Review Article
New Onset Diabetes Vs Long-Standing Diabetes Pathogenesis About Pancreatic Cancer: A Narrative Review
- Hawraa Tofaili 1,5
- Safaa Joumaa 1,5
- Rawan Tabaja 1, 5
- Nada Mchawrab 1,5
- Nour Soloh 1,5
- Zeinab Karaki 1,5
- Andrea Eid 2,5
- Hadi Abozeid 3,5
- Ahmad Jaber 4,5
- Jana Jaber 1,5
1Lebanese University, Faculty of Medical Sciences, Beirut, Lebanon.
2Holy Spirit University of Kaslik, Faculty of Medicine and Medical Sciences, Lebanon.
3Lebanese University, Faculty of Sciences, Beirut, Lebanon.
4Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Byblos, Lebanon.
5Medical Learning Skills Academy, Beirut, Lebanon.
*Corresponding Author: Jana Jaber, Lebanese University, Faculty of Medical Sciences, Beirut, Lebanon.
Citation: Tofaili H, Joumaa S, Tabaja R, Mchawrab N, Jaber J, et, al. (2024). New Onset Diabetes Vs Long-Standing Diabetes Pathogenesis About Pancreatic Cancer: A Narrative Review. Journal of Endocrinology and Diabetes Research, BioRes Scientia Publishers. 2(1):1-17. DOI: 10.59657/2996-3095.brs.24.009
Copyright: © 2024 Jana Jaber, this is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Received: December 06, 2023 | Accepted: December 28, 2023 | Published: January 08, 2024
Abstract
Pancreatic cancer (PC) is a deadly malignancy with a 5-year survival rate of less than 10%. More than 85% of PC patients have impaired glucose tolerance, indicating a complex bidirectional relationship. Diabetes can be both a risk factor and a complication of PC: the link between these two diseases is now a high priority for research. T2DM-associated PC is caused by insulin resistance, hyperinsulinemia, hyperglycemia, and chronic inflammation. Furthermore, recent research suggests that anti-diabetic medications may influence the risk of pancreatic cancer in type 2 diabetes; specifically, metformin reduces the risk of PC, whereas insulin therapy increases the risk. Diabetes also improves after pancreatic resection, implying that diabetes is caused by pancreatic cancer. Compared to the general population, people with new-onset diabetes have a 6-to-8-fold increased risk of developing pancreatic cancer. Diabetes of new-onset is an important screening tool for asymptomatic pancreatic cancer patients, and it improves pancreatic cancer survival. A deeper comprehension of the underlying pathophysiological mechanisms that distinguish T2DM from new-onset diabetes can help distinguish the two conditions. Additionally, evaluating the clinical features that set the two apart can help identify high-risk individuals for targeted screenings of suspected PC, which may help detect a tumor at a potentially treatable stage.
Keywords: diabetes mellitus; pancreatic cancer; inflammation; biomarkers; treatment
Introduction
Despite having a relatively low incidence of 12.9 per 100,000 person-years, pancreatic cancer (PC) is the seventh most common cause of cancer-related death worldwide, according to the World Health Organization (WHO) [1]. Most of these tumors are of the ductal adenocarcinoma histological subtype (PDAC), originating primarily from ductal epithelial cells of the exocrine pancreas [2]. Age, gender, and race/ethnicity all impact PC incidence and death rates. The disease is rare in young adults (45 years old), peaks in the seventh and eighth decades, and is slightly more common in men (M/F: 1.3/1) and Black individuals [3, 4]. With a 5-year survival rate of less than 10% and a high recurrence rate even after potentially curative resection, PC was associated with an extremely poor prognosis [5]. The 5-year survival rate for patients who had their entire tumor completely removed was only up to 25%. A number of risk factors have been found to contribute to the burden of PC, including smoking, genetic predisposition, and advanced age [6]. Diabetes is also regarded as a risk factor for PC, as patients with the condition have a risk of PC that is more than twice as high as that of people without the condition [7]. Actually, diabetes affects about half of patients with sporadic pancreatic cancer. Diabetes is thought to be secondary to malignancy, having developed 2-3 years prior to cancer diagnosis, and is diagnosed concurrently with or shortly after cancer diagnosis in nearly 50% of diabetic pancreatic cancer patients [8]. Numerous recent studies have demonstrated the complex and reciprocal nature of the relationship between PC and diabetes, and current efforts are being made to differentiate the relationship between PC and the various forms of diabetes that are involved [9]. According to the American Diabetes Association, there are several forms of diabetes; however, two of them—type 2 diabetes (T2DM) and type 3c diabetes—need to be addressed in relation to PC. Chronic non-communicable type 2 diabetes (T2DM) is typified by persistent insulin resistance (IR) combined with impaired insulin secretion as a result of beta cell dysfunction [10]. This leads to hyperglycemia. Type 3c diabetes is defined as diabetes that is linked to various exocrine pancreatic disorders, primarily chronic pancreatitis, and pancreatic cancer. In particular, new-onset diabetes (NODM), which appears in the context of PC between 2-3 years, is primarily linked to pancreatic cancer [11]. There is a significant correlation between the length of DM and the advancement of PC. Studies evaluating time-related factors consistently show a negative correlation between the length of diabetes and the risk of pancreatic cancer, with the risk rising to a peak soon after the diagnosis and then gradually declining but staying elevated for many years. These findings point to a bidirectional causal relationship, suggesting that diabetes raises the risk of pancreatic cancer, which may be the outcome of malignancy that is not yet manifest [12]. Regarding the field of early PDAC detection, the identification of NOD as a clinical manifestation of occult PDAC carries significant implications. Being able to differentiate between long-term T2DM and PC-caused NOD offers the chance to diagnose PC at an early stage, when treatment may be curative [13]. Recent research also indicates that anti-diabetic drugs may have a direct impact on the major mediators of the relationship between T2DM and PC, thereby affecting the course, outcome, and development of cancer [14]. In the context of long-term diabetes, this story will highlight the latest research on the pathophysiological mechanisms underlying PC development, the biological processes in PC that predispose PC patients to developing diabetes, and the importance of these associations for PC early diagnosis and anti-diabetic treatment selection.
Discussion
Long-standing PC T2DM risk factor
Diabetes is widely recognized as a risk factor for PC. Both men and women with DM have been found to have an elevated risk of PC [15], and people with type 2 diabetes have been found to have an increased mortality rate from PC [16]. Numerous data point to a low reported risk ratio—which ranges from 1.4 to 2.1—for long-term diabetes-related PDAC development [17]. While long-term T2DM and NODM present varying degrees of increased risk, with NODM posing a greater risk, both present an increase in risk [18]. The major impact of long-term diabetes on PC susceptibility has been explained by a number of mechanisms, the most significant of which are chronic hyperglycemia, insulin resistance, inflammation, and alterations in the tumor microenvironment.
Hyperglycemia and Advanced Glycation End Products
Chronic hyperglycemia, a hallmark of diabetes, in numerous ways, promotes the growth of cancer cells. The incidence of PDAC actually rises by 14% for every 0.56 mmol/l increase in fasting blood glucose levels [19]. The proliferation of cancer cells and the expansion of tumors are greatly aided by hyperglycemia [20]. Furthermore, greater phosphorylation of p38 MAPK (mitogen-activated protein kinase) in pancreatic cancer cells is caused by elevated blood sugar levels. It follows that this increases the cell's capacity to divide, evade apoptosis, infiltrate neighboring tissues, and migrate. It is possible to counteract the proliferation, invasion, migration, and epithelial-mesenchymal transition (EMT) process that hyperglycemia induces in tumor cells by inhibiting p38 MAPK [21]. Hyperglycemia also promotes endothelial dysfunction, angiogenesis, and cell proliferation [22]. By activating EGFRs, the receptors for epidermal growth factors, and their corresponding factors [23]. Moreover, advanced glycation end products (AGEs) are produced in large quantities and accumulate in a hyperglycemic environment [24]. AGEs cause pancreatic cells' receptors (RAGEs) to become active, which in turn triggers a number of signaling pathways linked to inflammation, cancer progression, and cell survival [25]. For example, upon binding between AGEs and RAGEs, the MAPK and NF-κB signaling pathways are activated, leading to an increase in HIF-1α and NF-κB transcription. These molecular processes produce a hypoxic microenvironment and protect cells from oxidative stress-induced cell death [26]. Solid tumors frequently experience hypoxia, which is essential for a number of reasons, including promoting angiogenesis, strengthening the tumor's defenses against the immune system, stifling anti-tumor responses, and reducing the effectiveness of treatment [27]. Additionally, the interaction between AGE and RAGE raises inflammatory signaling molecules. More inflammation results from this, and myeloid-derived suppressor cells (MDSCs), which weaken the immune system, are drawn in. In cells, AGEs/RAGEs binding also triggers the PI3K-AKT and KRAS signaling pathways. As a result, the TP53 protein, which typically triggers apoptosis, is reduced. Moreover, it stimulates mTOR, which regulates autophagy and cell proliferation. These alterations work together to give cancer cells the ability to multiply, endure longer, and stimulate the growth of pancreatic cancer [26].
