Abstract
Pancreatic ductal adenocarcinoma continues to be a lethal disease, for which efficient treatment options are very limited. Increasing efforts have been taken to understand how to prevent or intercept this disease at an early stage. There is convincing evidence from epidemiologic and preclinical studies that the anti-diabetic drug metformin possesses beneficial effects in pancreatic cancer, including reducing the risk of developing the disease and improving survival in patients with early stage disease. This review will summarize the current literature about the epidemiological data on metformin and pancreatic cancer as well as describe the preclinical evidence illustrating the anti-cancer effects of metformin in pancreatic cancer. Underlying mechanisms and targets of metformin will also be discussed. These include direct effects on transformed pancreatic epithelial cells and indirect, systemic effects on extra-pancreatic tissues.
Keywords: Metformin, pancreatic cancer, epidemiology, preclinical studies, mechanisms
Pancreatic cancer is a highly significant public health problem
Pancreatic ductal adenocarcinoma (PDAC), the most common histologic subtype of pancreatic malignancies, is still one of the most lethal types of cancer. The latest report of the American Cancer Society estimates 60,430 new cases (28,480 females and 31,950 males) of pancreatic cancer in the year 2021 [1]. An estimated 22,950 female and 25,270 male patients will succumb to this disease in 2021, putting pancreatic cancer as the fourth leading cause of cancer mortality in women and men [1]. Alarmingly, the incidence of pancreatic cancer has increased over the last years. In 2016, the American Cancer Society’s estimates for pancreatic cancer were 53,070 (25,400 females and 27,670 males) [2], marking an almost 13.9% increase (12.1% in females and 15.5% in males) in total estimated pancreatic cancer cases in the last 5 years. In fact, it has been projected that the mortality of pancreatic cancers will surpass the deaths from colorectal cancer by 2030, making pancreatic cancers the second leading cause of cancer-related deaths [3]. Only a minority of patients with PDAC are eligible for surgical resection due to the early local and distant spread. Although advances have been made in neoadjuvant and adjuvant chemotherapeutic regimens, the benefit in prolonging survival is still disappointingly small and recurrences occur often. In addition, despite significant progress in the discovery and understanding of molecular pathways driving PDAC growth, molecular targeted therapy has not yet resulted in significant breakthroughs. A different approach to reduce the burden of pancreatic cancer on patients and to lower the number of deaths from this disease is to identify strategies that either prevent or intercept PDAC [4, 5]. In this context, a detailed understanding of modifiable risk factors and repurposing currently used drugs will most likely guide the rapid implementation of new preventive strategies.
Preclinical animal models are invaluable to study the mechanisms of risk factors and to develop interventional strategies. It is well established now that most PDACs arise through a stepwise progression from precursor lesions [6], e.g. pancreatic intraepithelial neoplasias (PanINs). There is general consensus about the importance of mutated KRAS in the initiation of PDAC [7]. This notion is strongly supported by genetically engineered animal models of PDAC development [8, 9]. Accordingly, the model that faithfully recapitulates the progression of human PDAC in mice involves the expression of a mutant Kras (KrasG12D) from the endogenous Kras locus through a Cre recombinase that is under control of the pancreas-specific promoter p48 (LSL-KrasG12D; p48-Cre mice referred to as the KC model [10]). In this model the expression of mutated Kras in pancreatic progenitor cells during embryogenesis leads to PanIN formation and progression to PDAC [11]. However, in the conditional Kras mouse model (without additional genetic alterations) the development of invasive PDAC occurs very late (usually after 9 months) and only in ~ 10% of the animals [8]. Mutated Kras is necessary for the initiation but is not sufficient for driving the progression of PanINs to invasive PDAC. However, the presence of another mutation, e.g. in Trp53 (transformation-related protein 53), and/or certain environmental conditions greatly accelerate PDAC development [9].
Metabolic disorders, including obesity and type 2 diabetes mellitus (T2DM) are associated with an increased risk and worse clinical outcomes for developing PDAC and other cancers [12–17]. These connections have been recapitulated in preclinical mouse models of PDAC. In particular, diet-induced and genetic obesity strikingly accelerate the progression of PanINs to invasive cancer in mice [18–22]. Diet-related metabolic disturbances, including obesity and early stages of T2DM, which are prominent in the western world [23–26], are multifaceted but characterized by peripheral insulin resistance, compensatory overproduction of insulin, and increased bioavailability of IGF-1 (insulin-like growth factor-1). Elevated levels of insulin and IGF-1, which are potent growth factors for PDAC cells, are generally thought to be of great importance for the development and progression of cancers, including PDAC [27–30].
Metformin as an intriguing agent in pancreatic cancer: epidemiological evidence
Metformin (1,1-dimethylbiguanide hydrochloride) is the most widely prescribed drug for prevention and treatment of T2DM worldwide [31]. The primary systemic effect of metformin is the lowering of blood glucose levels predominantly through reduced hepatic gluconeogenesis. The molecular pathways responsible for this reduction in glucose production remain a subject of debate [32]. A number of studies suggest that a considerable proportion of the antidiabetic action of metformin derives from effects on the gastrointestinal (GI) tract [33–35], and this will revisited in a later section.
Over the past 10–15 years there have been many epidemiologic studies that analyzed a potential association of metformin use with a reduced risk of PDAC (preventive effect) or improved survival of patients with PDAC (therapeutic effect). Overall, the available literature suggests a beneficial effect of preventive and therapeutic efficacy of metformin in patients with underlying T2DM, with stronger evidence for a preventive effect, although the published reports are not universally consistent. Most epidemiologic studies found an association of metformin use with a reduced incidence of PDAC in patients with T2DM. In a single-institution, case-control study diabetic patients who had taken metformin had a significantly lower risk of PDAC compared with those who had not taken metformin (Odds Ratio [OR]: 0.38; 95% confidence interval [CI]: 0.22–0.69) [36]. Interestingly, diabetic patients who had taken insulin or insulin secretagogues had a significantly higher risk of PDAC compared with diabetic patients who had not taken these drugs [36], supporting the hypothesis that insulin is a potent mitogenic and cancer promoting factor. In contrast, a large cohort study from the Netherlands did not detect an association of current and cumulative use of metformin with a decreased risk of GI cancer, including PDAC [37]. A meta-analysis published in 2010, analyzing eleven studies, reported a significant inverse association between PDAC risk and metformin use [38]. Another meta-analysis in 2013 analyzing 37 studies comprising over 1,500,000 participants found a risk reduction of 46% for developing PDAC in metformin users [39]. Finally, a recent umbrella review on the potential effect of metformin on cancer, which included 21 systematic reviews and meta-analyses covering 11 major anatomical sites, found strong evidence for the association between metformin use and decreased pancreatic cancer incidence [40].
Besides strong epidemiologic evidence of an association of metformin use and decreased risk of PDAC, several studies assessed a potential association of metformin and improved survival of PDAC patients. A retrospective study of patients with diabetes and PDAC treated at the University of Texas MD Anderson Cancer Center showed a better survival in patients with metformin use [41]. The 2-year survival rate was 30.1% for the metformin group and 15.4% for the non-metformin group. The median overall survival time was 15.2 months for the metformin group, and 11.1 months for the non-metformin group [41]. In that study metformin use was significantly associated with longer survival only in patients with non-metastatic disease [41]. A nationwide population-based cohort study in Korea suggested that metformin might decrease cancer-specific mortality rates in localized resectable PDAC patients with pre-existing diabetes [42]. The cancer-specific survival (5-year, 31.9% vs. 22.2%) was significantly higher in the metformin users than in the diabetic metformin non-users [42]. In contrast, a large cohort study, which retrospectively analyzed 980 patients with PDAC failed to detect a benefit of metformin [43]. Median survival of metformin users versus nonusers was 9.9 versus 8.9 months, respectively and metformin use was not significantly associated with improved survival (Hazard Ratio [HR]=0.93; P=0.28) [43]. A double-blind, randomized, placebo-controlled phase 2 trial in the Netherlands in patients with advanced PDAC did not find a benefit of metformin when added to gemcitabine and erlotinib [44]. Overall survival at 6 months was 63.9% in the placebo group and 56.7% in the metformin group (P=0.41). There was no difference in overall survival between groups (median 7.6 months vs 6.8 months in the metformin group; HR=1.056; log-rank P=0.78) [44]. Several meta-analyses, however, described a beneficial association between metformin use and survival in PDAC patients. A systematic review and meta-analysis of nine studies found a relative survival benefit in patients with PDAC and preexisting T2DM associated with metformin treatment compared with non-metformin treatment in overall survival [45]. A recent meta-analysis in 2020 of 21 studies that included 38,772 patients with PDAC and concurrent T2DM found a significant survival benefit in the metformin treatment group compared with non-metformin group (HR=0.83) [46]. Interestingly, the survival benefit was observed in patients at early stage (HR=0.75) and mixed stage (HR=0.81), but not for patients at an advanced stage (HR=0.99) [46]. Also, the survival benefit was seen in patients receiving surgery (HR=0.82) and comprehensive treatment (HR=0.85), but not in the chemotherapy group (HR=0.99) [46]. Some of the contrasting results of epidemiologic studies may be related to weaknesses in the study design and data analysis. In fact, a recent study that included patients who only received metformin after surgery and corrected for immortal bias, a major weakness in many previous publications, concluded that metformin use is associated with a higher overall survival following pancreaticoduodenectomy in patients with T2DM and PDAC [47]. In contrast, another study assessing the impact of immortal time bias in previous cohort studies concluded that the correlation between the use of metformin and PDAC survival was exaggerated in several studies due to the existence of immortal time bias [48]. This contrasting conclusions highlight the importance of more rigorous statistical designs in investigating the effects of metformin on PDAC survival. Another potentially confounding and complicating factor in interpreting the efficacy of metformin on PDAC risk and survival in epidemiologic studies is the often concomitant use of several drugs, e.g. metformin, aspirin, statins, in patients with T2DM or metabolic syndrome [49]. A new retrospective analysis of 33,784 patients from the Korean National Health Insurance Service showed a reduction of all-cause mortality in diabetic cancer survivors using metformin [50]. Taken together the available epidemiologic evidence suggest a positive correlation between metformin use and survival in patients with early stage PDAC and co-existing T2DM. Indeed, a meta-analysis from 2017 indicated that the effects of metformin depend on tumor stage, with marked improved survival in patients with locally advanced disease but not in patients with metastatic PDAC [51].
