and novel therapies should be more extensively studied to lower or eliminate morbidity and mortality of PC.
CONFLICT OF INTEREST
All the authors confirm that there are no conflicts of interest.
AUTHOR CONTRIBUTIONS
BQL wrote the manuscript. AS, SS and RKS supervised activity planning and execution. All authors reviewed the manuscript.
ACKNOWLEDGEMENTS
We thank all the lab members for critical reading of the manuscript.
Quoc Lam B, Shrivastava SK, Shrivastava A, Shankar S, Srivastava RK. The Impact of obesity and diabetes mellitus on pancreatic cancer: Molecular mechanisms and clinical perspectives. J Cell Mol Med. 2020;24:7706–7716. 10.1111/jcmm.15413
REFERENCES
- 1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2020. CA Cancer J Clin. 2020;70:7‐30. [DOI] [PubMed] [Google Scholar]
- 2. Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM, Matrisian LM. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res. 2014;74:2913‐2921. [DOI] [PubMed] [Google Scholar]
- 3. Barone E, Corrado A, Gemignani F, Landi S. Environmental risk factors for pancreatic cancer: an update. Arch Toxicol. 2016;90:2617‐2642. [DOI] [PubMed] [Google Scholar]
- 4. Singh D, Upadhyay G, Srivastava RK, Shankar S. Recent advances in pancreatic cancer: biology, treatment, and prevention. Biochim Biophys Acta. 2015;1856:13‐27. [DOI] [PubMed] [Google Scholar]
- 5. Huxley RR, Ansary‐Moghaddam A, Clifton P, Czernichow S, Parr CL, Woodward M. The impact of dietary and lifestyle risk factors on risk of colorectal cancer: a quantitative overview of the epidemiological evidence. Int J Cancer. 2009;125:171‐180. [DOI] [PubMed] [Google Scholar]
- 6. Li Y, Yang H, Cao J. Association between alcohol consumption and cancers in the Chinese population–a systematic review and meta‐analysis. PLoS ONE. 2011;6:e18776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Chari ST, Leibson CL, Rabe KG, Ransom J, de Andrade M, Petersen GM. Probability of pancreatic cancer following diabetes: a population‐based study. Gastroenterology. 2005;129:504‐511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Permert J, Adrian TE, Jacobsson P, Jorfelt L, Fruin AB, Larsson J. Is profound peripheral insulin resistance in patients with pancreatic cancer caused by a tumor‐associated factor? Am J Surg. 1993;165:61‐66; discussion 6–7. [DOI] [PubMed] [Google Scholar]
- 9. Gupta N, Hoffman RP, Veng‐Pedersen P. Pharmacokinetic/pharmacodynamic insulin‐glucose analysis for differentiation of beta‐cell function: an 18 month follow‐up study in pre‐pubertal lean and obese children. Biopharm Drug Dispos. 2006;27:257‐265. [DOI] [PubMed] [Google Scholar]
- 10. Goh TT, Mason TM, Gupta N, et al. Lipid‐induced beta‐cell dysfunction in vivo in models of progressive beta‐cell failure. Am J Physiol Endocrinol Metab. 2007;292:E549‐E560. [DOI] [PubMed] [Google Scholar]
- 11. Pezzilli R. Alcohol abuse and pancreatic diseases: an overview. Recent Pat Inflamm Allergy Drug Discov. 2015;9:102‐106. [DOI] [PubMed] [Google Scholar]
- 12. Tramacere I, Scotti L, Jenab M, et al. Alcohol drinking and pancreatic cancer risk: a meta‐analysis of the dose‐risk relation. Int J Cancer. 2010;126:1474‐1486. [DOI] [PubMed] [Google Scholar]
- 13. Hermans MP, Levy JC, Morris RJ, Turner RC. Comparison of insulin sensitivity tests across a range of glucose tolerance from normal to diabetes. Diabetologia. 1999;42:678‐687. [DOI] [PubMed] [Google Scholar]
- 14. Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006;444:840‐846. [DOI] [PubMed] [Google Scholar]
- 15. McFarland MS, Knight TN, Brown A, Thomas J. The continuation of oral medications with the initiation of insulin therapy in type 2 diabetes: a review of the evidence. South Med J. 2010;103:58‐65. [DOI] [PubMed] [Google Scholar]
- 16. Muniraj T, Chari ST. Diabetes and pancreatic cancer. Minerva Gastroenterol Dietol. 2012;58:331‐345. [PMC free article] [PubMed] [Google Scholar]
- 17. Gullo L, Pezzilli R, Morselli‐Labate AM, Italian Pancreatic Cancer Study G . Diabetes and the risk of pancreatic cancer. N Engl J Med. 1994;331:81‐84. [DOI] [PubMed] [Google Scholar]
- 18. Jee SH, Ohrr H, Sull JW, Yun JE, Ji M, Samet JM. Fasting serum glucose level and cancer risk in Korean men and women. JAMA. 2005;293:194‐202. [DOI] [PubMed] [Google Scholar]
- 19. Ryu TY, Park J, Scherer PE. Hyperglycemia as a risk factor for cancer progression. Diabetes Metab J. 2014;38:330‐336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Wang L, Bai YY, Yang Y, et al. Diabetes mellitus stimulates pancreatic cancer growth and epithelial‐mesenchymal transition‐mediated metastasis via a p38 MAPK pathway. Oncotarget. 2016;7:38539‐38550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Ono K, Han J. The p38 signal transduction pathway: activation and function. Cell Signal. 2000;12:1‐13. [DOI] [PubMed] [Google Scholar]
- 22. Wagner EF, Nebreda AR. Signal integration by JNK and p38 MAPK pathways in cancer development. Nat Rev Cancer. 2009;9:537‐549. [DOI] [PubMed] [Google Scholar]