Insulin Resistance
Unlike hyperglycemia, insulin resistance shows a separate relationship with the chance of developing PC [28]. Moreover, elevated C-peptide levels show a relationship with a higher chance of the illness [29]. Studies have shown that PC tissue expresses more insulin receptors, and high levels of circulating insulin are often associated with a poor prognosis in PDAC cases [30, 31].Insulin mainly triggers two signaling pathways: the extracellular signal-regulated kinase (ERK) 1/2-MAPK pathway and the insulin receptor (IR)-insulin receptor substrates (IRS)-phosphatidylinositol 3-kinase (PI3K)-protein kinase B (Akt) signal pathway. Through a variety of mechanisms, including mediating metabolic reprogramming, interacting with insulin-like growth factor 1 (IGF-1), bolstering drug resistance, and encouraging the formation of a tumor microenvironment marked by inflammation, fibrosis, and neo-angiogenesis, these two pathways play critical roles in enhancing the development of PDAC [32].
Inflammation
One important mediator of the relationship between diabetes and pancreatic carcinogenesis is chronic inflammation. There is evidence of both systemic and localized inflammation. Chronic low-grade inflammation is also caused by obesity, which is closely linked to type 2 diabetes. Proinflammatory cytokines such as TNF-alpha, IL-1, IL-6, IL-18, and IFN-gamma, as well as inflammatory markers like CRP, are elevated in the bloodstream as a result of diabetes [33]. In addition to decreasing immune surveillance, inflammatory stimuli activate oncogenic transcription factors like NF-kB, STAT3, and NFAT, which raise the expression of genes governing cell survival, proliferation, and oncogenic transformation [26]. Furthermore, TNF-alpha, particularly at low concentrations, is crucial for the promotion of tumors because it generates reactive oxygen species (ROS) and reactive nitrogen species (RNS), both of which have the ability to damage DNA [34]. ROS triggers signaling cascades that propel the development and progression of cancer, such as PI3K/Akt/mTOR and MAPK/ERK [35].
Tumor Microenvironment Changes
Diabetes causes the pancreatic tumor microenvironment (TME) to significantly change, which accelerates the onset and spread of cancer. Initially, there is a correlation between diabetes and elevated adipose tissue accumulation and fatty infiltration in the pancreas. Diabetes patients had greater pancreatic fat content than controls, according to a meta-analysis [36]. It has been reported that intrapancreatic adipocyte accumulation increases the risk of developing pancreatic cancer through a variety of pathways, including the release of inflammatory cytokines and the activation of inflammatory pathways [37]. Diabetes also triggers the transformation of dormant pancreatic stellate cells (PSCs) into CAFs or cancer-associated fibroblasts. PSCs are the main progenitors of CAFs in the pancreas. In response to various stimuli, including obesity, hyperglycemia, and hyperinsulinemia, PSCs change from a quiescent state to activated myofibroblast-like cells (MFCs) [38]. Upon activation, they release cytokines that facilitate the growth, invasion, metastasis, EMT, and resistance to chemotherapy of tumor cells [24]. Above all, CAFs can prevent immune effector cells like CD8 + T cells from being recruited into tumor tissues by secreting different chemokines. Moreover, it has been shown that CAFs significantly contribute to immune suppression within the tumor by raising the quantity of immunosuppressive cells such as regulatory T (Treg) cells, myeloid-derived suppressor cells (MDSCs), and M2-type macrophages within the tumor's immune microenvironment (TIME) [39].
New onset DM an early manifestation of PC
Long-term type 2 diabetes is thought to increase the risk of developing prostate cancer (PC), but new-onset diabetes is thought to be an early sign of the disease that appears two to three years before the cancer is diagnosed [13]. Within three years of first meeting the glycemic criteria for new-onset diabetes, patients with NOD have an 8–10-fold increased risk of developing pancreatic cancer compared to the general population; the 3-year incidence of pancreatic cancer is 1% [40]. Patients who have been diagnosed with impaired glucose homeostasis within the last 12 months have the highest relative probability of an undiagnosed SPC [41]. Although the exact mechanisms underlying the association between PC and NOD are still unknown, the fact that more than 50% of patients experience a resolution of their diabetes following tumor removal through surgery suggests that the two conditions are causally related [42]. Since NOD can exist even before PC is radiologically detectable and has been demonstrated to improve following surgery, the theory that it is solely the result of PDAC's destruction of the endocrine pancreas is not a tenable explanation [43]. It is thought that tumor-secreted products trigger insulin resistance and insufficient beta cell response to stimuli, ultimately resulting in beta cell failure and new-onset diabetes [44].
Insulin resistance
Insulin action is significantly impaired by PC-associated NOD, according to early groundbreaking research [43]. The disordered glucose regulation typically appears gradually and appears as early as the PanIN period (a period that precedes pancreatic cancer) [32]. Actually, between 15 and 20 percent of PC patients have a normal β-cell function but are normoglycemic due to increased insulin resistance [45]. It is not insulin overexpression at pancreatic islet beta cells that causes this insulin resistance, but rather an increase in synthesized products that cause insulin resistance in the human body [32]. Muscle, adipose, and liver tissue from pancreatic cancer patients showed reduced insulin sensitivity; this is thought to be the consequence of exosome microRNAs secreted by pancreatic cancer cells [46]. Skeletal muscle from PC patients exhibits decreased phosphatidylinositol 3-kinase activity and impaired glucose transport when compared to controls; however, the early stages of insulin signaling (insulin receptor binding, tyrosine kinase activity, and insulin receptor substrate 1 content) are unaffected. According to these findings, exosomes inhibit the PI3K/Akt/FoxO1 signaling pathways and glucose transport, which in turn affects Glut4 translocation and glucose transport [47]. In addition to miRNAs, pancreatic cancer cells also produce galectin-3 and S100A9, which inhibit myotube cells' capacity to absorb glucose [19]. Additionally, a study found that the diabetic pancreatic cancer group had a lower fractional velocity of glycogen synthase, indicating that the insulin resistance linked to PC may have originated from a post-IR defect in the skeletal muscles [48]. Furthermore, because pancreatic cancer also targets adipose tissue, interactions between the two conditions may account for the development of insulin resistance and paraneoplastic weight loss in pancreatic cancer [49]. One potential mediator of this resistance is adrenomedullin (AM). This 52-amino acid peptide is expressed by pancreatic F-cells and has a variety of functions, including modulating tumor growth and progression through autocrine stimulation [50]. Adrenomedullin is produced by pancreatic cancer cells and fibroblasts in the pancreatic cancer microenvironment. This inhibits the adipocyte response to insulin signals by preventing beta cells from secreting insulin and enhances the insulin-inhibitory effect of PC cells both in vitro and in vivo. In patients with PC, particularly those with DM, a recent study discovered significant AM up-regulation in their plasma, at the gene and protein levels [51, 52]. Furthermore, Javeed et al. demonstrated that AM enters β cells by caveolin-mediated endocytosis after being transported from PC to β cells as exosomal cargo. This leads to insulin resistance via ADMR-AM interactions, reduced insulin secretion, overexpression of ER stress genes, and elevated reactive oxygen/nitrogen species. Remarkably, the inhibitory effect of PC-derived AM-positive exosomes on insulin secretion is eliminated upon blocking the interaction between AM and its receptors [53]. Additionally, PDA causes hepatic cells to become less sensitive to insulin, a sign of a lower level of liver gluconeogenesis [54]. Elevated blood glucose levels then stimulate insulin synthesis and secretion in pancreatic islet beta cells, resulting in endogenous hyperinsulinemia when a delayed and blunted insulin response occurs in PDA patients. Furthermore, diabetic PDA patients who take insulin regularly to control blood sugar were found to have worsened glucose control due to the high rate of insulin resistance. As a result of PDA-induced increased insulin resistance, more insulin will be used, which will raise exogenous hyperinsulinemia [32]. Moreover, it was recently demonstrated that PC patients with new-onset diabetes produce noticeably less glucose-dependent insulinotropic peptide (GIP), which is primarily secreted by enteroendocrine cells [52]. These patients have shown reduced levels of GIP, which may be the result of an increase in dipeptidyl peptidase IV levels brought on by the tumor and which can be reversed by tumor resection. As a result, low GIP levels may both contribute to and serve as a biomarker for PDAC-DM [55]. Additionally, it has been discovered that islet amyloid polypeptide (IAPP), which is typically secreted alongside insulin by β cells, is also a hormone that contributes to insulin resistance in patients with pancreatic cancer. According to a study, patients with diabetes who have pancreatic cancer have higher plasma levels of IAPP. This is explained by the PC cells stimulating β cells to secrete IAPP selectively and by changing β cells' reactivity to other secretagogues [56].