Metformin as an intriguing agent in pancreatic cancer: preclinical evidence
Early preclinical evidence of a potential preventive effect of metformin in pancreatic carcinogenesis came from hamster studies. Metformin administration in the drinking water prevented n-nitrosobis-(2-oxopropyl)amine-induced pancreatic carcinogenesis in hamster fed a high fat diet [52]. Using an allograft model, metformin significantly reduced tumor growth in prediabetic C57BL/6 mice with diet-induced obesity (DIO) [53]. The anti-cancer effect of metformin in allograft/xenograft models of pancreatic cancer has been corroborated by several other studies [54–58]. Metformin and phenformin (another related antidiabetic drug from the biguanide class) have also been shown to significantly reduce tumor growth in some patient-derived xenografts [59] but not all [60]. Our own earlier studies have shown that intraperitoneal administration of metformin (at 250 mg/kg) significantly reduced the growth of subcutaneous pancreatic cancer xenografts (using MIAPaCa-2 and PANC-1 human pancreatic cancer cell lines) in nu/nu mice [61]. Subsequently, we reported that metformin (administered intraperitoneally) dose-dependently inhibited subcutaneous pancreatic cancer xenograft growth with a significant growth inhibitory effect starting at 50 mg/kg [62]. Importantly, metformin also inhibited pancreatic cancer growth when xenografted orthotopically and administered orally (2.5 mg/ml in drinking water) [62].
Using a genetically engineered mouse model (LSL-Kras;Trp53), administration of metformin (intraperitoneally at 125 mg/kg) 1 or 3 weeks before tumor initiation (via direct orthotopic injection of adenoviral Cre constructs) and continued for another 6 weeks significantly reduced pancreatic tumor volume [63]. Other studies reported that daily oral gavage of metformin (at 200 mg/kg) inhibited tumor growth and pancreatitis and prolonged survival in LSL-KrasG12D/+;Pdx1-Cre (KC) and LSL-KrasG12D/+;Trp53fl/+;Pdx1-Cre (KPC) mice [64] as well as inhibited angiogenesis and enhanced chemosensitivity of gemcitabine in KPC mice [65]. Our own studies demonstrated that metformin decreased the incidence of PDAC in KrasG12D/+;p48-Cre (KC) mice with DIO [66, 67]. In that study male and female KC mice were randomly fed with a control diet (CD), high fat, high calorie diet (HFCD), or HFCD plus metformin (administered in the drinking water at 5 mg/ml) for 3 or 9 months. After 3 months, metformin prevented HFCD-induced weight gain, liver steatosis, formation of acinar-to-ductal metaplasia (ADM), and advanced PanIN lesions [66]. In addition to reversing hepatic and pancreatic histopathology, metformin normalized HFCD-induced hyperinsulinemia and hyperleptinemia in the 9-month cohort. Importantly, the HFCD-increased PDAC incidence was completely abrogated by metformin [66]. The chemopreventive effects of metformin in this model were associated with significant microbial changes in both the mucosal and luminal microbiome of the duodenum [67]. In contrast to our study, a recent report using KC mice with genetic obesity (Pdx1-Cre;LSL-KrasG12D/+ mice crossed with leptin-deficient [ob/ob] mice; KCO) did not detect a beneficial effect of metformin [22]. In that study, KCO mice were given metformin in the drinking water (2 mg/ml) starting at 6 weeks of age and continued for another 6 weeks. There are important differences, including rapid vs. gradual weight gain, lack of vs. elevated leptin levels, between our model with DIO and the genetic obesity model, which may explain the different response to metformin [22, 68]. Importantly, metformin was administered at a lower concentration (2 mg/ml) for only 6 weeks in the KCO model, which was not associated with weight loss and a decrease in insulin levels [22], while our study used metformin at 5 mg/ml for 3 and 9 months, which was associated with weight loss (also seen in diabetic patients on metformin) and a significant reduction in insulin levels [66]. Overall, there is overwhelming preclinical evidence from allograft/xenograft and genetically engineered mouse models of PDAC that metformin potently inhibits tumor growth and reduces the incidence of PDAC formation in obese and non-obese conditions.
Metformin as an intriguing agent in pancreatic cancer: underlying mechanisms
The mechanism of action of metformin remains incompletely understood. The beneficial effects of metformin on PDAC development and growth may thereby be mediated by direct effects on transformed pancreatic cells, by indirect, systemic extra-pancreatic effects or by a combination of both mechanisms. The systemic effects may include metformin action in the liver, skeletal muscle, adipose tissue, brain, and/or gut [7, 34, 69–75].
Direct cellular effects:
At the cellular level metformin indirectly activates AMPK (adenosine monophosphate-activated protein kinase) [76], although AMPK-independent mechanisms have also been described [69, 77–79]. Most of these studies were carried out in liver cells and attempted to identify the mechanism, by which metformin decreases glucose output. A number of studies were performed using PDAC cells, but an important limitation of most studies is that metformin was used at very high concentrations. While circulating levels of metformin in humans and animals range from 10 μM to 40 μM (approximately 40–70 μM in the portal vein), effects of metformin on signaling pathways and proliferation in PDAC cells were examined using drug concentrations in the culture medium as high as 5–30 mM [57, 80–82]. Therefore, the clinical and mechanistic significance of the effects of metformin on PDAC cells in vitro has been questioned.
Within cells, AMPK exists as heterotrimeric complexes, composed of a catalytic kinase α subunit (isoforms α1 and α2), a scaffolding β subunit (isoforms β1 and β2) and a regulatory γ (isoforms γ1, γ2 and γ3), giving rise to 12 possible α/β/γ combinations in different cell types [83, 84]. It is plausible that the different AMPK complexes have different regulation (e.g. expression, turnover, subcellular localization), regulatory properties (e.g. dependency on 5’-AMP/ADP [adenosine monophosphate/adenosine diphosphate] concentration), and functions in different cell types [85]. Activation of AMPK is achieved by phosphorylation in the activation loop by LKB-1/STK11 (liver kinase B-1/serine-threonine kinase 11) [86] in conditions where cellular levels of ATP (adenosine triphosphate) decrease and 5’-AMP and ADP concentrations rise [31]. Metformin does not act directly on either AMPK or LKB1, but is generally thought to inhibit the complex I of the mitochondrial respiratory chain [87] leading to a decline in ATP production accompanied by a concomitant increase in ADP and AMP levels [88]. The binding of AMP or ADP to the γ1/2 subunits induces a conformational change that facilitates the phosphorylation of the activation loop of the α subunit by LKB-1/STK11 at the highly conserved Thr172 [85]. Interestingly, AMPK with mutant subunits that do not bind AMP are not sensitive to metformin. However, changes in adenine nucleotides can affect the activity of other enzymes in the cell and thus, metformin can exert effects through AMPK independent pathways [32] especially at high concentrations [89–91] that cause pronounced changes in adenine nucleotide levels [90]. A different mechanism of AMPK activation involving a lysosomal pathway has also been proposed [92].
Using human PDAC cells exposed to medium containing a physiological concentration of glucose (5 mM), our studies showed that metformin inhibited cell proliferation at relevant concentrations (50–100 μM). Knock down of AMPKs prevented the inhibitory effect of low (but not high) concentrations of metformin on DNA synthesis in PDAC cells [90, 91]. These studies emphasized that the mechanism of the inhibitory effects of metformin on PDAC cell signaling and proliferation is sharply dose-dependent. At low concentrations, metformin inhibited cell proliferation via AMPK, whereas at high concentrations (>1 mM), metformin elicited cellular responses through AMPK-independent pathways.
A key pathway in KRAS signaling is phosphatidylinositol 3-kinase (PI3K)/Akt leading to mTOR (mechanistic target of rapamycin) activation [93–95]. This signaling module plays a pivotal role in stimulating proliferation of PDAC cells [96, 97], is activated in PDAC tissues and limits catabolic processes, including autophagy [98]. mTOR functions as a catalytic subunit in two distinct multi-protein complexes, mTORC1 (mechanistic target of rapamycin complex 1) and mTORC2 [99]. mTORC1, characterized by Raptor, phosphorylates and controls at least two regulators of protein synthesis, the 40S ribosomal protein subunit S6 kinase (S6K) and the inhibitor of protein synthesis 4E-binding protein 1, referred as 4EBP1 [100–103]. As depicted in Figure 1, AMPK has been shown to inhibit mTORC1 through several mechanisms. First, AMPK can phosphorylate TSC2 (tuberous sclerosis complex 2) on Ser1345 [104–106], thereby activating the complex, which in turn prevents the accumulation of Rheb-GTP (ras homolog enriched in brain-guanosine tri-phosphate), thus inhibiting mTORC1 activation, a process that occurs at the lysosomal membrane. Second, AMPK can also phosphorylate Raptor, which disrupts its binding to mTOR, thereby inhibiting mTORC1 [107]. Finally, AMPK has been shown to phosphorylate IRS-1 (insulin receptor substrate-1) at Ser794, which impedes activation of PI3K [108, 109], thereby leading to inhibition of mTORC1 activation induced by insulin and/or IGF-1. Our own studies have demonstrated that metformin at low concentrations activates AMPK in PDAC cells [90, 91, 96], as judged by the increase in the phosphorylation of both Raptor and ACC (acetyl-CoA carboxylase) Ser79 phosphorylation, thereby inhibiting mTORC1, ERK (extracellular signal-regulated kinase), and DNA synthesis. Accordingly, oral administration of metformin prevented the increase in PDAC incidence in KC mice with DIO [66], which was associated with an increase in AMPK activity in the pancreas (as measured by ACC Ser79 phosphorylation).
Fig. 1.

Schematic of direct molecular, AMPK-dependent effects of metformin
Metformin indirectly activates AMPK, which in turn phosphorylates multiple targets leading to inhibition of the Raf/MEK/ERK/RSK, mTORC1, and YAP/TAZ pathways, all critical oncogenic drivers in pancreatic cancer. Activating phosphorylations are depicted in black, inhibiting phosphorylations in red. Partly created with Servier Medical Art.