- 23. Gupta J, del Barco BI, Igea A, et al. Dual function of p38alpha MAPK in colon cancer: suppression of colitis‐associated tumor initiation but requirement for cancer cell survival. Cancer Cell. 2014;25:484‐500. [DOI] [PubMed] [Google Scholar]
- 24. Yasui H, Hideshima T, Ikeda H, et al. BIRB 796 enhances cytotoxicity triggered by bortezomib, heat shock protein (Hsp) 90 inhibitor, and dexamethasone via inhibition of p38 mitogen‐activated protein kinase/Hsp27 pathway in multiple myeloma cell lines and inhibits paracrine tumour growth. Br J Haematol. 2007;136:414‐423. [DOI] [PubMed] [Google Scholar]
- 25. Liu H, Ma Q, Li J. High glucose promotes cell proliferation and enhances GDNF and RET expression in pancreatic cancer cells. Mol Cell Biochem. 2011;347:95‐101. [DOI] [PubMed] [Google Scholar]
- 26. Li B, Wang Y, Liu Y, Ma J, Li Y. Altered gene expression involved in insulin signaling pathway in type II diabetic osteoporosis rats model. Endocrine. 2013;43:136‐146. [DOI] [PubMed] [Google Scholar]
- 27. Li Q, Zhang J, Zhou Y, Qiao L. Obesity and gastric cancer. Front Biosci. 2012;17:2383‐2390. [DOI] [PubMed] [Google Scholar]
- 28. Fisher SJ, Lekas MC, McCall RH, Shi ZQ, Giacca A, Vranic M. Determinants of glucose turnover in the pathophysiology of diabetes: an in vivo analysis in diabetic dogs. Diabetes Metab. 1996;22:111‐121. [PubMed] [Google Scholar]
- 29. Qureshi SA, Ding V, Li Z, et al. Activation of insulin signal transduction pathway and anti‐diabetic activity of small molecule insulin receptor activators. J Biol Chem. 2000;275:36590‐36595. [DOI] [PubMed] [Google Scholar]
- 30. Li J, Cao G, Ma Q, Liu H, Li W, Han L. The bidirectional interation between pancreatic cancer and diabetes. World J Surg Oncol. 2012;10:171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Ahmed AS, Li J, Schizas N, et al. Expressional changes in growth and inflammatory mediators during Achilles tendon repair in diabetic rats: new insights into a possible basis for compromised healing. Cell Tissue Res. 2014;357:109‐117. [DOI] [PubMed] [Google Scholar]
- 32. Gealekman O, Gurav K, Chouinard M, et al. Control of adipose tissue expandability in response to high fat diet by the insulin‐like growth factor‐binding protein‐4. J Biol Chem. 2014;289:18327‐18338. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Han L, Ma Q, Li J, et al. High glucose promotes pancreatic cancer cell proliferation via the induction of EGF expression and transactivation of EGFR. PLoS ONE. 2011;6:e27074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Li W, Ma Q, Li J, et al. Hyperglycemia enhances the invasive and migratory activity of pancreatic cancer cells via hydrogen peroxide. Oncol Rep. 2011;25:1279‐1287. [DOI] [PubMed] [Google Scholar]
- 35. Bhattacharyya S, Sul K, Krukovets I, Nestor C, Li J, Adognravi OS. Novel tissue‐specific mechanism of regulation of angiogenesis and cancer growth in response to hyperglycemia. J Am Heart Assoc. 2012;1:e005967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Lemoine AY, Ledoux S, Queguiner I, et al. Link between adipose tissue angiogenesis and fat accumulation in severely obese subjects. J Clin Endocrinol Metab. 2012;97:E775‐E780. [DOI] [PubMed] [Google Scholar]
- 37. Sparvero LJ, Asafu‐Adjei D, Kang R, et al. RAGE (Receptor for Advanced Glycation Endproducts), RAGE ligands, and their role in cancer and inflammation. J Transl Med. 2009;7:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Kang P, Tian C, Jia C. Association of RAGE gene polymorphisms with type 2 diabetes mellitus, diabetic retinopathy and diabetic nephropathy. Gene. 2012;500:1‐9. [DOI] [PubMed] [Google Scholar]
- 39. Grote VA, Nieters A, Kaaks R, et al. The associations of advanced glycation end products and its soluble receptor with pancreatic cancer risk: a case‐control study within the prospective EPIC Cohort. Cancer Epidemiol Biomark Prev. 2012;21:619‐628. [DOI] [PubMed] [Google Scholar]
- 40. Hirata K, Takada M, Suzuki Y, Kuroda Y. Expression of receptor for advanced glycation end products (RAGE) in human biliary cancer cells. Hepatogastroenterology. 2003;50:1205‐1207. [PubMed] [Google Scholar]
- 41. Kuniyasu H, Chihara Y, Kondo H. Differential effects between amphoterin and advanced glycation end products on colon cancer cells. Int J Cancer. 2003;104:722‐727. [DOI] [PubMed] [Google Scholar]
- 42. Abe R, Shimizu T, Sugawara H, et al. Regulation of human melanoma growth and metastasis by AGE‐AGE receptor interactions. J Investig Dermatol. 2004;122:461‐467. [DOI] [PubMed] [Google Scholar]
- 43. Abe R, Yamagishi S. AGE‐RAGE system and carcinogenesis. Curr Pharm Des. 2008;14:940‐945. [DOI] [PubMed] [Google Scholar]
- 44. Miyata Y, Sakai H. Thrombospondin‐1 in urological cancer: pathological role, clinical significance, and therapeutic prospects. Int J Mol Sci. 2013;14:12249‐12272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Jenkinson C, Elliott VL, Evans A, et al. Decreased serum thrombospondin‐1 levels in pancreatic cancer patients up to 24 months prior to clinical diagnosis: association with diabetes mellitus. Clin Cancer Res. 2016;22:1734‐1743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Dai G, Huang L, Du Y, Yang L, Yu P. Solid pseudopapillary neoplasms of the pancreas: clinical analysis of 45 cases. Int J Clin Exp Pathol. 2015;8:11400‐11406. [PMC free article] [PubMed] [Google Scholar]