Beta cell dysfunction
Patients diagnosed with PDAC typically experience a reduction in both the quantity and size of β cells within their islets [57]. PDAC patients also showed impairments in the β cell response, which includes the response to an oral glucose load, hyperglycemic clamp, or glucagon stimulation [58]. The increased activation of the TGF beta pathway is one of the several mechanisms that have been proposed to explain the pathophysiological mechanisms behind the beta cell dysfunction observed in patients with pancreatic cancer. Parajuli et al. found that as PDAC progressed, the TGF-β signaling pathway was expressed more frequently, which in turn caused β-cell mass to be lost through apoptosis.According to this study [59], there is a correlation between increased TGF-β signaling activation and increased caspase 3 cleavage within islets, which ultimately results in apoptotic β-cell death during PDAC progression and causes new-onset diabetes. Furthermore, it was discovered that necrotic stellate cells, a major collagen-producing cell type in PDAC, cause β-cell apoptosis and dysfunction by causing islet fibrosis and reducing β-cell insulin production, potentially contributing to the development of diabetes mellitus [60]. Macrophage migration inhibitory factor is another potential contender (MIF). Pro-inflammatory cytokine MIF influences glucose homeostasis as well as inflammation. It has been noted that pancreatic cancer cells overproduce it, and studies have shown how it regulates Ca2+ channels to inhibit glucose-stimulated insulin release from both isolated islets and β-cells. Furthermore, elevated serum MIF levels have been observed in patients with pancreatic cancer who also have diabetes, whereas no such rise has been observed in patients with pancreatic cancer who do not have diabetes or in newly diagnosed Type 2 diabetics who do not have cancer [61]. Adrenomedullin, a strong inhibitor of insulin secretion and an inducer of endoplasmic reticulum (ER) stress increased β-cell dysfunction, and death has also been demonstrated to be upregulated in cancer cells [62]. Every one of the physio-pathological scenarios listed above causes β-cells to be selectively depleted, which may eventually lead to reduced insulin secretion and the ensuing hyperglycemia and diabetes. Indeed, this may worsen the diabetes phenotype by further causing an imbalance in the ratio between β-cells and other endocrine cells that are not sensitive to PDAC [63]. Relevantly, recent research has revealed that a significant increase in the systemic glucagon/insulin ratio is experienced by the majority of PDAC patients diagnosed with diabetes. This phenomenon is thought to facilitate gluconeogenesis and the release of glucose into the circulatory system [64]. Apart from glucagon, another study found that PDAC and/or PanINs have no effect on δ- and PP cells, two other types of islet cells [65].
New-onset diabetes vs. long-standing diabetes
There is consistent evidence linking diabetes to pancreatic cancer [66]. Depending on how long it has been since the cancer diagnosis, diabetes mellitus linked to pancreatic cancer is classified into two main groups: long-standing diabetes mellitus (diagnosed more than three years ago) and new-onset diabetes mellitus (diagnosed less than three years ago) [67]. PC and diabetes have a reciprocal relationship. New-onset diabetes is an early symptom and a pathogenetic feature of PC, whereas long-term diabetes is thought to be an etiologic/risk factor of SPC [68]. Making the correct distinction between these two forms of diabetes is crucial to establishing an early PC diagnosis [69]. First, the risk of developing PC varies between diabetes with a long history and diabetes with a recent onset. Since PDA is not well documented in DM2, and PDA risk is comparable to other common cancers in these patients, DM2 is thought to be a relatively low PDA risk group [70]. Though more than 50% of PDA-related diabetic cases developed diabetes in the 36 months prior to the PDA diagnosis, NOD is thought to be a high-risk signal of PDA. In addition, this cohort study showed that NOD had a 2.3-fold higher risk of pancreatic cancer than did long-term diabetes mellitus [71]. Similarly, the relative risk of PDAC in patients with diabetes mellitus increased with a shorter duration of the disease: 6.69 for less than a year, 1.86 for 1-4 years, 1.72 for 5-9 years, and 1.36 for more than 10 years, according to a meta-analysis of 88 studies [72]. Remarkably, a different study found that although the relative risk of pancreatic cancer decreased with age at the onset of type 2 diabetes, the absolute risk of the disease increased with age [73]. Second, as previously mentioned, there are differences in the pathophysiological mechanisms underlying the development of each type of diabetes and how it relates to pancreatic cancer. Third, following the onset of diabetes, subjects with T2DM vs. T3cDM experience different changes in body weight [74, 75]. Patients with T3cDM are substantially more likely than those with T2DM to have a decrease in body weight at the diagnosis of prediabetes or diabetes, most likely as a result of the tumor-induced loss of subcutaneous fat tissue [76]. Body weight loss typically occurs before other systemic and local symptoms in PC. In fact, a study found that the mean time between the onset of diabetes in pancreatic cancer and the onset of symptoms was two months and thirteen months, respectively, before the cancer was diagnosed. According to these data, weight loss may be linked to the development of diabetes and may occur several months before the onset of diabetes and cancer-specific symptoms in pancreatic cancer [77]. Consequently, a patient with newly diagnosed prediabetes or diabetes should have suspicions about a paraneoplastic origin if their body weight decreases by more than 2 kg [68]. Conversely, compared to T3cDM, T2DM typically starts with a higher BMI and increased body weight linked to insulin resistance and hyperinsulinemia [14]. It's also crucial to note that, even in cases where DM2 patients do experience weight loss, this weight loss is typically linked to a gradual improvement in glycaemic control, as opposed to what is seen in people with NO diabetes, where weight loss is associated with worsening glycemia [78]. There have been mixed findings from earlier research on the effect of diabetes mellitus (DM) on PDAC patients' prognoses. According to research by Mizuno et al., patients with new-onset diabetes mellitus alone had a median survival time for pancreatic cancer that was about 10 months longer than that of patients without diabetes [79]. This better prognosis could be linked to early detection and treatment of diabetes-related pancreatic cancer. This study found that patients with pancreatic cancer had a poor prognosis when they had long-term diabetes [10]. Conversely, other research revealed that patients with DM who received chemotherapy for PDAC had smaller tumor sizes, a higher risk of dying from the treatment, and a worse prognosis than those with long-standing or recently developed diabetes [80, 81]. Furthermore, a recent study discovered that significant weight loss and long-term diabetes are risk factors for distant metastases. This implies that cancer cells may spread to distant locations more easily if they are exposed to diabetes-related metabolic abnormalities over an extended period of time [82]. Lastly, the finding that more than 50% of patients with new-onset DM who had their PaC resected experienced an improvement in their DM, whereas patients with long-standing DM did not show a discernible improvement in their hyperglycemic status after surgery, suggesting a causal relationship between PC and NO diabetes [8].