Another putative target of AMPK of high significance is the highly conserved Hippo pathway, which is attracting intense attention as a key regulator of development, organ-size, tissue regeneration and tumorigenesis [110, 111]. Hippo signals are transduced through a serine/threonine kinase cascade wherein Mst1/2 kinases (mammalian STE20-like protein kinases 1/2) phosphorylate and activate Lats1/2 (large tumor suppressor 1/2). In turn, Lats1/2 phosphorylates the transcriptional co-activators YAP (Yes-associated protein) and its paralog TAZ (WW-domain-containing transcriptional co-activator with PDZ-binding motif), two central effectors of the Hippo pathway [112–114]. The phosphorylation of YAP/TAZ by Lats1/2 restricts their activity, cellular localization, and stability. In the absence of phosphorylation, YAP/TAZ localizes to the nucleus where it binds and activates predominantly the TEA-domain DNA-binding transcription factors (TEAD 1–4) thereby stimulating the expression of genes. Besides its recognized role in the regulation of growth and development, recent studies show that Hippo kinases and YAP/TAZ transcriptional coactivators, are regulated by metabolism and conversely that the Hippo/YAP/TAZ pathway controls metabolic processes in physiological and pathologic conditions, including obesity and T2DM [115, 116]. Although YAP and TAZ have similar structural topologies, share nearly half of the overall amino acid sequence, and are thought to be largely redundant, they may differ in their regulation and downstream functions [117, 118]. The YAP/TAZ pathway assumes an added importance in PDAC given that YAP is a major downstream target of Ras required for PanIN progression into PDAC [119, 120]. YAP is overexpressed in human pancreatic cancer, particularly in the squamous subtype, and correlates to poor survival [121–126]. YAP/TAZ are upregulated in cerulein-induced acute pancreatitis and strongly expressed in ADM and PanIN lesions in the KC mouse model [120]. YAP has been shown to drive KrasG12D-independent tumor maintenance [123] and collaborates with oncogenic Kras to drive tumorigenesis [127].
Metformin has been shown to inhibit the activity or expression of YAP/TAZ in several (cancer) models [128–133]. The inhibition of YAP/TAZ by metformin is mediated by AMPK, which opposes YAP activity via multiple mechanisms, including direct YAP phosphorylation on Ser94 [134, 135], a site that is critical for the interaction of YAP with TEAD transcription factors. In addition, AMPK has been shown to phosphorylate HMG-CoA (hydroxy-methyl-glutaryl-coenzyme A) reductase on Ser872, which inhibits its activity and decreases mevalonic acid synthesis [136], a pathway critical for YAP regulation through the formation of GG-PP (geranyl-geranyl pyrophosphate) and prenylation of Rho GTPases [137]. In addition, AMPK phosphorylates and activates upstream regulators of the Hippo pathway [138], including LATS, leading to inhibition of YAP activity. In our own studies, we found that KC mice with DIO had markedly elevated YAP and TAZ expression in the pancreas, which was significantly prevented by oral administration of metformin [66]. A schematic overview of AMPK-dependent effects of metformin is shown in Figure 1.
Indirect systemic, extra-pancreatic effects:
As mentioned earlier, the anti-diabetic, glucose-lowering effects of metformin are mediated systemically by decreasing the production of glucose in the liver and by improving the insulin sensitivity by virtue of increasing the uptake of glucose into skeletal muscle cells and adipocytes [139]. In addition, metformin is also able to lower the levels of insulin and IGF-1 in diabetic and non-diabetic patients [140, 141]. There is substantial evidence that suggests a prominent role of insulin and IGF in PDAC development and progression [142–149]. The beneficial effects of metformin in PDAC patients, especially with co-existing obesity and/or T2DM, might thereby be mediated, at least in part, by reducing or normalizing the systemically elevated insulin and IGF levels. This was also seen in our animal study, in which oral administration of metformin normalized the hyperinsulinemia in KC mice with DIO, which was associated with a marked reduction in PDAC incidence [66]. Interestingly, in that study metformin treatment also correlated with a substantial improvement of liver steatosis seen in obese KC mice [66]. In this context it is important to note that non-alcoholic fatty liver disease (NAFLD), often seen in obesity, has been found to be associated with an increased risk of PDAC and with a poorer survival of patients with PDAC [150–152]. However, further research is needed to determine whether the link between NAFLD and PDAC merely reflects the general promotional effect of obesity (and visceral adiposity) on PDAC development or whether specific mechanisms exists that are unique to hepatic steatosis and PDAC formation.
Over the last decade there has been growing evidence indicating the importance of the gut microbiome in obesity and other metabolic conditions [153, 154]. Accordingly, a number of studies have found that the anti-diabetic and anti-obesity actions of metformin may be mediated by its effects on the gut, including the gut microbiome. In fact, the concentration of metformin in the intestine is 30–300 times higher than the levels of the drug in the circulation, implying striking accumulation of metformin in enterocytes [155]. A large observational study and a randomized controlled trial have identified a microbiome signature of metformin [156, 157]. The beneficial anti-diabetic effects of metformin can be transferred to germ free mice through fecal transplantation, providing strong evidence that changes of the gut microbiome underlie at least some of glucose lowering effects of metformin [157]. Findings from human studies have been corroborated by animal studies. Oral administration of metformin to high fat diet-fed mice for 6 weeks improved the glycemic profile, which was associated with a higher abundance of the mucin-degrading bacterium Akkermansia [158]. Oral administration of Akkermansia muciniphila to mice fed the high fat diet but not treated with metformin significantly improved glucose tolerance [158] providing a causal link between microbiome effects of metformin to its anti-diabetic actions. There has been increasing evidence of the importance of the gut microbiome in pancreatic cancer [159–161]. In our own studies we found that KC mice fed a high fat high calorie diet for 3 months displayed a significant shift in the microbial composition and α-diversity in the duodenum, a sign of dysbiosis or microbial imbalance [67]. Oral administration of metformin, which was associated with an attenuation of PDAC development, prevented the changes in the duodenal microbiome. Specifically, the high fat, high calorie diet increased substantially the genus Clostridium sensu stricto within the duodenum, which was prevented by metformin [67]. Importantly, Clostridium sensu stricto negatively correlated with the extent of intact pancreatic acini in KC mice [67]. Overall, there is increasing evidence that at least some of the beneficial effects of metformin in reducing PDAC development may be mediated by its actions on the gut microbiome. However, there is a paucity of causal links and further experimental work is needed to elucidate the role of the intestinal microbiota and their metabolites in PDAC de and in the inhibitory effect of metformin on PDAC development.
In addition to its actions on the gut microbiome, the glucose-lowering effects of metformin in diabetic patients have also been shown to be mediated by increasing the plasma levels of the gut incretin hormone, glucagon-like peptide 1 (GLP1) [162, 163], which is secreted by intestinal enteroendocrine L cells. In turn, GLP1 released locally in the gut can activate a gut-brain-liver neuronal network to inhibit hepatic glucose production [164]. Moreover, a recent elegant study has demonstrated that oral metformin increases circulating growth differentiation factor 15 (GDF15), expressed mainly in the intestine and kidney, which is necessary for metformin’s beneficial effects on energy balance, food intake, and body weight [73]. The effects of metformin on GDF15 were also produced in intestinal organoids growing in three-dimensional cultures [165]. The reduction of food intake and body weight by metformin are also thought to contribute to the anti-cancer activities of metformin, especially in obese and diabetic patients. Figure 2 illustrates some of the indirect, systemic extra-pancreatic effects of metformin.
Fig. 2.

Overview of indirect, systemic extra-pancreatic effects of metformin
Besides its direct effects on transformed pancreatic cells, metformin acts on several other organs/tissues, including liver, skeletal muscle, fat, gut, and brain, leading to a reduction of blood glucose and insulin levels, reduced food intake, weight loss, and gut microbiome changes that all together may also have profound opposing/preventive effects on pancreatic cancer development and growth. Partly created with Servier Medical Art.
Conclusion
There is overwhelming evidence from epidemiologic and preclinical studies that the anti-diabetic drug metformin reduces the risk of developing pancreatic cancer and improves outcome in patients with early stage pancreatic cancer. These anti-cancer effects of metformin may thereby be mediated by its direct actions on transformed pancreatic cells or indirectly by its systemic actions on extra-pancreatic tissues/organs. Among the direct mechanisms on transformed pancreatic cells, inhibition of the YAP/TAZ transcriptional co-activators by metformin may be of paramount importance. In addition, the glucose-lowering properties of metformin together with a decrease of the levels of insulin and IGF as well as its normalizing effects on the gut dysbiosis in diabetic and/or obese patients may contribute to attenuating PDAC development and progression. Further mechanistic studies are needed to dissect the individual contributions of the direct and indirect effects of metformin. However, we postulate that metformin is an exciting and promising drug for PDAC prevention and therapy exactly because it counteracts PDAC development and growth on several levels (direct and indirect).
Acknowledgement:
Guido Eibl is supported by the National Institutes of Health, National Cancer Institute (P01CA236585) and the Hirshberg Foundation for Pancreatic Cancer Research. Enrique Rozengurt is supported by the National Institutes of Health, National Cancer Institute (P01CA236585), National Institute of Allergy and Infectious Diseases (R21AI156592), the Veterans Administration (I01BX003801) and the Hirshberg Foundation for Pancreatic Cancer Research.
Funding:
NIH/NCI (P01CA236585); NIH/NIAID (R21AI156592); VA (I01BX003801).
Footnotes
Conflicts of interest/Competing interests: The authors declare that they have no conflict of interest.