- 47. Dai X, Pang W, Zhou Y, et al. Altered profile of serum microRNAs in pancreatic cancer‐associated new‐onset diabetes mellitus. J Diabetes. 2016;8:422‐433. [DOI] [PubMed] [Google Scholar]
- 48. Belkina AC, Denis GV. BET domain co‐regulators in obesity, inflammation and cancer. Nat Rev Cancer. 2012;12:465‐477. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Schilderink R, Bell M, Reginato E, et al. BET bromodomain inhibition reduces maturation and enhances tolerogenic properties of human and mouse dendritic cells. Mol Immunol. 2016;79:66‐76. [DOI] [PubMed] [Google Scholar]
- 50. Nicholas DA, Andrieu G, Strissel KJ, Nikolajczyk BS, Denis GV. BET bromodomain proteins and epigenetic regulation of inflammation: implications for type 2 diabetes and breast cancer. Cell Mol Life Sci. 2017;74:231‐243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Padmanabhan B, Mathur S, Manjula R, Tripathi S. Bromodomain and extra‐terminal (BET) family proteins: New therapeutic targets in major diseases. J Biosci. 2016;41:295‐311. [DOI] [PubMed] [Google Scholar]
- 52. Huang M, Qiu Q, Xiao Y, et al. BET bromodomain suppression inhibits VEGF‐induced angiogenesis and vascular permeability by blocking VEGFR2‐mediated activation of PAK1 and eNOS. Sci Rep. 2016;6:23770. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Zaware N, Zhou MM. Chemical modulators for epigenome reader domains as emerging epigenetic therapies for cancer and inflammation. Curr Opin Chem Biol. 2017;39:116‐125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Zhou M, Huang K, Jung KJ, et al. Bromodomain protein Brd4 regulates human immunodeficiency virus transcription through phosphorylation of CDK9 at threonine 29. J Virol. 2009;83:1036‐1044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Chan CH, Fang C, Yarilina A, Prinjha RK, Qiao Y, Ivashkiv LB. BET bromodomain inhibition suppresses transcriptional responses to cytokine‐Jak‐STAT signaling in a gene‐specific manner in human monocytes. Eur J Immunol. 2015;45:287‐297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Xiao Y, Liang L, Huang M, et al. Bromodomain and extra‐terminal domain bromodomain inhibition prevents synovial inflammation via blocking IkappaB kinase‐dependent NF‐kappaB activation in rheumatoid fibroblast‐like synoviocytes. Rheumatology. 2016;55:173‐184. [DOI] [PubMed] [Google Scholar]
- 57. Nadeem A, Al‐Harbi NO, Al‐Harbi MM, et al. Imiquimod‐induced psoriasis‐like skin inflammation is suppressed by BET bromodomain inhibitor in mice through RORC/IL‐17A pathway modulation. Pharmacol Res. 2015;99:248‐257. [DOI] [PubMed] [Google Scholar]
- 58. Juwono J, Martinus RD. Does Hsp60 provide a link between mitochondrial stress and inflammation in diabetes mellitus? J Diabetes Res. 2016;2016:8017571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Winer S, Winer DA. The adaptive immune system as a fundamental regulator of adipose tissue inflammation and insulin resistance. Immunol Cell Biol. 2012;90:755‐762. [DOI] [PubMed] [Google Scholar]
- 60. Song MJ, Kim KH, Yoon JM, Kim JB. Activation of Toll‐like receptor 4 is associated with insulin resistance in adipocytes. Biochem Biophys Res Commun. 2006;346:739‐745. [DOI] [PubMed] [Google Scholar]
- 61. Hotamisligil GS. Inflammation, metaflammation and immunometabolic disorders. Nature. 2017;542:177‐185. [DOI] [PubMed] [Google Scholar]
- 62. Shaul ME, Bennett G, Strissel KJ, Greenberg AS, Obin MS. Dynamic, M2‐like remodeling phenotypes of CD11c+ adipose tissue macrophages during high‐fat diet–induced obesity in mice. Diabetes. 2010;59:1171‐1181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Winer N, Branger B, Azria E, et al. L‐Arginine treatment for severe vascular fetal intrauterine growth restriction: a randomized double‐bind controlled trial. Clin Nutr. 2009;28:243‐248. [DOI] [PubMed] [Google Scholar]
- 64. Eljaafari A, Robert M, Chehimi M, et al. Adipose tissue‐derived stem cells from obese subjects contribute to inflammation and reduced insulin response in adipocytes through differential regulation of the Th1/Th17 balance and monocyte activation. Diabetes. 2015;64:2477‐2488. [DOI] [PubMed] [Google Scholar]
- 65. Guo H, Xu BC, Yang XG, et al. A high frequency of peripheral blood IL‐22(+) CD4(+) T cells in patients with new onset type 2 diabetes mellitus. J Clin Lab Anal. 2016;30:95‐102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. DeFuria J, Belkina AC, Jagannathan‐Bogdan M, et al. B cells promote inflammation in obesity and type 2 diabetes through regulation of T‐cell function and an inflammatory cytokine profile. Proc Natl Acad Sci USA. 2013;110:5133‐5138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. McDonnell ME, Umpierrez GE. Insulin therapy for the management of hyperglycemia in hospitalized patients. Endocrinol Metab Clin North Am. 2012;41:175‐201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Coffelt SB, Kersten K, Doornebal CW, et al. IL‐17‐producing gammadelta T cells and neutrophils conspire to promote breast cancer metastasis. Nature. 2015;522:345‐348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Olefsky JM, Glass CK. Macrophages, inflammation, and insulin resistance. Annu Rev Physiol. 2010;72:219‐246. [DOI] [PubMed] [Google Scholar]