Early detection of pancreatic cancer in patients with new onset diabetes mellitus
The majority of the time, pancreatic cancer is discovered in an advanced stage. The depressing 5-year survival rate of 3–15% is a result of such late detection, which also limits treatment options [83]. Only 15-20% of cases are currently diagnosed at a level where curative surgery is a possibility [84]. As a result, early detection of pancreatic cancer is critical. The majority of patients with pancreatic intraepithelial neoplasia (PanIN), stage I, are either asymptomatic or exhibit non-specific symptoms like pyrosis, abdominal pain, and weight loss in the early stages of the disease. When PC-related symptoms appear, the illness has already progressed [85]. Given the rarity of PC, it is strongly advised against doing a generalized screening of asymptomatic adults. Moreover, none of the primary imaging technologies for PC identification are affordable or easily applied to a larger scale of screening. In order to identify high-risk individuals for focused screenings, numerous attempts have been made recently [5,86]. In order to choose which population should be screened, Chari suggests using two "sieves": the first is for high-risk individuals (hereditary PC, NOD), and the second is for particular traits of these high-risk groups (known risk factors, suggestive symptoms, serum biomarkers, non-invasive imaging) [87].
Screening models
New-onset diabetes (NOD) is one of the most promising high-risk factors for pancreatic cancer because elderly patients with NOD have a higher risk of sporadic PC than the general population [40]. A score to evaluate the risk of PC in patients with NOD has been developed in recent years. To assess the risk of pancreatic cancer in patients with new-onset diabetes, Sharma et al. created a model known as enriching new-onset diabetes for pancreatic cancer (END-PAC). The blood glucose, weight, and age changes at glycemically defined new-onset diabetes are measured by the model. Patients with new-onset diabetes are more likely to develop PC if they have more significant changes in blood glucose, less significant weight changes, and higher ages [88]. The END-PAC model has been verified by multiple investigations. At the 3+ threshold, for instance, the sensitivity and specificity of the END-PAC model were 62.6% and 78.5%, respectively, according to a study conducted on 13,947 NOD patients in a healthcare setting [89]. The Health Improvement Network (THIN) model from the United Kingdom is another model that should be brought up. The population with a 5% 3-year predicted risk of ductal PC among individuals with NOD can be identified using this Boursi et al. model with 11% sensitivity and 99.7% specificity. To identify the population with a 5% 3-year predicted risk of ductal PC among people with NOD, this model takes into account factors such as age, changes in body mass index (BMI), smoking, diabetes medications, proton pump inhibitors, changes in hemoglobin A1c, total cholesterol, creatinine, and alkaline phosphatase [90].
Biomarkers associated with PC and new-onset diabetes
No single biomarker currently seems appropriate for clinical use for the early detection of pancreatic ductal cancer, despite multiple attempts. In patients with NOD, the amount of carbohydrate antigen 19-9 (CA19-9) secreted by cancer cells may be a good predictor of PC [91]. Higher CA19-9) levels above the upper normal limit were associated with a 5.5 times greater risk of developing PC within 2 years of diagnosis in asymptomatic NOD patients, according to a study by Choe et al [92]. Another study suggested that patients with diabetes with or without PC could be distinguished using a cut-off serum CA19-9 level of 75 U/mL. The CA19-9 for PC had a sensitivity of 69.5% and a specificity of 98.2% at this cut-off [93]. It is crucial to note, though, that other benign diseases affecting the pancreas, biliary tract, and other organs can also cause an increase in CA19-9 levels. However, some data suggests that its levels are much higher in malignant than in benign diseases [94].Osteoprotegerin is another potential biomarker (OPG). OPG is a member of the TNF receptor superfamily (TNFRSF) and a soluble decoy receptor for TNF-related apoptosis-inducing ligand (TRAIL) [95]. In a meta-analysis of gene expression microarray datasets, Shi W et al. discovered that patients with PC-related DM had elevated serum OPG levels. Serum OPG showed a sensitivity of 68%, specificity of 73.9%, and area under the curve (AUC) of 73.7% in identifying PC in patients with NOD [96]. Furthermore, the systemic inflammatory response to the tumor raises C-reactive protein (CRP), and tumor necrosis factor-α (TNF-α) is an upregulating factor of CRP in PCa patients. Higher levels of sTNF-R2 were found to tend to increase PC risk, and this association was stronger in people with diabetes, according to Grote VA et al. [97]. Promising biomarkers for identifying PC in NOD patients include other pancreatic metabolites like docosahexaenoic acid, LysoPC and histidyl-lysine46, and changes in microRNA blood levels like miR-20b-5p, miR-29a, and miR-18a-5p [98].
Biomarkers that help differentiate between NOD and long-standing T2DM in PC patients
In clinical practice, it can be difficult to distinguish PC-induced NOD from the more prevalent T2DM based on clinical and biochemical factors. As a result, a number of biomarkers are proposed to help PC patients distinguish between the two types. One of these is islet amyloid polypeptide (IAPP), a hormone released by pancreatic beta cells that has been shown to have diabetogenic effects both in vivo and in vitro and is thought to be the root cause of insulin resistance in diabetes mellitus associated with PCa. While it was found to be normal or low in patients with type 2 DM, it was found to be elevated in patients with PCa-associated DM [99]. Other biomarkers include the protein S100A9, which is connected to inflammation through toll-like receptor-4, and Galectin-3, a β-galactoside-binding lectin involved in PC cell proliferation, migration, and invasion [42]. According to Liao et al., PDAC-related DM had higher levels of galectin-3 and S100A9 than T2DM. Additionally, they discovered that PDAC-related DM and T2DM can be distinguished from one another by serum levels of the proteins galectin-3 and S100A9 [100]. Thrombospondin-1 (TSP-1), a multimeric protein with anti-angiogenic characteristics, is another intriguing biomarker. TSP-1 levels were observed to be lower in PDAC patients than in non-diabetic individuals, especially in those with diabetes. Significantly, whereas lower TSP-1 levels were not observed in long-term T2DM, they were in PDAC-associated diabetes [101]. Furthermore, it was discovered that the enzyme vascular non-inflammatory molecule-1, or vanin 1, was linked to paraneoplastic islet cell dysfunction and may be a useful biomarker for identifying PC in patients with diabetes [102]. According to Huang et al., PDAC was found to have significantly higher Vanin-1 gene levels, and PDAC-associated diabetes could be distinguished from type 2 diabetes by measuring serum levels of Vanin-1 and matrix metalloproteinase 9 (MMP9) using quantitative real-time polymerase chain reaction [103].Additionally, a microRNA panel consisting of miR-483-5p, miR-19a, miR-29a, miR-20a, miR-24, and miR-25 was reported by Dai et al. to have an AUC of 0.887 in differentiating PC-related diabetes from T2DM [104].