References:
- 1.Siegel RL, Miller KD, Fuchs HE and Jemal A (2021). Cancer Statistics, 2021. CA Cancer J Clin, 71(1), 7–33. 10.3322/caac.21654 [DOI] [PubMed] [Google Scholar]
- 2.Siegel RL, Miller KD and Jemal A (2016). Cancer statistics, 2016. CA Cancer J Clin, 66(1), 7–30. 10.3322/caac.21332 [DOI] [PubMed] [Google Scholar]
- 3.Rahib L, Smith BD, Aizenberg R, Rosenzweig AB, Fleshman JM and Matrisian LM (2014). Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res, 74(11), 2913–2921. 10.1158/0008-5472.CAN-14-0155 [DOI] [PubMed] [Google Scholar]
- 4.Albini A, DeCensi A, Cavalli F and Costa A (2016). Cancer Prevention and Interception: A New Era for Chemopreventive Approaches. Clinical Cancer Research, 22(17), 4322–4327. [DOI] [PubMed] [Google Scholar]
- 5.Meyskens FL, Mukhtar H, Rock CL, Cuzick J, Kensler TW, Yang CS, Ramsey SD, Lippman SM and Alberts DS (2016). Cancer Prevention: Obstacles, Challenges, and the Road Ahead. J Natl Cancer Inst, 108(2), djv309. 10.1093/jnci/djv309 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Maitra A, Fukushima N, Takaori K and Hruban RH (2005). Precursors to invasive pancreatic cancer. Adv Anat Pathol, 12(2), 81–91. [DOI] [PubMed] [Google Scholar]
- 7.Eibl G and Rozengurt E (2019). KRAS, YAP, and obesity in pancreatic cancer: A signaling network with multiple loops. Semin Cancer Biol, 54, 50–62. 10.1016/j.semcancer.2017.10.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Hingorani SR, Petricoin EF, Maitra A, Rajapakse V, King C, Jacobetz MA, Ross S, Conrads TP, Veenstra TD, Hitt BA, Kawaguchi Y, Johann D, Liotta LA, Crawford HC, Putt ME, Jacks T, Wright CV, Hruban RH, Lowy AM, et al. (2003). Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell, 4(6), 437–450. [DOI] [PubMed] [Google Scholar]
- 9.Hingorani SR, Wang L, Multani AS, Combs C, Deramaudt TB, Hruban RH, Rustgi AK, Chang S and Tuveson DA (2005). Trp53R172H and KrasG12D cooperate to promote chromosomal instability and widely metastatic pancreatic ductal adenocarcinoma in mice. Cancer Cell, 7(5), 469–483. 10.1016/j.ccr.2005.04.023 [DOI] [PubMed] [Google Scholar]
- 10.Ying H, Dey P, Yao W, Kimmelman AC, Draetta GF, Maitra A and DePinho RA (2016). Genetics and biology of pancreatic ductal adenocarcinoma. Genes Dev, 30(4), 355–385. 10.1101/gad.275776.115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Collins MA, Bednar F, Zhang Y, Brisset J-C, Galbán S, Galbán CJ, Rakshit S, Flannagan KS, Adsay NV and Pasca di Magliano M (2012). Oncogenic Kras is required for both the initiation and maintenance of pancreatic cancer in mice. J Clin Invest, 122(2), 639–653. 10.1172/JCI59227 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Arslan AA, Helzlsouer KJ, Kooperberg C, Shu X-O, Steplowski E, Bueno-de-Mesquita HB, Fuchs CS, Gross MD, Jacobs EJ, LaCroix AZ, Petersen GM, Stolzenberg-Solomon RZ, Zheng W, Albanes D, Amundadottir L, Bamlet WR, Barricarte A, Bingham SA, Boeing H, et al. (2010). Anthropometric Measures, Body Mass Index, and Pancreatic Cancer: A Pooled Analysis From the Pancreatic Cancer Cohort Consortium (PanScan). Arch Intern Med, 170(9), 791–802. 10.1001/archinternmed.2010.63 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Giovannucci E, Harlan DM, Archer MC, Bergenstal RM, Gapstur SM, Habel LA, Pollak M, Regensteiner JG and Yee D (2010). Diabetes and Cancer: A Consensus Report. CA: A Cancer Journal for Clinicians, 60, 207–221. [DOI] [PubMed] [Google Scholar]
- 14.Rebours V, Gaujoux S, d’Assignies G, Sauvanet A, Ruszniewski P, Lévy P, Paradis V, Bedossa P and Couvelard A (2015). Obesity and Fatty Pancreatic Infiltration Are Risk Factors for Pancreatic Precancerous Lesions (PanIN). Clinical Cancer Research, 21(15), 3522–3528. 10.1158/1078-0432.ccr-14-2385 [DOI] [PubMed] [Google Scholar]
- 15.Lauby-Secretan B, Scoccianti C, Loomis D, Grosse Y, Bianchini F and Straif K (2016). Body Fatness and Cancer — Viewpoint of the IARC Working Group. New Engl J Med, 375(8), 794–798. 10.1056/NEJMsr1606602 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bethea TN, Kitahara CM, Sonderman J, Patel AV, Harvey C, Knutsen SF, Park Y, Park SY, Fraser GE, Jacobs EJ, Purdue MP, Stolzenberg-Solomon RZ, Gillanders EM, Blot WJ, Palmer JR and Kolonel LN (2014). A pooled analysis of body mass index and pancreatic cancer mortality in African Americans. Cancer Epidemiol Biomarkers Prev, 23(10), 2119–2125. 10.1158/1055-9965.EPI-14-0422 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Lega IC and Lipscombe LL (2019). Review: Diabetes, Obesity, and Cancer—Pathophysiology and Clinical Implications. Endocr Rev, 41(1), 33–52. 10.1210/endrev/bnz014 [DOI] [PubMed] [Google Scholar]
- 18.Dawson DW, Hertzer K, Moro A, Donald G, Chang HH, Go VL, Pandol SJ, Lugea A, Gukovskaya AS, Li G, Hines OJ, Rozengurt E and Eibl GE (2013). High Fat, High Calorie Diet Promotes Early Pancreatic Neoplasia in the Conditional KrasG12D Mouse Model. Cancer Prev Res (Phila), 6, 1064–1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chang H-H, Moro A, Takakura K, Su H-Y, Mo A, Nakanishi M, Waldron RT, French SW, Dawson DW, Hines OJ, Li G, Go VLW, Sinnett-Smith J, Pandol SJ, Lugea A, Gukovskaya AS, Duff MO, Rosenberg DW, Rozengurt E, et al. (2017). Incidence of pancreatic cancer is dramatically increased by a high fat, high calorie diet in KrasG12D mice. PLoS ONE, 12(9), e0184455. 10.1371/journal.pone.0184455 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Lashinger LM, Harrison LM, Rasmussen AJ, Logsdon CD, Fischer SM, McArthur MJ and Hursting SD (2013). Dietary energy balance modulation of Kras- and Ink4a/Arf+/−-driven pancreatic cancer: the role of insulin-like growth factor-I. Cancer Prev Res (Phila), 6(10), 1046–1055. 10.1158/1940-6207.CAPR-13-0185 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Philip B, Roland CL, Daniluk J, Liu Y, Chatterjee D, Gomez SB, Ji B, Huang H, Wang H, Fleming JB, Logsdon CD and Cruz-Monserrate Z (2013). A high-fat diet activates oncogenic Kras and COX2 to induce development of pancreatic ductal adenocarcinoma in mice. Gastroenterology, 145(6), 1449–1458. 10.1053/j.gastro.2013.08.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chung KM, Singh J, Lawres L, Dorans KJ, Garcia C, Burkhardt DB, Robbins R, Bhutkar A, Cardone R, Zhao X, Babic A, Vayrynen SA, Dias Costa A, Nowak JA, Chang DT, Dunne RF, Hezel AF, Koong AC, Wilhelm JJ, et al. (2020). Endocrine-Exocrine Signaling Drives Obesity-Associated Pancreatic Ductal Adenocarcinoma. Cell, 181(4), 832–847 e818. 10.1016/j.cell.2020.03.062 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Flier JS (2004). Obesity wars: molecular progress confronts an expanding epidemic. Cell, 116(2), 337–350. [DOI] [PubMed] [Google Scholar]
- 24.Calle EE and Kaaks R (2004). Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat Rev Cancer, 4(8), 579–591. 10.1038/nrc1408 [DOI] [PubMed] [Google Scholar]
- 25.Calle EE and Thun MJ (2004). Obesity and cancer. Oncogene, 23(38), 6365–6378. [DOI] [PubMed] [Google Scholar]
- 26.Muoio DM and Newgard CB (2006). Obesity-Related Derangements in Metabolic Regulation. Annu Rev Biochem, 75, 367–401. [DOI] [PubMed] [Google Scholar]
- 27.Abbruzzese JL, Andersen DK, Borrebaeck CAK, Chari ST, Costello E, Cruz-Monserrate Z, Eibl G, Engleman EG, Fisher WE, Habtezion A, Kim SK, Korc M, Logsdon C, Lyssiotis CA, Pandol SJ, Rustgi A, Wolfe BM, Zheng L and Powers AC (2018). The Interface of Pancreatic Cancer With Diabetes, Obesity, and Inflammation: Research Gaps and Opportunities: Summary of a National Institute of Diabetes and Digestive and Kidney Diseases Workshop. Pancreas, 47(5), 516–525. 10.1097/MPA.0000000000001037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Andersen DK, Andren-Sandberg A, Duell EJ, Goggins M, Korc M, Petersen GM, Smith JP and Whitcomb DC (2013). Pancreatitis-diabetes-pancreatic cancer: summary of an NIDDK-NCI workshop. Pancreas, 42(8), 1227–1237. 10.1097/MPA.0b013e3182a9ad9d [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Andersen DK, Korc M, Petersen GM, Eibl G, Li D, Rickels MR, Chari ST and Abbruzzese JL (2017). Diabetes, Pancreatogenic Diabetes, and Pancreatic Cancer. Diabetes, 66(5), 1103–1110. 10.2337/db16-1477 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Eibl G, Cruz-Monserrate Z, Korc M, Petrov MS, Goodarzi MO, Fisher WE, Habtezion A, Lugea A, Pandol SJ, Hart PA, Andersen DK, Consortium for the Study of Chronic Pancreatitis, D. and Pancreatic, C. (2018). Diabetes Mellitus and Obesity as Risk Factors for Pancreatic Cancer. J Acad Nutr Diet, 118(4), 555–567. 10.1016/j.jand.2017.07.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Kahn BB, Alquier T, Carling D and Hardie DG (2005). AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab, 1(1), 15–25. 10.1016/j.cmet.2004.12.003 [DOI] [PubMed] [Google Scholar]
- 32.Foretz M, Hébrard S, Leclerc J, Zarrinpashneh E, Soty M, Mithieux G, Sakamoto K, Andreelli F and Viollet B (2010). Metformin inhibits hepatic gluconeogenesis in mice independently of the LKB1/AMPK pathway via a decrease in hepatic energy state. J Clin Invest, 120(7), 2355–2369. 10.1172/jci40671 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wu T, Horowitz M and Rayner CK (2017). New insights into the anti-diabetic actions of metformin: from the liver to the gut. Expert Rev Gastroenterol Hepatol, 11(2), 157–166. 10.1080/17474124.2017.1273769 [DOI] [PubMed] [Google Scholar]