- 70. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. Macrophage polarization: tumor‐associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002;23:549‐555. [DOI] [PubMed] [Google Scholar]
- 71. Jourdan T, Godlewski G, Cinar R, et al. Activation of the Nlrp3 inflammasome in infiltrating macrophages by endocannabinoids mediates beta cell loss in type 2 diabetes. Nat Med. 2013;19:1132‐1140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Westwell‐Roper CY, Ehses JA, Verchere CB. Resident macrophages mediate islet amyloid polypeptide‐induced islet IL‐1beta production and beta‐cell dysfunction. Diabetes. 2014;63:1698‐1711. [DOI] [PubMed] [Google Scholar]
- 73. Eguchi K, Manabe I, Oishi‐Tanaka Y, et al. Saturated fatty acid and TLR signaling link beta cell dysfunction and islet inflammation. Cell Metab. 2012;15:518‐533. [DOI] [PubMed] [Google Scholar]
- 74. Andersson G, Wennersten C, Borgquist S, Jirstrom K. Pancreatic cancer risk in relation to sex, lifestyle factors, and pre‐diagnostic anthropometry in the Malmo Diet and Cancer Study. Biol Sex Differ. 2016;7:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Becker AE, Hernandez YG, Frucht H, Lucas AL. Pancreatic ductal adenocarcinoma: risk factors, screening, and early detection. World J Gastroenterol. 2014;20:11182‐11198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Davoodi SH, Malek‐Shahabi T, Malekshahi‐Moghadam A, Shahbazi R, Esmaeili S. Obesity as an important risk factor for certain types of cancer. Iran J Cancer Prev. 2013;6:186‐194. [PMC free article] [PubMed] [Google Scholar]
- 77. Catanzaro R, Cuffari B, Italia A, Marotta F. Exploring the metabolic syndrome: Nonalcoholic fatty pancreas disease. World J Gastroenterol. 2016;22:7660‐7675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Di Ciaula A, Fat PP. epigenome and pancreatic diseases. Interplay and common pathways from a toxic and obesogenic environment. Eur J Intern Med. 2014;25:865‐873. [DOI] [PubMed] [Google Scholar]
- 79. Matsuda M, Shimomura I. Increased oxidative stress in obesity: implications for metabolic syndrome, diabetes, hypertension, dyslipidemia, atherosclerosis, and cancer. Obes Res Clin Pract. 2013;7:e330‐e341. [DOI] [PubMed] [Google Scholar]
- 80. Incio J, Liu H, Suboj P, et al. Obesity‐induced inflammation and desmoplasia promote pancreatic cancer progression and resistance to chemotherapy. Cancer Discov. 2016;6:852‐869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Pervanidou P, Chouliaras G, Akalestos A, et al. Increased placental growth factor (PlGF) concentrations in children and adolescents with obesity and the metabolic syndrome. Hormones. 2014;13:369‐374. [DOI] [PubMed] [Google Scholar]
- 82. Lappas M. Markers of endothelial cell dysfunction are increased in human omental adipose tissue from women with pre‐existing maternal obesity and gestational diabetes. Metabolism. 2014;63:860‐873. [DOI] [PubMed] [Google Scholar]
- 83. Hiratsuka S. Vasculogenensis, angiogenesis and special features of tumor blood vessels. Front Biosci. 2011;16:1413‐1427. [DOI] [PubMed] [Google Scholar]
- 84. Incio J, Tam J, Rahbari NN, et al. PlGF/VEGFR‐1 signaling promotes macrophage polarization and accelerated tumor progression in obesity. Clin Cancer Res. 2016;22:2993‐3004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Li D. Diabetes and pancreatic cancer. Mol Carcinog. 2012;51:64‐74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Zhou B, Wu D, Liu H, et al. Obesity and pancreatic cancer: an update of epidemiological evidence and molecular mechanisms. Pancreatology. 2019;19:941‐950. [DOI] [PubMed] [Google Scholar]
- 87. Jiang J, Fan Y, Zhang W, et al. Adiponectin suppresses human pancreatic cancer growth through attenuating the beta‐catenin signaling pathway. Int J Biol Sci. 2019;15:253‐264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Hosogai N, Fukuhara A, Oshima K, et al. Adipose tissue hypoxia in obesity and its impact on adipocytokine dysregulation. Diabetes. 2007;56:901‐911. [DOI] [PubMed] [Google Scholar]
- 89. Rausch ME, Weisberg S, Vardhana P, Tortoriello DV. Obesity in C57BL/6J mice is characterized by adipose tissue hypoxia and cytotoxic T‐cell infiltration. Int J Obesity. 2008;32:451‐463. [DOI] [PubMed] [Google Scholar]
- 90. Hummasti S, Hotamisligil GS. Endoplasmic reticulum stress and inflammation in obesity and diabetes. Circ Res. 2010;107:579‐591. [DOI] [PubMed] [Google Scholar]
- 91. Halberg N, Wernstedt‐Asterholm I, Scherer PE. The adipocyte as an endocrine cell. Endocrinol Metab Clin North Am. 2008;37: 753‐768, x–xi. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Winer DA, Winer S, Shen L, Chng MH, Engleman EG. B lymphocytes as emerging mediators of insulin resistance. Int J Obesity Suppl. 2012;2:S4‐S7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Khodabandehloo H, Gorgani‐Firuzjaee S, Panahi G, Meshkani R. Molecular and cellular mechanisms linking inflammation to insulin resistance and beta‐cell dysfunction. Translational Res. 2016;167:228‐256. [DOI] [PubMed] [Google Scholar]