Shared genetic predisposition between PC and DM
Significant advancements in our knowledge of complex diseases, such as cancer and type 2 diabetes, have been made since the introduction of genome-wide association studies (GWAS) in 2005 [105]. According to GWAS, patients with type 2 diabetes may be at increased risk of developing pancreatic cancer if they have mutations in genes related to embryonic development and the control of pancreas-specific genes like Nr5a3, HNF1A (hepatocyte nuclear factor 1), and PDX1 (pancreatic and duodenal homeobox 1) [106,107]. For instance, normal pancreatic development, appropriate β-cell function, and insulin secretion all depend on the HFN1A and HNF1B genes [18]. It has been observed that mutations in these genes, which are linked to type 2 diabetes, also contribute to the development of pancreatic cancer [22]. Two (in FTO and MTNR1B) and one (in BCL11A) of the 37 type 2 diabetes risk alleles were found to have nominally positive associations with pancreatic cancer risk, according to a different study [108]. Furthermore, et al found that test cohorts for PC and T2DM shared 44 genes primarily enriched in the regulation of endodermal cell fate specification, suggesting a potential shared mechanism between the two conditions. A particularly noteworthy finding in validation cohorts was the identification of a hub-shared gene, S100A6, which is important for the progression of both PC and T2DM and may be used as a predictive biomarker to identify covert PC in T2DM patients [109]. S100A6 is a member of the S100 family and a calcium-binding protein with an EF-hand. According to earlier research, S100A16 primarily contributes to lipid metabolism and stimulates the release of insulin in response to calcium, a condition linked to hyperglycemia [110,111]. Furthermore, it has been discovered to have a role in a number of cancers, including pancreatic cancer [112]. S100A6 was found to be significantly more expressed in PC tissues than in non-tumor tissues in the study. Because S100A6 is involved in stimulating PC cell proliferation and migration, PC patients with high expression levels of the protein had a lower overall survival (OS) than those with low expression levels [109].Moreover, the major C-allele of the diabetes-related SNP GCKR rs780094 may raise the risk of pancreatic cancer. GCKR controls glucokinase (GCK) activity in pancreatic islet cells and liver cells, respectively.High levels of insulin and fasting glucose may be caused by a high degree of glucokinase inhibition in CC carriers, which may have a mitogenic effect on the exocrine pancreatic cells. Therefore, through increased insulin levels and/or diabetes, GCKR may influence the development of pancreatic cancer [113].The promoter genes of CDH1 AND CDKN2A are other variants that are important to note. According to a study, PDC frequently experienced both CDH1 and CDKN2A promoter gene methylation, and the frequency of positive cases was higher in long-DM patients than in non-DM and short-DM patients. Promoter methylation of CDH1 was more prevalent in pancreatic cancer with long-term diabetes mellitus and was positively connected with decreased expression of E-cadherin. Therefore, it is highly likely that diabetes itself will cause promoter methylation [114]. Last but not least, reverse causation should also be taken into account because type 2 diabetes can both cause and be preceded by pancreatic cancer [115]. Only the correlation between LINC-PINT rs6971499 and T2DM risk was validated among the 14 known PC susceptibility variants, albeit at a weak significance level that vanished upon multiple comparison adjustments [116].
Table 1: Compares new-onset diabetes to late-onset diabetes and its association with pancreatic cancer in terms of signs and symptoms, pathology, and treatment.
New onset of Diabetes | Late onset of diabetes | |
Symptoms and signs | Symptoms may include unexplained weight loss, abdominal pain, jaundice (yellowing of the skin and eyes), and changes in stool color. | It may not exhibit specific symptoms directly related to pancreatic cancer if it has been present for several years. |
Pathology | Pancreatic cancer has the potential to cause new-onset diabetes by impairing the pancreas' ability to produce insulin. Pancreatic tumors can obstruct the pancreatic duct, resulting in impaired insulin secretion. | Diabetes mellitus which has been present for a long time is a risk factor for the development of pancreatic cancer. The pathology in these cases typically involves a longer duration of diabetes before the cancer diagnosis. |
The underlying pathology frequently involves the formation of tumors in the pancreatic head, which can disrupt normal pancreatic function and result in elevated blood sugar levels. | Pathological features may include more widespread tumor involvement within the pancreas or metastasis to nearby tissues or lymph nodes, emphasizing the importance of regular monitoring for people with diabetes who have had it for a long time. | |
Treatment | Cases of newly diagnosed diabetes may provide a "window of opportunity" for early detection and intervention, potentially expanding treatment options and improving outcomes. | Treatment options for people with long-standing diabetes may be limited due to the cancer's advanced stage. |
Effect of Anti-Diabetic Medication on Pancreatic Cancer
Insulin therapy
Insulin is safe for the majority of diabetic patients because pancreatic cancer is not common in the general population. On the other hand, individuals with high risk of PDA, such as those with NODM, and PDA patients, should use insulin with greater caution [32]. Numerous studies have demonstrated that insulin therapy may raise the incidence of PC [117, 118]. In fact, a retrospective cohort analysis of the UK GP database (The Health Information Network; THIN) [119] supported the yearly incremental risk of cancer in individuals receiving insulin treatment. Bosetti et al. examined the risk association between PC and insulin therapy by examining 15 case-control studies, comprising 13,987 controls and 8,305 patient cases. Research findings revealed that while long-term insulin use (≥15 years) was not linked to an increased risk of PC, short-term insulin use (<5>
Metformin therapy
Worldwide, metformin is frequently prescribed as the first-line treatment for type 2 diabetes. Traditionally, metformin lowers hepatic gluconeogenesis in order to act as an antidiabetic [122]. Numerous epidemiologic studies conducted in the last few years have suggested that metformin may be associated with a lower risk of PDAC [59]. Metformin-using diabetics have a lower risk of PC than non-users, according to a meta-analysis of 29 papers involving over 2 million participants [123]. In a similar vein, a different study found that taking metformin reduced the risk of PC by 37% when compared to other diabetes treatments [124]. Metformin's anti-cancer mechanism is not entirely understood, but it is primarily thought to be through activating liver kinase B1 (LKB1) and adenosine monophosphate protein-activated kinase (AMPK), which can reduce hepatic tissue insulin resistance and prolong survival in cancerous metformin [125]. In pancreatic cancer, metformin induces LKB1 activation to break the crosstalk between IR and GPCR, which reduces cancer growth and fibrosis. Additionally, research showed that metformin could trigger ferroptosis and apoptosis in pancreatic cancer by activating the AMPK signal pathway, which is inhibited by high glucose status [1]. Furthermore, metformin may stimulate the STING/IRF3/IFN-β pathway by blocking AKT signaling in PDAC cells, which would facilitate the infiltration of immune cells into the tumor microenvironment (TME), according to research by Ren et al. [126]. In addition, through the MTOR signaling pathway, metformin can suppress PDAC by promoting the expression of pro-apoptosis proteins caspase-3 and Bax and reducing the expression of anti-apoptosis protein Bcl-2 [127]. Remarkably, metformin was only found to prolong survival in diabetic PDA patients; no effect was observed in the overall PDA population. This suggests that metformin primarily reduces insulin resistance rather than having an anticancer effect [128].
Incretin-based therapy
Incretin-based therapies, which include dipeptidyl peptidase-4 inhibitors (DPP-4 inhibitors) and glucagon-like peptide-1 receptor agonists (GLP-1 RAs), are another class of frequently used anti-diabetic medications. Data regarding the risk for PC were inconsistent, despite the initial preclinical and adverse database review studies raising concerns about acute pancreatitis and PC associated with their chronic use [129]. It was previously hypothesized that incretin-based treatments would cause the GLP-1 receptor to become overstimulated, which would raise the risk of PC [130]. However, Nreu et al. found in 2020 [131, 132] that there was no relationship between GLP1-RA use and PC. It is noteworthy to note that certain research has linked GLP-1RAs to an antitumor effect on pancreatic cancers in humans [133]. It was proposed that GLP-RAs may help reverse insulin resistance caused by pancreatic cancer cells, resulting in a lower blood insulin level. This is because they lower blood glucose by encouraging energy storage in adipose tissue and inhibiting glucagon secretion [134]. However, there may not be enough information to make such a determination regarding the connection between PC and this incretin-based drug. A serine protease called dipeptidyl peptidase-4 (DPP4) facilitates the breakdown of GLP-1.52 Consequently, DPP4-inhibitor (DDP4i) can be employed as a type of hypoglycemic agent by inhibiting the degradation of GLP-1 [135, 136, 137].