- 34.Rena G, Hardie DG and Pearson ER (2017). The mechanisms of action of metformin. Diabetologia, 60(9), 1577–1585. 10.1007/s00125-017-4342-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Duca FA, Côté CD, Rasmussen BA, Zadeh-Tahmasebi M, Rutter GA, Filippi BM and Lam TK (2015). Metformin activates a duodenal Ampk-dependent pathway to lower hepatic glucose production in rats. Nat Med, 21(5), 506–511. 10.1038/nm.3787 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Li D, Yeung SC, Hassan MM, Konopleva M and Abbruzzese JL (2009). Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology, 137(2), 482–488. 10.1053/j.gastro.2009.04.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.de Jong RG, Burden AM, de Kort S, van Herk-Sukel MP, Vissers PA, Janssen PK, Haak HR, Masclee AA, de Vries F and Janssen-Heijnen ML (2017). No Decreased Risk of Gastrointestinal Cancers in Users of Metformin in The Netherlands; A Time-Varying Analysis of Metformin Exposure. Cancer Prev Res (Phila), 10(5), 290–297. 10.1158/1940-6207.CAPR-16-0277 [DOI] [PubMed] [Google Scholar]
- 38.Decensi A, Puntoni M, Goodwin P, Cazzaniga M, Gennari A, Bonanni B and Gandini S (2010). Metformin and cancer risk in diabetic patients: a systematic review and meta-analysis. Cancer Prev Res (Phila), 3(11), 1451–1461. 10.1158/1940-6207.CAPR-10-0157 [DOI] [PubMed] [Google Scholar]
- 39.Zhang P, Li H, Tan X, Chen L and Wang S (2013). Association of metformin use with cancer incidence and mortality: a meta-analysis. Cancer Epidemiol, 37(3), 207–218. 10.1016/j.canep.2012.12.009 [DOI] [PubMed] [Google Scholar]
- 40.Yu H, Zhong X, Gao P, Shi J, Wu Z, Guo Z, Wang Z and Song Y (2019). The Potential Effect of Metformin on Cancer: An Umbrella Review. Front Endocrinol (Lausanne), 10, 617. 10.3389/fendo.2019.00617 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Sadeghi N, Abbruzzese JL, Yeung SC, Hassan M and Li D (2012). Metformin use is associated with better survival of diabetic patients with pancreatic cancer. Clin Cancer Res, 18(10), 2905–2912. 10.1158/1078-0432.CCR-11-2994 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Jang WI, Kim MS, Kang SH, Jo AJ, Kim YJ, Tchoe HJ, Park CM, Kim HJ, Choi JA, Choi HJ, Paik EK, Seo YS, Yoo HJ, Kang JK, Han CJ, Kim YJ, Kim SB and Ko MJ (2017). 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, 8(6), 9587–9596. 10.18632/oncotarget.14525 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Chaiteerakij R, Petersen GM, Bamlet WR, Chaffee KG, Zhen DB, Burch PA, Leof ER, Roberts LR and Oberg AL (2016). Metformin Use and Survival of Patients With Pancreatic Cancer: A Cautionary Lesson. J Clin Oncol, 34(16), 1898–1904. 10.1200/JCO.2015.63.3511 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kordes S, Pollak MN, Zwinderman AH, Mathot RA, Weterman MJ, Beeker A, Punt CJ, Richel DJ and Wilmink JW (2015). Metformin in patients with advanced pancreatic cancer: a double-blind, randomised, placebo-controlled phase 2 trial. Lancet Oncol, 16(7), 839–847. 10.1016/S1470-2045(15)00027-3 [DOI] [PubMed] [Google Scholar]
- 45.Zhou PT, Li B, Liu FR, Zhang MC, Wang Q, Li YY, Xu C, Liu YH, Yao Y and Li D (2017). Metformin is associated with survival benefit in pancreatic cancer patients with diabetes: a systematic review and meta-analysis. Oncotarget, 8(15), 25242–25250. 10.18632/oncotarget.15692 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Shi YQ, Zhou XC, Du P, Yin MY, Xu L, Chen WJ and Xu CF (2020). Relationships are between metformin use and survival in pancreatic cancer patients concurrent with diabetes: A systematic review and meta-analysis. Medicine (Baltimore), 99(37), e21687. 10.1097/MD.0000000000021687 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Yoo D, Kim N, Hwang DW, Song KB, Lee JH, Lee W, Kwon J, Park Y, Hong S, Lee JW, Hwang K, Shin D, Tak E and Kim SC (2020). Association between Metformin Use and Clinical Outcomes Following Pancreaticoduodenectomy in Patients with Type 2 Diabetes and Pancreatic Ductal Adenocarcinoma. J Clin Med, 9(6), E1953. 10.3390/jcm9061953 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Wei M, Liu Y, Bi Y and Zhang ZJ (2019). Metformin and pancreatic cancer survival: Real effect or immortal time bias? Int J Cancer, 145(7), 1822–1828. 10.1002/ijc.32254 [DOI] [PubMed] [Google Scholar]
- 49.Tamburrino D, Guarneri G, Pagnanelli M, Crippa S, Partelli S, Belfiori G, Capurso G and Falconi M (2021). Chemopreventive Agents After Pancreatic Resection for Ductal Adenocarcinoma: Legend or Scientific Evidence? Ann Surg Oncol, 28(4), 2312–2322. 10.1245/s10434-020-09097-y [DOI] [PubMed] [Google Scholar]
- 50.Kim J, Bae YJ, Lee JW, Kim YS, Kim Y, You HS, Kim HS, Choi EA, Han YE and Kang HT (2021). Metformin use in cancer survivors with diabetes reduces all-cause mortality, based on the Korean National Health Insurance Service between 2002 and 2015. Medicine (Baltimore), 100(11), e25045. 10.1097/MD.0000000000025045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Li X, Li T, Liu Z, Gou S and Wang C (2017). The effect of metformin on survival of patients with pancreatic cancer: a meta-analysis. Sci Rep, 7(1), 5825. 10.1038/s41598-017-06207-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Schneider MB, Matsuzaki H, Haorah J, Ulrich A, Standop J, Ding XZ, Adrian TE and Pour PM (2001). Prevention of pancreatic cancer induction in hamsters by metformin. Gastroenterology, 120(5), 1263–1270. 10.1053/gast.2001.23258 [DOI] [PubMed] [Google Scholar]
- 53.Cifarelli V, Lashinger LM, Devlin KL, Dunlap SM, Huang J, Kaaks R, Pollak MN and Hursting SD (2015). Metformin and Rapamycin Reduce Pancreatic Cancer Growth in Obese Prediabetic Mice by Distinct MicroRNA-Regulated Mechanisms. Diabetes, 64(5), 1632–1642. 10.2337/db14-1132 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Yue W, Zheng X, Lin Y, Yang CS, Xu Q, Carpizo D, Huang H, DiPaola RS and Tan XL (2015). Metformin combined with aspirin significantly inhibit pancreatic cancer cell growth in vitro and in vivo by suppressing anti-apoptotic proteins Mcl-1 and Bcl-2. Oncotarget, 6(25), 21208–21224. 10.18632/oncotarget.4126 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Incio J, Suboj P, Chin SM, Vardam-Kaur T, Liu H, Hato T, Babykutty S, Chen I, Deshpande V, Jain RK and Fukumura D (2015). Metformin Reduces Desmoplasia in Pancreatic Cancer by Reprogramming Stellate Cells and Tumor-Associated Macrophages. PLoS One, 10(12), e0141392. 10.1371/journal.pone.0141392 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kato K, Iwama H, Yamashita T, Kobayashi K, Fujihara S, Fujimori T, Kamada H, Kobara H and Masaki T (2016). The anti-diabetic drug metformin inhibits pancreatic cancer cell proliferation in vitro and in vivo: Study of the microRNAs associated with the antitumor effect of metformin. Oncol Rep, 35(3), 1582–1592. 10.3892/or.2015.4496 [DOI] [PubMed] [Google Scholar]
- 57.Shi Y, He Z, Jia Z and Xu C (2016). Inhibitory effect of metformin combined with gemcitabine on pancreatic cancer cells in vitro and in vivo. Mol Med Rep, 14(4), 2921–2928. 10.3892/mmr.2016.5592 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wang C, Zhang T, Liao Q, Dai M, Guo J, Yang X, Tan W, Lin D, Wu C and Zhao Y (2021). Metformin inhibits pancreatic cancer metastasis caused by SMAD4 deficiency and consequent HNF4G upregulation. Protein Cell, 12(2), 128–144. 10.1007/s13238-020-00760-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Rajeshkumar NV, Yabuuchi S, Pai SG, De Oliveira E, Kamphorst JJ, Rabinowitz JD, Tejero H, Al-Shahrour F, Hidalgo M, Maitra A and Dang CV (2017). Treatment of Pancreatic Cancer Patient-Derived Xenograft Panel with Metabolic Inhibitors Reveals Efficacy of Phenformin. Clin Cancer Res, 23(18), 5639–5647. 10.1158/1078-0432.CCR-17-1115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Lipner MB, Marayati R, Deng Y, Wang X, Raftery L, O’Neil BH and Yeh JJ (2016). Metformin Treatment Does Not Inhibit Growth of Pancreatic Cancer Patient-Derived Xenografts. PLoS One, 11(1), e0147113. 10.1371/journal.pone.0147113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Kisfalvi K, Eibl G, Sinnett-Smith J and Rozengurt E (2009). Metformin disrupts crosstalk between G protein-coupled receptor and insulin receptor signaling systems and inhibits pancreatic cancer growth. Cancer Res, 69(16), 6539–6545. 10.1158/0008-5472.CAN-09-0418 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Kisfalvi K, Moro A, Sinnett-Smith J, Eibl G and Rozengurt E (2013). Metformin inhibits the growth of human pancreatic cancer xenografts. Pancreas, 42(5), 781–785. 10.1097/MPA.0b013e31827aec40 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Tan XL, Bhattacharyya KK, Dutta SK, Bamlet WR, Rabe KG, Wang E, Smyrk TC, Oberg AL, Petersen GM and Mukhopadhyay D (2015). Metformin suppresses pancreatic tumor growth with inhibition of NFkappaB/STAT3 inflammatory signaling. Pancreas, 44(4), 636–647. 10.1097/MPA.0000000000000308 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Chen K, Qian W, Jiang Z, Cheng L, Li J, Sun L, Zhou C, Gao L, Lei M, Yan B, Cao J, Duan W and Ma Q (2017). Metformin suppresses cancer initiation and progression in genetic mouse models of pancreatic cancer. Mol Cancer, 16(1), 131. 10.1186/s12943-017-0701-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Qian W, Li J, Chen K, Jiang Z, Cheng L, Zhou C, Yan B, Cao J, Ma Q and Duan W (2018). Metformin suppresses tumor angiogenesis and enhances the chemosensitivity of gemcitabine in a genetically engineered mouse model of pancreatic cancer. Life Sci, 208, 253–261. 10.1016/j.lfs.2018.07.046 [DOI] [PubMed] [Google Scholar]