- 94. Yeung KTD, Reddy M, Purkayastha S. Surgical options for glycaemic control in Type 1 diabetes. Diabet Med. 2019;36:414‐423. [DOI] [PubMed] [Google Scholar]
- 95. Mulla CM, Storino A, Yee EU, et al. Insulinoma after bariatric surgery: diagnostic dilemma and therapeutic approaches. Obes Surg. 2016;26:874‐881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Wu C, Kraft P, Stolzenberg‐Solomon R, et al. Genome‐wide association study of survival in patients with pancreatic adenocarcinoma. Gut. 2014;63:152‐160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Wei P, Tang H, Li D. Insights into pancreatic cancer etiology from pathway analysis of genome‐wide association study data. PLoS ONE. 2012;7:e46887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Prizment AE, Gross M, Rasmussen‐Torvik L, Peacock JM, Anderson KE. Genes related to diabetes may be associated with pancreatic cancer in a population‐based case‐control study in Minnesota. Pancreas. 2012;41:50‐53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Pierce BL, Ahsan H. Genome‐wide, "pleiotropy scan" identifies HNF1A region as a novel pancreatic cancer susceptibility locus. Cancer Res. 2011;71:4352‐4358. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Pierce BL, Austin MA, Ahsan H. Association study of type 2 diabetes genetic susceptibility variants and risk of pancreatic cancer: an analysis of PanScan‐I data. Cancer Causes Control. 2011;22:877‐883. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Frayling TM, McCarthy MI. Genetic studies of diabetes following the advent of the genome‐wide association study: where do we go from here? Diabetologia. 2007;50:2229‐2233. [DOI] [PubMed] [Google Scholar]
- 102. Arslan E, Yiginer O, Yavasoglu I, Ozcelik F, Kardesoglu E, Nalbant S. Effect of uncomplicated obesity on QT interval in young men. Pol Arch Med Wewn. 2010;120:209‐213. [PubMed] [Google Scholar]
- 103. Ying H, Dey P, Yao W, et al. Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev. 2016;30:355‐385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Chari ST, Kelly K, Hollingsworth MA, et al. Early detection of sporadic pancreatic cancer: summative review. Pancreas. 2015;44:693‐712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Martinez‐Useros J, Garcia‐Foncillas J. The role of BRCA2 mutation status as diagnostic, predictive, and prognosis biomarker for pancreatic cancer. Biomed Res Int. 2016;2016:1869304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Goggins M, Hruban RH, Kern SE. BRCA2 is inactivated late in the development of pancreatic intraepithelial neoplasia: evidence and implications. Am J Pathol. 2000;156:1767‐1771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Rowley M, Ohashi A, Mondal G, et al. Inactivation of Brca2 promotes Trp53‐associated but inhibits KrasG12D‐dependent pancreatic cancer development in mice. Gastroenterology. 2011;140:1303‐1313.e1‐3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Jones S, Hruban RH, Kamiyama M, et al. Exomic sequencing identifies PALB2 as a pancreatic cancer susceptibility gene. Science. 2009;324:217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Xia B, Sheng Q, Nakanishi K, et al. Control of BRCA2 cellular and clinical functions by a nuclear partner, PALB2. Mol Cell. 2006;22:719‐729. [DOI] [PubMed] [Google Scholar]
- 110. Mehenni H, Resta N, Park JG, Miyaki M, Guanti G, Costanza MC. Cancer risks in LKB1 germline mutation carriers. Gut. 2006;55:984‐990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Birnbaum DJ, Adelaide J, Mamessier E, et al. Genome profiling of pancreatic adenocarcinoma. Genes Chromosom Cancer. 2011;50:456‐465. [DOI] [PubMed] [Google Scholar]
- 112. Birnbaum MJ, Shaw RJ. Genomics: drugs, diabetes and cancer. Nature. 2011;470:338‐339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Whatcott CJ, Han H, Von Hoff DD. Orchestrating the tumor microenvironment to improve survival for patients with pancreatic cancer: normalization. Not Destruction. Cancer J. 2015;21:299‐306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Hingorani SR, Wang L, Multani AS, et al. Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell. 2005;7:469‐483. [DOI] [PubMed] [Google Scholar]
- 115. Wong KM, Horton KJ, Coveler AL, Hingorani SR, Harris WP. Targeting the tumor stroma: the biology and clinical development of pegylated recombinant human hyaluronidase (PEGPH20). Curr Oncol Rep. 2017;19:47. [DOI] [PubMed] [Google Scholar]