Sodium-Glucose Cotransporter 2 Inhibitors
By reducing the reabsorption of glucose and raising the excretion of urine glucose, sodium-dependent glucose transporter-2 (SGLT-2) inhibitors, another type of oral antidiabetic medication, contribute to the maintenance of glycaemic control [138]. Apart from their significant cardiovascular advantages, they exhibit the capacity to impede the advancement of Parkinson's disease. In cells of pancreatic cancer, SGLT-2 was found to be highly expressed. In preclinical pancreatic cancer models, SGLT-2 inhibitor treatment was found to reduce cancer proliferation and lower PDA risk in SGLT-2 inhibitor users. According to these results, SGLT2 inhibitors might be helpful in the treatment of cancer [139].
Conclusion
Diabetes has a broad spectrum of morbidity, while PC is a highly aggressive and deadly malignancy. The complex relationships between PC and diabetes are demonstrated by epidemiological studies. The mechanisms underlying the correlation between PC and DM include chronic inflammation, hyperglycemia, hyperinsulinemia, and insulin resistance. Metformin and insulin are the two main medical treatments for type 2 diabetes. Studies have shown that metformin lowers the risk of PC, but insulin therapy raises the risk of PC; for this reason, metformin has the potential to act as an anticancer drug and can be used to stop the growth of malignant lesions. Surgical resection is the only potentially curative treatment for PC; however, evidence suggests that T2DM is a significant comorbidity that predicts worse outcomes in patients undergoing PC resection and is associated with decreased survival.Consequently, research has been concentrating on novel screening models and biomarkers to identify PC risk in individuals with recently developed diabetes and encourage early detection of PC while it is still treatable.
Declarations
Funding: Not applicable
Conflicts of interest: The authors declare no conflict of interest.
Ethics approval: Not applicable
Consent to participate: Not applicable
Consent for publication: Not applicable
Availability of data and material (data transparency): Not applicable
Code availability: Not applicable
Acknowledgment: The authors thank Dr. Hiba Hamdar (hiba.hamdar@live.com), who supervised us reviewed our article, and gave us her valuable comments.
Authors' contributions: Each author has contributed in the same manner to this manuscript.
References
- Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. (2018). Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries CA Cancer. J Clin, 68(6):394-424.
Publisher | Google Scholor - Grant TJ, Hua K, Singh A. (2016). Molecular Pathogenesis of Pancreatic Cancer. Prog Mol Biol Transl Sci, 144:241-275.
Publisher | Google Scholor - GBD 2017 Pancreatic Cancer Collaborators. (2019). The global, regional, and national burden of pancreatic cancer and its attributable risk factors in 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol, 4(12):934-947.
Publisher | Google Scholor - Gallo M, Adinolfi V, Morviducci L, et al. (2021). Early prediction of pancreatic cancer from new-onset diabetes: an Associazione Italiana Oncologia Medica (AIOM)/Associazione Medici Diabetologi (AMD)/Società Italiana Endocrinologia (SIE)/Società Italiana Farmacologia (SIF) multidisciplinary consensus position paper. ESMO Open, 6(3):100155.
Publisher | Google Scholor - Li Y, Bian X, Wei S, He M, Yang Y. (2019). The relationship between pancreatic cancer and type 2 diabetes: cause and consequence. Cancer Manag Res, 11:8257-8268.
Publisher | Google Scholor - Momayez Sanat Z, Masoudi S, Mansouri M, Ghamarzad Shishavan N, Jameshorani M, Pourshams A. (2021). Diabetes Mellitus, Obesity, and Risk of Pancreatic Ductal Adenocarcinoma: a Large Case-Control Study from Iran. Middle East J Dig Dis, 13(1):15-20.
Publisher | Google Scholor - Grote VA, Becker S, Kaaks R. (2010). Diabetes mellitus type 2 - an independent risk factor for cancer?. Exp Clin Endocrinol Diabetes, 118(1):4-8.
Publisher | Google Scholor - Pannala R, Basu A, Petersen GM, Chari ST. (2009). New-onset diabetes: a potential clue to the early diagnosis of pancreatic cancer. Lancet Oncol, 10(1):88-95.
Publisher | Google Scholor - Păunică I, Giurgiu M, Dumitriu AS, et al. (2023). The Bidirectional Relationship between Periodontal Disease and Diabetes Mellitus-A Review. Diagnostics (Basel), 13(4):681.
Publisher | Google Scholor - American Diabetes Association (2014). Diagnosis and classification of diabetes mellitus. Diabetes Care,37 (S1):81-90.
Publisher | Google Scholor - Pannala R, Leirness JB, Bamlet WR, Basu A, Petersen GM, Chari ST. (2008). Prevalence and clinical profile of pancreatic cancer-associated diabetes mellitus. Gastroenterology, 134(4):981-987.
Publisher | Google Scholor - Pizzato M, Turati F, Rosato V, La Vecchia C. (2019). Exploring the link between diabetes and pancreatic cancer. Expert Rev Anticancer Ther, 19(8):681-687.
Publisher | Google Scholor - Sharma A, Chari ST. (2018). Pancreatic Cancer and Diabetes Mellitus. Curr Treat Options Gastroenterol.16(4):466-478.
Publisher | Google Scholor - Andersen DK, Korc M, Petersen GM, et al. (2017). Diabetes, Pancreatogenic Diabetes, and Pancreatic Cancer. Diabetes, 66(5):1103-1110.
Publisher | Google Scholor - Ben Q, Xu M, Ning X, et al. (2011). Diabetes mellitus and risk of pancreatic cancer: A meta-analysis of cohort studies. Eur J Cancer, 47(13):1928-1937.
Publisher | Google Scholor - Ling S, Brown K, Miksza JK, Howells L, Morrison A, Issa E, Yates T, Khunti K, Davies MJ, Zaccardi F. (2020). Association of Type 2 Diabetes with Cancer: A Meta-analysis With Bias Analysis for Unmeasured Confounding in 151 Cohorts Comprising 32 million People. Diabetes Care, 43:2313-2322.
Publisher | Google Scholor - Batabyal P, Vander Hoorn S, Christophi C, Nikfarjam M. (2014). Association of diabetes mellitus and pancreatic adenocarcinoma: a meta-analysis of 88 studies. Ann Surg Oncol, 21: 2453-2462.
Publisher | Google Scholor - Li D. (2012). Diabetes and pancreatic cancer. Mol Carcinog, 51(1):64-74.
Publisher | Google Scholor - Liao WC, Tu YK, Wu MS, Lin JT, Wang HP, Chien KL. (2015). Blood glucose concentration and risk of pancreatic cancer: systematic review and dose-response meta-analysis. BMJ, 350:7371.
Publisher | Google Scholor - Quoc Lam B, Shrivastava SK, Shrivastava A, Shankar S, Srivastava RK. (2020). The Impact of obesity and diabetes mellitus on pancreatic cancer: Molecular mechanisms and clinical perspectives. J Cell Mol Med, 24(14):7706-7716.
Publisher | Google Scholor - Wang, Lishan, et al. (2016).
Publisher | Google Scholor - Bhattacharyya S, Sul K, Krukovets I, Nestor C, Li J, Adognravi OS. (2012). Novel tissue-specific mechanism of regulation of angiogenesis and cancer growth in response to hyperglycemia. J Am Heart Assoc, 1(6):005967.
Publisher | Google Scholor - Han L, Ma Q, Li J, et al. (2011). High glucose promotes pancreatic cancer cell proliferation via the induction of EGF expression and transactivation of EGFR. PLoS One, 6(11):27074.
Publisher | Google Scholor - Singh VP, Bali A, Singh N, Jaggi AS. (2014). Advanced glycation end products and diabetic complications. Korean J Physiol Pharmacol, 18(1):1-14.
Publisher | Google Scholor - El-Far AH, Sroga G, Jaouni SKA, Mousa SA. (2020). Role and Mechanisms of RAGE-Ligand Complexes and RAGE-Inhibitors in Cancer Progression. Int J Mol Sci, 21(10):3613.
Publisher | Google Scholor - Ruze R, Song J, Yin X, et al. (2023). Mechanisms of obesity- and diabetes mellitus-related pancreatic carcinogenesis: a comprehensive and systematic review. Signal Transduct Target Ther, 8(1):139.