- 66.Chang HH, Moro A, Chou CEN, Dawson DW, French S, Schmidt AI, Sinnett-Smith J, Hao F, Hines OJ, Eibl G and Rozengurt E (2018). Metformin Decreases the Incidence of Pancreatic Ductal Adenocarcinoma Promoted by Diet-induced Obesity in the Conditional KrasG12D Mouse Model. Sci Rep, 8(1), 5899. 10.1038/s41598-018-24337-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Dong TS, Chang HH, Hauer M, Lagishetty V, Katzka W, Rozengurt E, Jacobs JP and Eibl G (2019). Metformin alters the duodenal microbiome and decreases the incidence of pancreatic ductal adenocarcinoma promoted by diet-induced obesity. Am J Physiol Gastrointest Liver Physiol, 317(6), G763–G772. 10.1152/ajpgi.00170.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Eibl G (2020). Endocrine-exocrine signals in obesity-associated pancreatic cancer. Nat Rev Gastroenterol Hepatol, 17(8), 455–456. 10.1038/s41575-020-0324-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Foretz M, Guigas B, Bertrand L, Pollak M and Viollet B (2014). Metformin: from mechanisms of action to therapies. Cell Metab, 20(6), 953–966. 10.1016/j.cmet.2014.09.018 [DOI] [PubMed] [Google Scholar]
- 70.Pollak M (2017). The effects of metformin on gut microbiota and the immune system as research frontiers. Diabetologia, 60(9), 1662–1667. 10.1007/s00125-017-4352-x [DOI] [PubMed] [Google Scholar]
- 71.Pollak MN (2012). Investigating metformin for cancer prevention and treatment: the end of the beginning. Cancer Discov, 2(9), 778–790. 10.1158/2159-8290.CD-12-0263 [DOI] [PubMed] [Google Scholar]
- 72.Pollak M (2010). Metformin and other biguanides in oncology: advancing the research agenda. Cancer Prev Res (Phila), 3(9), 1060–1065. 10.1158/1940-6207.CAPR-10-0175 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Coll AP, Chen M, Taskar P, Rimmington D, Patel S, Tadross JA, Cimino I, Yang M, Welsh P, Virtue S, Goldspink DA, Miedzybrodzka EL, Konopka AR, Esponda RR, Huang JT, Tung YCL, Rodriguez-Cuenca S, Tomaz RA, Harding HP, et al. (2020). GDF15 mediates the effects of metformin on body weight and energy balance. Nature, 578(7795), 444–448. 10.1038/s41586-019-1911-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Rozengurt E and Eibl G (2019). Central role of Yes-associated protein and WW-domain-containing transcriptional co-activator with PDZ-binding motif in pancreatic cancer development. World J Gastroenterol, 25(15), 1797–1816. 10.3748/wjg.v25.i15.1797 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Foretz M, Guigas B and Viollet B (2019). Understanding the glucoregulatory mechanisms of metformin in type 2 diabetes mellitus. Nat Rev Endocrinol, 15(10), 569–589. 10.1038/s41574-019-0242-2 [DOI] [PubMed] [Google Scholar]
- 76.Hawley SA, Ross FA, Chevtzoff C, Green KA, Evans A, Fogarty S, Towler MC, Brown LJ, Ogunbayo OA, Evans AM and Hardie DG (2010). Use of cells expressing gamma subunit variants to identify diverse mechanisms of AMPK activation. Cell Metab, 11(6), 554–565. 10.1016/j.cmet.2010.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Ben Sahra I, Laurent K, Loubat A, Giorgetti-Peraldi S, Colosetti P, Auberger P, Tanti JF, Le Marchand-Brustel Y and Bost F (2008). The antidiabetic drug metformin exerts an antitumoral effect in vitro and in vivo through a decrease of cyclin D1 level. Oncogene, 27(25), 3576–3586. 10.1038/sj.onc.1211024 [DOI] [PubMed] [Google Scholar]
- 78.Kalender A, Selvaraj A, Kim SY, Gulati P, Brule S, Viollet B, Kemp BE, Bardeesy N, Dennis P, Schlager JJ, Marette A, Kozma SC and Thomas G (2010). Metformin, independent of AMPK, inhibits mTORC1 in a rag GTPase-dependent manner. Cell Metab, 11(5), 390–401. 10.1016/j.cmet.2010.03.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.LaMoia TE and Shulman GI (2021). Cellular and Molecular Mechanisms of Metformin Action. Endocr Rev, 42(1), 77–96. 10.1210/endrev/bnaa023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Wang LW, Li ZS, Zou DW, Jin ZD, Gao J and Xu GM (2008). Metformin induces apoptosis of pancreatic cancer cells. World J Gastroenterol, 14(47), 7192–7198. 10.3748/wjg.14.7192 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Bao B, Wang Z, Ali S, Ahmad A, Azmi AS, Sarkar SH, Banerjee S, Kong D, Li Y, Thakur S and Sarkar FH (2012). Metformin inhibits cell proliferation, migration and invasion by attenuating CSC function mediated by deregulating miRNAs in pancreatic cancer cells. Cancer Prev Res (Phila), 5(3), 355–364. 10.1158/1940-6207.CAPR-11-0299 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Nair V, Sreevalsan S, Basha R, Abdelrahim M, Abudayyeh A, Rodrigues Hoffman A and Safe S (2014). Mechanism of metformin-dependent inhibition of mammalian target of rapamycin (mTOR) and Ras activity in pancreatic cancer: role of specificity protein (Sp) transcription factors. J Biol Chem, 289(40), 27692–27701. 10.1074/jbc.M114.592576 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Yan Y, Zhou XE, Xu HE and Melcher K (2018). Structure and Physiological Regulation of AMPK. Int J Mol Sci, 19(11), 3534. 10.3390/ijms19113534 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Hardie DG (2015). AMPK: positive and negative regulation, and its role in whole-body energy homeostasis. Curr Opin Cell Biol, 33, 1–7. 10.1016/j.ceb.2014.09.004 [DOI] [PubMed] [Google Scholar]
- 85.Olivier S, Foretz M and Viollet B (2018). Promise and challenges for direct small molecule AMPK activators. Biochem Pharmacol, 153, 147–158. 10.1016/j.bcp.2018.01.049 [DOI] [PubMed] [Google Scholar]
- 86.Hezel AF and Bardeesy N (2008). LKB1; linking cell structure and tumor suppression. Oncogene, 27(55), 6908–6919. 10.1038/onc.2008.342 [DOI] [PubMed] [Google Scholar]
- 87.Bridges HR, Jones AJ, Pollak MN and Hirst J (2014). Effects of metformin and other biguanides on oxidative phosphorylation in mitochondria. Biochem J, 462(3), 475–487. 10.1042/bj20140620 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Pryor R and Cabreiro F (2015). Repurposing metformin: an old drug with new tricks in its binding pockets. Biochem J, 471(3), 307–322. 10.1042/bj20150497 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Hardie DG, Ross FA and Hawley SA (2012). AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nature Reviews Molecular Cell Biology, 13(4), 251–262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Ming M, Sinnett-Smith J, Wang J, Soares HP, Young SH, Eibl G and Rozengurt E (2014). Dose-Dependent AMPK-Dependent and Independent Mechanisms of Berberine and Metformin Inhibition of mTORC1, ERK, DNA Synthesis and Proliferation in Pancreatic Cancer Cells. PLoS ONE, 9(12), e114573. 10.1371/journal.pone.0114573 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sinnett-Smith J, Kisfalvi K, Kui R and Rozengurt E (2013). Metformin inhibition of mTORC1 activation, DNA synthesis and proliferation in pancreatic cancer cells: dependence on glucose concentration and role of AMPK. Biochem Biophys Res Commun, 430(1), 352–357. 10.1016/j.bbrc.2012.11.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Carroll B and Dunlop EA (2017). The lysosome: a crucial hub for AMPK and mTORC1 signalling. Biochem J, 474(9), 1453–1466. 10.1042/BCJ20160780 [DOI] [PubMed] [Google Scholar]
- 93.Taniguchi CM, Emanuelli B and Kahn CR (2006). Critical nodes in signalling pathways: insights into insulin action. Nature Reviews Molecular Cell Biology, 7(2), 85–96. [DOI] [PubMed] [Google Scholar]
- 94.Rozengurt E (2014). Mechanistic target of rapamycin (mTOR): a point of convergence in the action of insulin/IGF-1 and G protein-coupled receptor agonists in pancreatic cancer cells. Front Physiol, 5, 357. 10.3389/fphys.2014.00357 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Rozengurt E, Soares HP and Sinnet-Smith J (2014). Suppression of Feedback Loops Mediated by PI3K/mTOR Induces Multiple Overactivation of Compensatory Pathways: An Unintended Consequence Leading to Drug Resistance. Mol Cancer Ther, 13(11), 2477–2488. 10.1158/1535-7163.mct-14-0330 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Soares HP, Ni Y, Kisfalvi K, Sinnett-Smith J and Rozengurt E (2013). Different patterns of Akt and ERK feedback activation in response to rapamycin, active-site mTOR inhibitors and metformin in pancreatic cancer cells. PLoS One, 8(2), e57289. 10.1371/journal.pone.0057289 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Soares HP, Ming M, Mellon M, Young SH, Han L, Sinnet-Smith J and Rozengurt E (2015). Dual PI3K/mTOR Inhibitors Induce Rapid Overactivation of the MEK/ERK Pathway in Human Pancreatic Cancer Cells through Suppression of mTORC2. Mol Cancer Ther, 14(4), 1014–1023. 10.1158/1535-7163.mct-14-0669 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Lee JW, Park S, Takahashi Y and Wang H-G (2010). The Association of AMPK with ULK1 Regulates Autophagy. PLoS ONE, 5(11), e15394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Saxton RA and Sabatini DM (2017). mTOR Signaling in Growth, Metabolism, and Disease. Cell, 168(6), 960–976. 10.1016/j.cell.2017.02.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Um SH, D’Alessio D and Thomas G (2006). Nutrient overload, insulin resistance, and ribosomal protein S6 kinase 1, S6K1. Cell Metab, 3(6), 393–402. [DOI] [PubMed] [Google Scholar]