- 116. Olive KP, Jacobetz MA, Davidson CJ, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324:1457‐1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Delitto D, Zhang D, Han S, et al. Nicotine reduces survival via augmentation of paracrine HGF‐MET signaling in the pancreatic cancer microenvironment. Clin Cancer Res. 2016;22:1787‐1799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Delitto D, Delitto AE, DiVita BB, et al. Human pancreatic cancer cells induce a MyD88‐dependent stromal response to promote a tumor‐tolerant immune microenvironment. Cancer Res. 2017;77:672‐683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Mohammed S, Sukumaran S, Bajgain P, et al. Improving chimeric antigen receptor‐modified T cell function by reversing the immunosuppressive tumor microenvironment of pancreatic cancer. Mol Ther. 2017;25:249‐258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Huang Y, Nahar S, Nakagawa A, et al. Regulation of GLI underlies a role for BET bromodomains in pancreatic cancer growth and the tumor microenvironment. Clin Cancer Res. 2016;22:4259‐4270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Rhim AD, Oberstein PE, Thomas DH, et al. Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 2014;25:735‐747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Ozdemir BC, Pentcheva‐Hoang T, Carstens JL, et al. Depletion of carcinoma‐associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell. 2014;25:719‐734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer Cell. 2012;21:418‐429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Zhang Y, McAllister F, Pasca di Magliano M. Immune cells in pancreatic cancer: Joining the dark side. Oncoimmunology. 2014;3:e29125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Feig C, Jones JO, Kraman M, et al. Targeting CXCL12 from FAP‐expressing carcinoma‐associated fibroblasts synergizes with anti‐PD‐L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci USA. 2013;110:20212‐20217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Delitto D, Black BS, Sorenson HL, et al. The inflammatory milieu within the pancreatic cancer microenvironment correlates with clinicopathologic parameters, chemoresistance and survival. BMC Cancer. 2015;15:783. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Muerkoster S, Wegehenkel K, Arlt A, et al. Tumor stroma interactions induce chemoresistance in pancreatic ductal carcinoma cells involving increased secretion and paracrine effects of nitric oxide and interleukin‐1beta. Cancer Res. 2004;64:1331‐1337. [DOI] [PubMed] [Google Scholar]
- 128. Marsh JL, Jackman CP, Tang SN, Shankar S, Srivastava RK. Embelin suppresses pancreatic cancer growth by modulating tumor immune microenvironment. Front Biosci. 2014;19:113‐125. [DOI] [PubMed] [Google Scholar]
- 129. Greer JB, Brand RE. New developments in pancreatic cancer. Curr Gastroenterol Rep. 2011;13:131‐139. [DOI] [PubMed] [Google Scholar]
- 130. Lopez‐Garcia E, Schulze MB, Fung TT, et al. Major dietary patterns are related to plasma concentrations of markers of inflammation and endothelial dysfunction. Am J Clin Nutr. 2004;80:1029‐1035. [DOI] [PubMed] [Google Scholar]
- 131. Esmaillzadeh A, Kimiagar M, Mehrabi Y, Azadbakht L, Hu FB, Willett WC. Dietary patterns and markers of systemic inflammation among Iranian women. J Nutr. 2007;137:992‐998. [DOI] [PubMed] [Google Scholar]
- 132. Esposito K, Marfella R, Ciotola M, et al. Effect of a mediterranean‐style diet on endothelial dysfunction and markers of vascular inflammation in the metabolic syndrome: a randomized trial. JAMA. 2004;292:1440‐1446. [DOI] [PubMed] [Google Scholar]
- 133. Bosetti C, Turati F, Dal Pont A, et al. The role of Mediterranean diet on the risk of pancreatic cancer. Br J Cancer. 2013;109:1360‐1366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Antwi SO, Oberg AL, Shivappa N, et al. Pancreatic cancer: associations of inflammatory potential of diet, cigarette smoking and long‐standing diabetes. Carcinogenesis. 2016;37:481‐490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Shivappa N, Bosetti C, Zucchetto A, Serraino D, La Vecchia C, Hebert JR. Dietary inflammatory index and risk of pancreatic cancer in an Italian case‐control study. Br J Nutr. 2015;113:292‐298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Farrow B, Evers BM. Inflammation and the development of pancreatic cancer. Surg Oncol. 2002;10:153‐169. [DOI] [PubMed] [Google Scholar]
- 137. Zhao Z, Yin Z, Pu Z, Zhao Q. Association between consumption of red and processed meat and pancreatic cancer risk: a systematic review and meta‐analysis. Clin Gastroenterol Hepatol. 2017;15:486‐93 e10. [DOI] [PubMed] [Google Scholar]
- 138. Bae JM, Lee EJ, Guyatt G. Citrus fruit intake and pancreatic cancer risk: a quantitative systematic review. Pancreas. 2009;38:168‐174. [DOI] [PubMed] [Google Scholar]
- 139. Huang JY, Butler LM, Wang R, Jin A, Koh WP, Yuan JM. Dietary intake of one‐carbon metabolism‐related nutrients and pancreatic cancer risk: the Singapore Chinese Health Study. Cancer Epidemiol Biomark Prev. 2016;25:417‐424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Hidaka A, Shimazu T, Sawada N, et al. Fish, n‐3 PUFA consumption, and pancreatic cancer risk in Japanese: a large, population‐based, prospective cohort study. Am J Clin Nutr. 2015;102:1490‐1497. [DOI] [PubMed] [Google Scholar]