Publisher | Google Scholor - Emami Nejad A, Najafgholian S, Rostami A, et al. (2021). The role of hypoxia in the tumor microenvironment and development of cancer stem cell: a novel approach to developing treatment. Cancer Cell Int, 21(1):62.
Publisher | Google Scholor - Wolpin, Brian M., et al. (2013).
Publisher | Google Scholor - Michaud DS, Wolpin B, Giovannucci E, et al. (2007). Prediagnostic plasma C-peptide and pancreatic cancer risk in men and women. Cancer Epidemiol Biomarkers Prev, 16(10):2101-2109.
Publisher | Google Scholor - Kim NH, Chang Y, Lee SR, Ryu S, Kim HJ. (2020). Glycemic Status, Insulin Resistance, and Risk of Pancreatic Cancer Mortality in Individuals with and Without Diabetes. Am J Gastroenterol, 115(11):1840-1848.
Publisher | Google Scholor - Heckl SM, Kercher L, Abdullazade S, et al. (2021). Insulin Receptor in Pancreatic Cancer-Crown Witness in Cross Examination. Cancers (Basel), 13(19):4988.
Publisher | Google Scholor - Deng J, Guo Y, Du J, et al. (2022). The Intricate Crosstalk Between Insulin and Pancreatic Ductal Adenocarcinoma: A Review from Clinical to Molecular. Front Cell Dev Biol, 10:844028.
Publisher | Google Scholor - Goldberg RB. (2009). Cytokine and cytokine-like inflammation markers, endothelial dysfunction, and imbalanced coagulation in development of diabetes and its complications. J Clin Endocrinol Metab, 94(9):3171-3182.
Publisher | Google Scholor - Padoan A, Plebani M, Basso D. (2019). Inflammation and Pancreatic Cancer: Focus on Metabolism, Cytokines, and Immunity. Int J Mol Sci, 20(3):676.
Publisher | Google Scholor - Chio IIC, Tuveson DA. (2017). ROS in Cancer: The Burning Question. Trends Mol Med, 23(5):411-429.
Publisher | Google Scholor - Garcia TS, Rech TH, Leitão CB. (2017). Pancreatic size and fat content in diabetes: A systematic review and meta-analysis of imaging studies. PLoS One, 12(7):0180911.
Publisher | Google Scholor - Lilly, A. C., Astsaturov, I., & Golemis, E. A. (2023). Intrapancreatic fat, pancreatitis, and pancreatic cancer. Cellular and molecular life sciences: CMLS, 80(8):206.
Publisher | Google Scholor - Uchida C, Mizukami H, Hara Y, et al. (2021). Diabetes in Humans Activates Pancreatic Stellate Cells via RAGE in Pancreatic Ductal Adenocarcinoma. Int J Mol Sci, 22(21):11716.
Publisher | Google Scholor - Mao X, Xu J, Wang W, et al. (2021). Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: new findings and future perspectives. Mol Cancer, 20(1):131.
Publisher | Google Scholor - Magruder JT, Elahi D, Andersen DK. (2011). Diabetes and pancreatic cancer: chicken or egg?. Pancreas, 40(3):339-351.
Publisher | Google Scholor - Ben Q, Xu M, Ning X, et al. (2011). Diabetes mellitus and risk of pancreatic cancer: A meta-analysis of cohort studies. Eur J Cancer, 47(13):1928-1937.
Publisher | Google Scholor - Roy A, Sahoo J, Kamalanathan S, Naik D, Mohan P, Kalayarasan R. (2021). Diabetes and pancreatic cancer: Exploring the two-way traffic. World J Gastroenterol, 27(30):4939-4962.
Publisher | Google Scholor - Permert J, Ihse I, Jorfeldt L, von Schenck H, Arnquist HJ, Larsson J. (1993). Improved glucose metabolism after subtotal pancreatectomy for pancreatic cancer. Br J Surg, 80:1047-1050.
Publisher | Google Scholor - Sah RP, Nagpal SJ, Mukhopadhyay D, Chari ST. (2013). New insights into pancreatic cancer-induced paraneoplastic diabetes. Nat Rev Gastroenterol Hepatol, 10(7):423-433.
Publisher | Google Scholor - Wang F, Larsson J, Adrian TE, Gasslander T, Permert J. (1998). In vitro influences between pancreatic adenocarcinoma cells and pancreatic islets. J Surg Res. 79(1):13-19.
Publisher | Google Scholor - Wang L, Zhang B, Zheng W, et al. (2017). Exosomes derived from pancreatic cancer cells induce insulin resistance in C2C12 myotube cells through the PI3K/Akt/FoxO1 pathway. Sci Rep, 7(1):5384.
Publisher | Google Scholor - Isaksson B, Strömmer L, Friess H, et al. (2003). Impaired insulin action on phosphatidylinositol 3-kinase activity and glucose transport in skeletal muscle of pancreatic cancer patients. Pancreas, 26(2):173-177.
Publisher | Google Scholor - Liu J, Knezetic JA, Strömmer L, Permert J, Larsson J, Adrian TE. (2000). The intracellular mechanism of insulin resistance in pancreatic cancer patients. J Clin Endocrinol Metab, 85(3):1232-1238.
Publisher | Google Scholor - Kahn SE, Hull RL, Utzschneider KM. (2006). Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature, 444(7121):840-846.
Publisher | Google Scholor - Ramachandran V, Arumugam T, Hwang RF, Greenson JK, Simeone DM, Logsdon CD. (2007). Adrenomedullin is expressed in pancreatic cancer and stimulates cell proliferation and invasion in an autocrine manner via the adrenomedullin receptor, ADMR. Cancer Res, 67(6):2666-2675.
Publisher | Google Scholor - Aggarwal G, Ramachandran V, Javeed N, et al. (2012). Adrenomedullin is up-regulated in patients with pancreatic cancer and causes insulin resistance in β cells and mice. Gastroenterology, 143(6):1510-1517.
Publisher | Google Scholor - Zhang Y, Huang S, Li P, et al. (2018). Pancreatic cancer-derived exosomes suppress the production of GIP and GLP-1 from STC-1 cells in vitro by down-regulating the PCSK1/3. Cancer Lett, 431:190-200.
Publisher | Google Scholor - Javeed N, Sagar G, Dutta SK, et al. (2015). Pancreatic Cancer-Derived Exosomes Cause Paraneoplastic β-cell Dysfunction. Clin Cancer Res, 21(7):1722-1733.
Publisher | Google Scholor - Basso D, Valerio A, Brigato L, et al. (1997). An unidentified pancreatic cancer cell product alters some intracellular pathways of glucose metabolism in isolated rat hepatocytes. Pancreas, 15(2):132-138.
Publisher | Google Scholor - Škrha J, Bušek P, Uhrová J, et al. (2017). Lower plasma levels of glucose-dependent insulinotropic peptide (GIP) and pancreatic polypeptide (PP) in patients with ductal adenocarcinoma of the pancreas and their relation to the presence of impaired glucoregulation and weight loss. Pancreatology, 17(1):89-94.
Publisher | Google Scholor - Ding X, Flatt PR, Permert J, Adrian TE. (1998). Pancreatic cancer cells selectively stimulate islet beta cells to secrete amylin. Gastroenterology, 114(1):130-138.
Publisher | Google Scholor - Nagpal SJS, Kandlakunta H, Sharma A, Sannapaneni S, Velamala P, Majumder S, et al. (2018). Endocrinopathy in Pancreatic Cancer Is Characterized by Reduced Islet Size and Density with Preserved Endocrine Composition as Compared to Type 2 Diabetes: Presidential Poster Award: 45. American Journal of Gastroenterology, 113:26-S28
Publisher | Google Scholor - Cersosimo E, Pisters PW, Pesola G, McDermott K, Bajorunas D, Brennan MF. (1991). Insulin secretion and action in patients with pancreatic cancer. Cancer, 67(2):486-493.
Publisher | Google Scholor - Fu YB, Yang G, Qiu JD, Zhang TP. (2022). Diabetes Mellitus and Early Detection of Pancreatic Cancer. Cancer Screening and Prevention, 1(1):64-71.