- 101.Ali SM and Sabatini DM (2005). Structure of S6 Kinase 1 Determines whether Raptor-mTOR or Rictor-mTOR Phosphorylates Its Hydrophobic Motif Site. Journal of Biological Chemistry, 280, 19445–19448. [DOI] [PubMed] [Google Scholar]
- 102.Long X, Muller F and Avruch J (2004). TOR action in mammalian cells and in Caenorhabditis elegans. Curr Top Microbiol Immunol, 279, 115–138. [DOI] [PubMed] [Google Scholar]
- 103.Armengol G, Rojo F, Castellvi J, Iglesias C, Cuatrecasas M, Pons B, Baselga J and Ramon y Cajal S (2007). 4E-binding protein 1: a key molecular “funnel factor” in human cancer with clinical implications. Cancer Res, 67(16), 7551–7555. [DOI] [PubMed] [Google Scholar]
- 104.Inoki K, Zhu T and Guan KL (2003). TSC2 mediates cellular energy response to control cell growth and survival. Cell, 115(5), 577–590. 10.1016/s0092-8674(03)00929-2 [DOI] [PubMed] [Google Scholar]
- 105.Shaw RJ, Bardeesy N, Manning BD, Lopez L, Kosmatka M, DePinho RA and Cantley LC (2004). The LKB1 tumor suppressor negatively regulates mTOR signaling. Cancer Cell, 6(1), 91–99. 10.1016/j.ccr.2004.06.007 [DOI] [PubMed] [Google Scholar]
- 106.Inoki K, Ouyang H, Zhu T, Lindvall C, Wang Y, Zhang X, Yang Q, Bennett C, Harada Y, Stankunas K, Wang CY, He X, MacDougald OA, You M, Williams BO and Guan KL (2006). TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell, 126(5), 955–968. 10.1016/j.cell.2006.06.055 [DOI] [PubMed] [Google Scholar]
- 107.Gwinn DM, Shackelford DB, Egan DF, Mihaylova MM, Mery A, Vasquez DS, Turk BE and Shaw RJ (2008). AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol Cell, 30(2), 214–226. 10.1016/j.molcel.2008.03.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Tzatsos A and Tsichlis PN (2007). Energy depletion inhibits phosphatidylinositol 3-kinase/Akt signaling and induces apoptosis via AMP-activated protein kinase-dependent phosphorylation of IRS-1 at Ser-794. J Biol Chem, 282(25), 18069–18082. 10.1074/jbc.M610101200 [DOI] [PubMed] [Google Scholar]
- 109.Ning J and Clemmons DR (2010). AMP-activated protein kinase inhibits IGF-I signaling and protein synthesis in vascular smooth muscle cells via stimulation of insulin receptor substrate 1 S794 and tuberous sclerosis 2 S1345 phosphorylation. Mol Endocrinol, 24(6), 1218–1229. 10.1210/me.2009-0474 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Piccolo S, Dupont S and Cordenonsi M (2014). The biology of YAP/TAZ: hippo signaling and beyond. Physiol Rev, 94(4), 1287–1312. 10.1152/physrev.00005.2014 [DOI] [PubMed] [Google Scholar]
- 111.Yu FX, Zhao B and Guan KL (2015). Hippo Pathway in Organ Size Control, Tissue Homeostasis, and Cancer. Cell, 163(4), 811–828. 10.1016/j.cell.2015.10.044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Enzo E, Santinon G, Pocaterra A, Aragona M, Bresolin S, Forcato M, Grifoni D, Pession A, Zanconato F, Guzzo G, Bicciato S and Dupont S (2015). Aerobic glycolysis tunes YAP/TAZ transcriptional activity. EMBO J, 34(10), 1349–1370. 10.15252/embj.201490379 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Wang Z, Wu Y, Wang H, Zhang Y, Mei L, Fang X, Zhang X, Zhang F, Chen H, Liu Y, Jiang Y, Sun S, Zheng Y, Li N and Huang L (2014). Interplay of mevalonate and Hippo pathways regulates RHAMM transcription via YAP to modulate breast cancer cell motility. Proc Natl Acad Sci U S A, 111(1), E89–98. 10.1073/pnas.1319190110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Santinon G, Pocaterra A and Dupont S (2016). Control of YAP/TAZ Activity by Metabolic and Nutrient-Sensing Pathways. Trends Cell Biol, 26(4), 289–299. 10.1016/j.tcb.2015.11.004 [DOI] [PubMed] [Google Scholar]
- 115.Ardestani A, Lupse B and Maedler K (2018). Hippo Signaling: Key Emerging Pathway in Cellular and Whole-Body Metabolism. Trends Endocrinol Metab, 29(7), 492–509. 10.1016/j.tem.2018.04.006 [DOI] [PubMed] [Google Scholar]
- 116.Koo JH and Guan KL (2018). Interplay between YAP/TAZ and Metabolism. Cell Metab, 28(2), 196–206. 10.1016/j.cmet.2018.07.010 [DOI] [PubMed] [Google Scholar]
- 117.Plouffe SW, Lin KC, Moore JL 3rd, Tan FE, Ma S, Ye Z, Qiu Y, Ren B and Guan KL (2018). The Hippo pathway effector proteins YAP and TAZ have both distinct and overlapping functions in the cell. J Biol Chem, 293(28), 11230–11240. 10.1074/jbc.RA118.002715 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Callus BA, Finch-Edmondson ML, Fletcher S and Wilton SD (2019). YAPping about and not forgetting TAZ. FEBS Lett, 593(3), 253–276. 10.1002/1873-3468.13318 [DOI] [PubMed] [Google Scholar]
- 119.Zhang W, Nandakumar N, Shi Y, Manzano M, Smith A, Graham G, Gupta S, Vietsch EE, Laughlin SZ, Wadhwa M, Chetram M, Joshi M, Wang F, Kallakury B, Toretsky J, Wellstein A and Yi C (2014). Downstream of mutant KRAS, the transcription regulator YAP is essential for neoplastic progression to pancreatic ductal adenocarcinoma. Sci Signal, 7(324), ra42. 10.1126/scisignal.2005049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Gruber R, Panayiotou R, Nye E, Spencer-Dene B, Stamp G and Behrens A (2016). YAP1 and TAZ Control Pancreatic Cancer Initiation in Mice by Direct Up-regulation of JAK-STAT3 Signaling. Gastroenterology, 151(3), 526–539. 10.1053/j.gastro.2016.05.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Tu B, Yao J, Ferri-Borgogno S, Zhao J, Chen S, Wang Q, Yan L, Zhou X, Zhu C, Bang S, Chang Q, Bristow CA, Kang Y, Zheng H, Wang H, Fleming JB, Kim M, Heffernan TP, Draetta GF, et al. (2019). YAP1 oncogene is a context-specific driver for pancreatic ductal adenocarcinoma. JCI Insight, 4(21), 10.1172/jci.insight.130811 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Rozengurt E, Sinnett-Smith J and Eibl G (2018). Yes-associated protein (YAP) in pancreatic cancer: at the epicenter of a targetable signaling network associated with patient survival. Signal Transduct Target Ther, 3, 11. 10.1038/s41392-017-0005-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Kapoor A, Yao W, Ying H, Hua S, Liewen A, Wang Q, Zhong Y, Wu CJ, Sadanandam A, Hu B, Chang Q, Chu GC, Al-Khalil R, Jiang S, Xia H, Fletcher-Sananikone E, Lim C, Horwitz GI, Viale A, et al. (2014). Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell, 158(1), 185–197. 10.1016/j.cell.2014.06.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Yang S, Zhang L, Purohit V, Shukla SK, Chen X, Yu F, Fu K, Chen Y, Solheim J, Singh PK, Song W and Dong J (2015). Active YAP promotes pancreatic cancer cell motility, invasion and tumorigenesis in a mitotic phosphorylation-dependent manner through LPAR3. Oncotarget, 6(34), 36019–36031. 10.18632/oncotarget.5935 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Morvaridi S, Dhall D, Greene MI, Pandol SJ and Wang Q (2015). Role of YAP and TAZ in pancreatic ductal adenocarcinoma and in stellate cells associated with cancer and chronic pancreatitis. Sci Rep, 5, 16759. 10.1038/srep16759 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Murakami S, Shahbazian D, Surana R, Zhang W, Chen H, Graham GT, White SM, Weiner LM and Yi C (2017). Yes-associated protein mediates immune reprogramming in pancreatic ductal adenocarcinoma. Oncogene, 36(9), 1232–1244. 10.1038/onc.2016.288 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Mueller S, Engleitner T, Maresch R, Zukowska M, Lange S, Kaltenbacher T, Konukiewitz B, Ollinger R, Zwiebel M, Strong A, Yen HY, Banerjee R, Louzada S, Fu B, Seidler B, Gotzfried J, Schuck K, Hassan Z, Arbeiter A, et al. (2018). Evolutionary routes and KRAS dosage define pancreatic cancer phenotypes. Nature, 554(7690), 62–68. 10.1038/nature25459 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Hajimoradi Javarsiani M, Sajedianfard J and Haghjooy Javanmard S (2020). The effects of metformin on the hippo pathway in the proliferation of melanoma cancer cells: a preclinical study. Arch Physiol Biochem, 1–6. 10.1080/13813455.2020.1760304 [DOI] [PubMed] [Google Scholar]
- 129.Liu J, Li J, Chen H, Wang R, Li P, Miao Y and Liu P (2020). Metformin suppresses proliferation and invasion of drug-resistant breast cancer cells by activation of the Hippo pathway. J Cell Mol Med, 24(10), 5786–5796. 10.1111/jcmm.15241 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Jin D, Guo J, Wu Y, Chen W, Du J, Yang L, Wang X, Gong K, Dai J, Miao S, Li X and Su G (2020). Metformin-repressed miR-381-YAP-snail axis activity disrupts NSCLC growth and metastasis. J Exp Clin Cancer Res, 39(1), 6. 10.1186/s13046-019-1503-6 [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 131.Wu Y, Zheng Q, Li Y, Wang G, Gao S, Zhang X, Yan X, Zhang X, Xie J, Wang Y, Sun X, Meng X, Yin B and Wang B (2019). Metformin targets a YAP1-TEAD4 complex via AMPKalpha to regulate CCNE1/2 in bladder cancer cells. J Exp Clin Cancer Res, 38(1), 376. 10.1186/s13046-019-1346-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Yuan X, Wei W, Bao Q, Chen H, Jin P and Jiang W (2018). Metformin inhibits glioma cells stemness and epithelial-mesenchymal transition via regulating YAP activity. Biomed Pharmacother, 102, 263–270. 10.1016/j.biopha.2018.03.031 [DOI] [PubMed] [Google Scholar]