- 141. Jansen RJ, Robinson DP, Frank RD, et al. Fatty acids found in dairy, protein and unsaturated fatty acids are associated with risk of pancreatic cancer in a case‐control study. Int J Cancer. 2014;134:1935‐1946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Larsson SC, Bergkvist L, Wolk A. Consumption of sugar and sugar‐sweetened foods and the risk of pancreatic cancer in a prospective study. Am J Clin Nutr. 2006;84:1171‐1176. [DOI] [PubMed] [Google Scholar]
- 143. Larsson SC, Giovannucci E, Wolk A. Folate intake, MTHFR polymorphisms, and risk of esophageal, gastric, and pancreatic cancer: a meta‐analysis. Gastroenterology. 2006;131:1271‐1283. [DOI] [PubMed] [Google Scholar]
- 144. Larsson SC, Giovannucci E, Wolk A. Folate intake and stomach cancer incidence in a prospective cohort of Swedish women. Cancer Epidemiol Biomark Prev. 2006;15:1409‐1412. [DOI] [PubMed] [Google Scholar]
- 145. Hou TC, Lin JJ, Wen HC, Chen LC, Hsu SP, Lee WS. Folic acid inhibits endothelial cell migration through inhibiting the RhoA activity mediated by activating the folic acid receptor/cSrc/p190RhoGAP‐signaling pathway. Biochem Pharmacol. 2013;85:376‐384. [DOI] [PubMed] [Google Scholar]
- 146. Matters GL, Cooper TK, McGovern CO, et al. Cholecystokinin mediates progression and metastasis of pancreatic cancer associated with dietary fat. Dig Dis Sci. 2014;59:1180‐1191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Shankar S, Ganapathy S, Srivastava RK. Green tea polyphenols: biology and therapeutic implications in cancer. Front Biosci. 2007;12:4881‐4899. [DOI] [PubMed] [Google Scholar]
- 148. Shankar S, Siddiqui I, Srivastava RK. Molecular mechanisms of resveratrol (3,4,5‐trihydroxy‐trans‐stilbene) and its interaction with TNF‐related apoptosis inducing ligand (TRAIL) in androgen‐insensitive prostate cancer cells. Mol Cell Biochem. 2007;304:273‐285. [DOI] [PubMed] [Google Scholar]
- 149. Huang M, Tang SN, Upadhyay G, et al. Embelin suppresses growth of human pancreatic cancer xenografts, and pancreatic cancer cells isolated from KrasG12D mice by inhibiting Akt and Sonic hedgehog pathways. PLoS ONE. 2014;9:e92161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Huang M, Tang SN, Upadhyay G, et al. Rottlerin suppresses growth of human pancreatic tumors in nude mice, and pancreatic cancer cells isolated from Kras(G12D) mice. Cancer Lett. 2014;353:32‐40. [DOI] [PubMed] [Google Scholar]
- 151. Li SH, Fu J, Watkins DN, Srivastava RK, Shankar S. Sulforaphane regulates self‐renewal of pancreatic cancer stem cells through the modulation of Sonic hedgehog‐GLI pathway. Mol Cell Biochem. 2013;373:217‐227. [DOI] [PubMed] [Google Scholar]
- 152. Rodova M, Fu J, Watkins DN, Srivastava RK, Shankar S. Sonic hedgehog signaling inhibition provides opportunities for targeted therapy by sulforaphane in regulating pancreatic cancer stem cell self‐renewal. PLoS ONE. 2012;7:e46083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Shankar S, Ganapathy S, Hingorani SR, Srivastava RK. EGCG inhibits growth, invasion, angiogenesis and metastasis of pancreatic cancer. Front Biosci. 2008;13:440‐452. [DOI] [PubMed] [Google Scholar]
- 154. Shankar S, Nall D, Tang SN, et al. Resveratrol inhibits pancreatic cancer stem cell characteristics in human and KrasG12D transgenic mice by inhibiting pluripotency maintaining factors and epithelial‐mesenchymal transition. PLoS ONE. 2011;6:e16530. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Shankar S, Suthakar G, Srivastava RK. Epigallocatechin‐3‐gallate inhibits cell cycle and induces apoptosis in pancreatic cancer. Front Biosci. 2007;12:5039‐5051. [DOI] [PubMed] [Google Scholar]
- 156. Singh BN, Kumar D, Shankar S, Srivastava RK. Rottlerin induces autophagy which leads to apoptotic cell death through inhibition of PI3K/Akt/mTOR pathway in human pancreatic cancer stem cells. Biochem Pharmacol. 2012;84:1154‐1163. [DOI] [PubMed] [Google Scholar]
- 157. Srivastava RK, Tang SN, Zhu W, Meeker D, Shankar S. Sulforaphane synergizes with quercetin to inhibit self‐renewal capacity of pancreatic cancer stem cells. Front Biosci (Elite Ed). 2011;3:515‐528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Verma RK, Yu W, Singh SP, Shankar S, Srivastava RK. Anthothecol‐encapsulated PLGA nanoparticles inhibit pancreatic cancer stem cell growth by modulating sonic hedgehog pathway. Nanomedicine. 2015;11:2061‐2070. [DOI] [PubMed] [Google Scholar]
- 159. Zhao M, Tang SN, Marsh JL, Shankar S, Srivastava RK. Ellagic acid inhibits human pancreatic cancer growth in Balb c nude mice. Cancer Lett. 2013;337:210‐217. [DOI] [PubMed] [Google Scholar]
- 160. Upadhyay G, Shankar S, Srivastava RK. Stem cells in neurological disorders: emerging therapy with stunning hopes. Mol Neurobiol. 2015;52:610‐625. [DOI] [PubMed] [Google Scholar]
- 161. Verma RK, Yu W, Shrivastava A, Shankar S, Srivastava RK. alpha‐Mangostin‐encapsulated PLGA nanoparticles inhibit pancreatic carcinogenesis by targeting cancer stem cells in human, and transgenic (Kras(G12D), and Kras(G12D)/tp53R270H) mice. Sci Rep. 2016;6:32743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Sharma SB, Gupta R. Drug development from natural resource: a systematic approach. Mini Rev Med Chem. 2015;15:52‐57. [DOI] [PubMed] [Google Scholar]
- 163. Zhang HM, Liang FX, Chen R. Ancient records and modern research on the mechanisms of chinese herbal medicines in the treatment of diabetes mellitus. Evid Based Complement Alternat Med. 2015;2015:747982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Bosetti C, Rosato V, Li D, et al. Diabetes, antidiabetic medications, and pancreatic cancer risk: an analysis from the International Pancreatic Cancer Case‐Control Consortium. Ann Oncol. 2014;25:2065‐2072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Zhang P, Li H, Tan X, Chen L, Wang S. Association of metformin use with cancer incidence and mortality: a meta‐analysis. Cancer Epidemiol. 2013;37:207‐218. [DOI] [PubMed] [Google Scholar]
- 166. Zhou DC, Gong H, Tan CQ, Luo JQ. Prognostic significance of anti‐diabetic medications in pancreatic cancer: a meta‐analysis. Oncotarget. 2017;8:62349‐62357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Dong YW, Shi YQ, He LW, Cui XY, Su PZ. Effects of metformin on survival outcomes of pancreatic cancer: a meta‐analysis. Oncotarget. 2017;8:55478‐55488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Jy E, Graber JM, Lu SE, Lin Y, Lu‐Yao G, Tan XL. Effect of metformin and statin use on survival in pancreatic cancer patients: a systematic literature review and meta‐analysis. Curr Med Chem. 2018;25:2595‐2607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Chaiteerakij R, Petersen GM, Bamlet WR, et al. Metformin Use and Survival of Patients With Pancreatic Cancer: A Cautionary Lesson. J Clin Oncol. 2016;34:1898‐1904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Franciosi M, Lucisano G, Lapice E, Strippoli GF, Pellegrini F, Nicolucci A. Metformin therapy and risk of cancer in patients with type 2 diabetes: systematic review. PLoS ONE. 2013;8:e71583. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Jang WI, Kim MS, Kang SH, et al. Association between metformin use and mortality in patients with type 2 diabetes mellitus and localized resectable pancreatic cancer: a nationwide population‐based study in korea. Oncotarget. 2017;8:9587‐9596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172. Choi Y, Kim TY, Oh DY, et al. The impact of diabetes mellitus and metformin treatment on survival of patients with advanced pancreatic cancer undergoing chemotherapy. Cancer Res Treat. 2016;48:171‐179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173. Rozengurt E, Sinnett‐Smith J, Kisfalvi K. Crosstalk between insulin/insulin‐like growth factor‐1 receptors and G protein‐coupled receptor signaling systems: a novel target for the antidiabetic drug metformin in pancreatic cancer. Clin Cancer Res. 2010;16:2505‐2511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Gong J, Robbins LA, Lugea A, Waldron RT, Jeon CY, Pandol SJ. Diabetes, pancreatic cancer, and metformin therapy. Front Physiol. 2014;5:426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Perez‐Jimenez J, Neveu V, Vos F, Scalbert A. Identification of the 100 richest dietary sources of polyphenols: an application of the Phenol‐Explorer database. Eur J Clin Nutr. 2010;64(Suppl 3):S112‐S120. [DOI] [PubMed] [Google Scholar]
- 176. Perez‐Jimenez J, Neveu V, Vos F, Scalbert A. Systematic analysis of the content of 502 polyphenols in 452 foods and beverages: an application of the phenol‐explorer database. J Agric Food Chem. 2010;58:4959‐4969. [DOI] [PubMed] [Google Scholar]
- 177. Ramos S. Effects of dietary flavonoids on apoptotic pathways related to cancer chemoprevention. J Nutr Biochem. 2007;18:427‐442. [DOI] [PubMed] [Google Scholar]
- 178. Oi N, Jeong CH, Nadas J, et al. Resveratrol, a red wine polyphenol, suppresses pancreatic cancer by inhibiting leukotriene A(4)hydrolase. Cancer Res. 2010;70:9755‐9764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179. Boreddy SR, Srivastava SK. Pancreatic cancer chemoprevention by phytochemicals. Cancer Lett. 2013;334:86‐94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180. Anajafi T, Yu J, Sedigh A, et al. Nuclear localizing peptide‐conjugated, redox‐sensitive polymersomes for delivering curcumin and doxorubicin to pancreatic cancer microtumors. Mol Pharm. 2017;14:1916‐1928. [DOI] [PubMed] [Google Scholar]
- 181. Cao S, Hu Y, Gao X, Liao Q, Zhao Y. Serum carbohydrate antigen 19–9 in differential diagnosis of benign and malignant pancreatic cystic neoplasms: a meta‐analysis. PLoS ONE. 2016;11:e0166406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Imaoka H, Shimizu Y, Senda Y, et al. Post‐adjuvant chemotherapy CA19‐9 levels predict prognosis in patients with pancreatic ductal adenocarcinoma: a retrospective cohort study. Pancreatology. 2016;16:658‐664. [DOI] [PubMed] [Google Scholar]
- 183. Sogawa K, Takano S, Iida F, et al. Identification of a novel serum biomarker for pancreatic cancer, C4b‐binding protein alpha‐chain (C4BPA) by quantitative proteomic analysis using tandem mass tags. Br J Cancer. 2016;115:949‐956. [DOI] [PMC free article] [PubMed] [Google Scholar]