- 133.Tian Y, Tang B, Wang C, Sun D, Zhang R, Luo N, Han Z, Liang R, Gao Z and Wang L (2016). Metformin mediates resensitivity to 5-fluorouracil in hepatocellular carcinoma via the suppression of YAP. Oncotarget, 7(29), 46230–46241. 10.18632/oncotarget.10079 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Mo JS, Meng Z, Kim YC, Park HW, Hansen CG, Kim S, Lim DS and Guan KL (2015). Cellular energy stress induces AMPK-mediated regulation of YAP and the Hippo pathway. Nat Cell Biol, 17(4), 500–510. 10.1038/ncb3111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Wang W, Xiao ZD, Li X, Aziz KE, Gan B, Johnson RL and Chen J (2015). AMPK modulates Hippo pathway activity to regulate energy homeostasis. Nat Cell Biol, 17(4), 490–499. 10.1038/ncb3113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Omkumar RV, Darnay BG and Rodwell VW (1994). Modulation of Syrian hamster 3-hydroxy-3-methylglutaryl-CoA reductase activity by phosphorylation. Role of serine 871. J Biol Chem, 269(9), 6810–6814. [PubMed] [Google Scholar]
- 137.Moroishi T, Hansen CG and Guan KL (2015). The emerging roles of YAP and TAZ in cancer. Nat Rev Cancer, 15(2), 73–79. 10.1038/nrc3876 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.DeRan M, Yang J, Shen CH, Peters EC, Fitamant J, Chan P, Hsieh M, Zhu S, Asara JM, Zheng B, Bardeesy N, Liu J and Wu X (2014). Energy stress regulates hippo-YAP signaling involving AMPK-mediated regulation of angiomotin-like 1 protein. Cell Rep, 9(2), 495–503. 10.1016/j.celrep.2014.09.036 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Shaw RJ, Lamia KA, Vasquez D, Koo SH, Bardeesy N, Depinho RA, Montminy M and Cantley LC (2005). The kinase LKB1 mediates glucose homeostasis in liver and therapeutic effects of metformin. Science, 310(5754), 1642–1646. 10.1126/science.1120781 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Berker B, Emral R, Demirel C, Corapcioglu D, Unlu C and Kose K (2004). Increased insulin-like growth factor-I levels in women with polycystic ovary syndrome, and beneficial effects of metformin therapy. Gynecol Endocrinol, 19(3), 125–133. 10.1080/09513590400007309 [DOI] [PubMed] [Google Scholar]
- 141.Goodwin PJ, Pritchard KI, Ennis M, Clemons M, Graham M and Fantus IG (2008). Insulin-lowering effects of metformin in women with early breast cancer. Clin Breast Cancer, 8(6), 501–505. 10.3816/CBC.2008.n.060 [DOI] [PubMed] [Google Scholar]
- 142.Mutgan AC, Besikcioglu HE, Wang S, Friess H, Ceyhan GO and Demir IE (2018). Insulin/IGF-driven cancer cell-stroma crosstalk as a novel therapeutic target in pancreatic cancer. Mol Cancer, 17(1), 66. 10.1186/s12943-018-0806-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Zheng Y, Wu C, Yang J, Zhao Y, Jia H, Xue M, Xu D, Yang F, Fu D, Wang C, Hu B, Zhang Z, Li T, Yan S, Wang X, Nelson PJ, Bruns C, Qin L and Dong Q (2020). Insulin-like growth factor 1-induced enolase 2 deacetylation by HDAC3 promotes metastasis of pancreatic cancer. Signal Transduct Target Ther, 5(1), 53. 10.1038/s41392-020-0146-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Trajkovic-Arsic M, Kalideris E and Siveke JT (2013). The role of insulin and IGF system in pancreatic cancer. J Mol Endocrinol, 50(3), R67–74. 10.1530/JME-12-0259 [DOI] [PubMed] [Google Scholar]
- 145.Renehan AG, Frystyk J and Flyvbjerg A (2006). Obesity and cancer risk: the role of the insulin-IGF axis. Trends Endocrinol Metab, 17(8), 328–336. 10.1016/j.tem.2006.08.006 [DOI] [PubMed] [Google Scholar]
- 146.Subramani R, Lopez-Valdez R, Arumugam A, Nandy S, Boopalan T and Lakshmanaswamy R (2014). Targeting insulin-like growth factor 1 receptor inhibits pancreatic cancer growth and metastasis. PLoS One, 9(5), e97016. 10.1371/journal.pone.0097016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Gong J, Robbins LA, Lugea A, Waldron RT, Jeon CY and Pandol SJ (2014). Diabetes, pancreatic cancer, and metformin therapy. Front Physiol, 5, 426. 10.3389/fphys.2014.00426 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Hao F, Xu Q, Zhao Y, Stevens JV, Young SH, Sinnett-Smith J and Rozengurt E (2017). Insulin Receptor and GPCR Crosstalk Stimulates YAP via PI3K and PKD in Pancreatic Cancer Cells. Mol Cancer Res, 15(7), 929–941. 10.1158/1541-7786.MCR-17-0023 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Rozengurt E, Sinnett-Smith J and Kisfalvi K (2010). 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, 16(9), 2505–2511. 10.1158/1078-0432.CCR-09-2229 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Liu SS, Ma XF, Zhao J, Du SX, Zhang J, Dong MZ and Xin YN (2020). Association between nonalcoholic fatty liver disease and extrahepatic cancers: a systematic review and meta-analysis. Lipids Health Dis, 19(1), 118. 10.1186/s12944-020-01288-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Chang CF, Tseng YC, Huang HH, Shih YL, Hsieh TY and Lin HH (2018). Exploring the relationship between nonalcoholic fatty liver disease and pancreatic cancer by computed tomographic survey. Intern Emerg Med, 13(2), 191–197. 10.1007/s11739-017-1774-x [DOI] [PubMed] [Google Scholar]
- 152.Murphy N, Jenab M and Gunter MJ (2018). Adiposity and gastrointestinal cancers: epidemiology, mechanisms and future directions. Nat Rev Gastroenterol Hepatol, 15(11), 659–670. 10.1038/s41575-018-0038-1 [DOI] [PubMed] [Google Scholar]
- 153.Maruvada P, Leone V, Kaplan LM and Chang EB (2017). The Human Microbiome and Obesity: Moving beyond Associations. Cell Host Microbe, 22(5), 589–599. 10.1016/j.chom.2017.10.005 [DOI] [PubMed] [Google Scholar]
- 154.Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER and Gordon JI (2006). An obesity-associated gut microbiome with increased capacity for energy harvest. Nature, 444(7122), 1027–1031. 10.1038/nature05414 [DOI] [PubMed] [Google Scholar]
- 155.He L (2020). Metformin and Systemic Metabolism. Trends Pharmacol Sci, 41(11), 868–881. 10.1016/j.tips.2020.09.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S, Prifti E, Vieira-Silva S, Gudmundsdottir V, Pedersen HK, Arumugam M, Kristiansen K, Voigt AY, Vestergaard H, Hercog R, Costea PI, Kultima JR, Li J, Jorgensen T, et al. (2015). Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature, 528(7581), 262–266. 10.1038/nature15766 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Wu H, Esteve E, Tremaroli V, Khan MT, Caesar R, Manneras-Holm L, Stahlman M, Olsson LM, Serino M, Planas-Felix M, Xifra G, Mercader JM, Torrents D, Burcelin R, Ricart W, Perkins R, Fernandez-Real JM and Backhed F (2017). Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat Med, 23(7), 850–858. 10.1038/nm.4345 [DOI] [PubMed] [Google Scholar]
- 158.Shin NR, Lee JC, Lee HY, Kim MS, Whon TW, Lee MS and Bae JW (2014). An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut, 63(5), 727–735. 10.1136/gutjnl-2012-303839 [DOI] [PubMed] [Google Scholar]
- 159.Riquelme E, Zhang Y, Zhang L, Montiel M, Zoltan M, Dong W, Quesada P, Sahin I, Chandra V, San Lucas A, Scheet P, Xu H, Hanash SM, Feng L, Burks JK, Do KA, Peterson CB, Nejman D, Tzeng CD, et al. (2019). Tumor Microbiome Diversity and Composition Influence Pancreatic Cancer Outcomes. Cell, 178(4), 795–806 e712. 10.1016/j.cell.2019.07.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Sethi V, Kurtom S, Tarique M, Lavania S, Malchiodi Z, Hellmund L, Zhang L, Sharma U, Giri B, Garg B, Ferrantella A, Vickers SM, Banerjee S, Dawra R, Roy S, Ramakrishnan S, Saluja A and Dudeja V (2018). Gut Microbiota Promotes Tumor Growth in Mice by Modulating Immune Response. Gastroenterology, 155(1), 33–37 e36. 10.1053/j.gastro.2018.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Pushalkar S, Hundeyin M, Daley D, Zambirinis CP, Kurz E, Mishra A, Mohan N, Aykut B, Usyk M, Torres LE, Werba G, Zhang K, Guo Y, Li Q, Akkad N, Lall S, Wadowski B, Gutierrez J, Kochen Rossi JA, et al. (2018). The Pancreatic Cancer Microbiome Promotes Oncogenesis by Induction of Innate and Adaptive Immune Suppression. Cancer Discov, 8(4), 403–416. 10.1158/2159-8290.CD-17-1134 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Mannucci E, Tesi F, Bardini G, Ognibene A, Petracca MG, Ciani S, Pezzatini A, Brogi M, Dicembrini I, Cremasco F, Messeri G and Rotella CM (2004). Effects of metformin on glucagon-like peptide-1 levels in obese patients with and without Type 2 diabetes. Diabetes Nutr Metab, 17(6), 336–342. [PubMed] [Google Scholar]
- 163.Napolitano A, Miller S, Nicholls AW, Baker D, Van Horn S, Thomas E, Rajpal D, Spivak A, Brown JR and Nunez DJ (2014). Novel gut-based pharmacology of metformin in patients with type 2 diabetes mellitus. PLoS One, 9(7), e100778. 10.1371/journal.pone.0100778 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Duca FA, Bauer PV, Hamr SC and Lam TK (2015). Glucoregulatory Relevance of Small Intestinal Nutrient Sensing in Physiology, Bariatric Surgery, and Pharmacology. Cell Metab, 22(3), 367–380. 10.1016/j.cmet.2015.07.003 [DOI] [PubMed] [Google Scholar]
- 165.Yang M, Darwish T, Larraufie P, Rimmington D, Cimino I, Goldspink DA, Jenkins B, Koulman A, Brighton CA, Ma M, Lam BYH, Coll AP, O’Rahilly S, Reimann F and Gribble FM (2021). Inhibition of mitochondrial function by metformin increases glucose uptake, glycolysis and GDF-15 release from intestinal cells. Sci Rep, 11(1), 2529. 10.1038/s41598-021-81349-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