Publisher | Google Scholor - Mekapogu, Alpha R., Srinivasa P. Pothula, Romano C. Pirola, Jeremy S. Wilson, & Minoti V. Apte. (2019).
Publisher | Google Scholor - Tan L, Ye X, Zhou Y, Yu M, Fu Z, Chen R, Zhuang B, Zeng B, Ye H, Gao W, Lin Q, Li Z, Zhou Q, Chen R. (2014). Macrophage migration inhibitory factor is overexpressed in pancreatic cancer tissues and impairs insulin secretion functionof β-cell. Journal of Translational Medicine.
Publisher | Google Scholor - Martinez A, Weaver C, Lopez J, Bhathena SJ, Elsasser TH, Miller MJ, Moody TW, Unsworth EJ, Cuttitta F. (1996). Regulation of insulin secretion and blood glucose metabolism by adrenomedullin. Endocrinology, 137:2626-2632.
Publisher | Google Scholor - Parajuli P, Nguyen TL, Prunier C, Razzaque MS, Xu K, Atfi A. (2020). Pancreatic cancer triggers diabetes through TGF-β-mediated selective depletion of islet β-cells. Life Sci Alliance. 3(6):201900573.
Publisher | Google Scholor - Kolb A, Rieder S, Born D, et al. (2009). Glucagon/insulin ratio as a potential biomarker for pancreatic cancer in patients with new-onset diabetes mellitus. Cancer Biol Ther, 8(16):1527-1533.
Publisher | Google Scholor - Hauge-Evans AC, King AJ, Carmignac D, et al. (2009). Somatostatin secreted by islet delta-cells fulfills multiple roles as a paracrine regulator of islet function. Diabetes, 58(2):403-411.
Publisher | Google Scholor - Tan JX, You Y, Guo F, Xu JH, Dai HS, Bie P. (2017). Association of elevated risk of pancreatic cancer in diabetic patients: A systematic review and meta-analysis. Oncology Letters, 13:1247-1255.
Publisher | Google Scholor - Jeon CY, Li D, Cleary S, et al. (2018). The Association of Recently Diagnosed Diabetes and Long-term Diabetes with Survival in Pancreatic Cancer Patients: A Pooled Analysis. Pancreas, 47(3):314-320.
Publisher | Google Scholor - Škrha, Jan, et al. (2018). ‘Sporadic Pancreatic Cancer: Glucose Homeostasis and Pancreatogenic Type 3 Diabetes’. Advances in Pancreatic Cancer.
Publisher | Google Scholor - Muniraj T, Chari ST. (2012). Diabetes and pancreatic cancer. Minerva Gastroenterol Dietol, 58(4):331-345.
Publisher | Google Scholor - Molina-Montes E, Coscia C, Gómez-Rubio P On behalf of the PanGenEU Study Investigators, et al. (2021). Deciphering the complex interplay between pancreatic cancer, diabetes mellitus subtypes and obesity/BMI through causal inference and mediation analyses Gut. 70:319-329.
Publisher | Google Scholor - Setiawan VW, Stram DO, Porcel J, et al. (2019). Pancreatic Cancer Following Incident Diabetes in African Americans and Latinos: The Multiethnic Cohort. J Natl Cancer Inst, 111(1):27-33.
Publisher | Google Scholor - Batabyal P, Vander Hoorn S, Christophi C, Nikfarjam M. (2014). Association of diabetes mellitus and pancreatic adenocarcinoma: a meta-analysis of 88 studies. Ann Surg Oncol, 21(7):2453-2462.
Publisher | Google Scholor - Shen B, Li Y, Sheng CS, et al. (2022). Association between age at diabetes onset or diabetes duration and subsequent risk of pancreatic cancer: Results from a longitudinal cohort and mendelian randomization study. Lancet Reg Health West Pac, 30:100596.
Publisher | Google Scholor - Wang F, Herrington M, Larsson J, Permert J. (2003). The relationship between diabetes and pancreatic cancer. Mol Cancer, 2:4.
Publisher | Google Scholor - Yuan C, Babic A, Khalaf N, Nowak JA, Brais LK, Rubinson DA, Ng K, Aguirre AJ, Pandharipande PV, Fuchs CS, Giovannucci EL, Stampfer MJ, Rosenthal MH, Sander C, Kraft P, Wolpin BM. (2020). Diabetes, Weight Change, and Pancreatic Cancer Risk. JAMA Oncol, 6(10):202948.
Publisher | Google Scholor - Sagar G, Sah RP, Javeed N, Dutta SK, Smyrk TC, Lau JS, Giorgadze N, Tchkonia T, Kirkland J, Chari ST, Mukhopadhyay D. (2016). Pathogenesis of pancreatic cancer exosome-induced lipolysis in adipose tissue. Gut, 65:1165-1174.
Publisher | Google Scholor - Liu Z, Hayashi H, Matsumura K, et al. (2023). Biological and Clinical Impacts of Glucose Metabolism in Pancreatic Ductal Adenocarcinoma. Cancers (Basel), 15(2):498.
Publisher | Google Scholor - Pannala R, Leibson CL, Rabe KG, et al. (2009). Temporal association of changes in fasting blood glucose and body mass index with diagnosis of pancreatic cancer. Am J Gastroenterol, 104(9):2318-2325.
Publisher | Google Scholor - Mizuno S, Nakai Y, Isayama H, et al. (2013). Diabetes is a useful diagnostic clue to improve the prognosis of pancreatic cancer. Pancreatology, 13(3):285-289.
Publisher | Google Scholor - Shen H, Zhan M, Wang W, Yang D, Wang J. (2016). Impact of diabetes mellitus on the survival of pancreatic cancer: a meta-analysis. Onco Targets Ther. 9:1679-1688.
Publisher | Google Scholor - Ma J, Wang J, Ge L, Long B, Zhang J. (2019). The impact of diabetes mellitus on clinical outcomes following chemotherapy for the patients with pancreatic cancer: a meta-analysis. Acta Diabetol. 56(10):1103-1111.
Publisher | Google Scholor - Ma M, Li W, Xu L, Ping F, Zhang H, Li Y. (2022). Diabetes duration and weight loss are associated with onset age and remote metastasis of pancreatic cancer in patients with diabetes mellitus. J Diabetes, 14(4):261-270.
Publisher | Google Scholor - Roalsø M, Aunan JR, Søreide K. (2020). Refined TNM-staging for pancreatic adenocarcinoma - Real progress or much ado about nothing?. Eur J Surg Oncol, 46(8):1554-1557.
Publisher | Google Scholor - Ansari D, Gustafsson A, Andersson R. (2015). Update on the management of pancreatic cancer: surgery is not enough. World J Gastroenterol, 21(11):3157-3165.
Publisher | Google Scholor - Holly EA, Chaliha I, Bracci PM, Gautam M. (2004). Signs and symptoms of pancreatic cancer: a population-based case-control study in the San Francisco Bay area. Clin Gastroenterol Hepatol, 2(6):510-517.
Publisher | Google Scholor - US Preventive Services Task Force; Owens DK, Davidson KW, Krist AH, Barry MJ, Cabana M, Caughey AB, Curry SJ, Doubeni CA, Epling JW Jr, Kubik M, Landefeld CS, Mangione CM, Pbert L, Silverstein M, Simon MA, Tseng CW, Wong JB. (2019). Screening for Pancreatic Cancer: US Preventive Services Task Force Reaffirmation Recommendation Statement. JAMA, 322(5):438-444.
Publisher | Google Scholor - Chari ST. (2007). Detecting early pancreatic cancer: problems and prospects. Semin Oncol, 34(4):284-294.
Publisher | Google Scholor - Sharma A, Kandlakunta H, Nagpal SJS, Feng Z, Hoos W, Petersen GM, Chari ST. (2018). Model to Determine Risk of Pancreatic Cancer in Patients with New-Onset Diabetes. Gastroenterology, 155(3):730-739.
Publisher | Google Scholor - Chen W, Butler RK, Lustigova E, Chari ST, Wu BU. (2021). Validation of the Enriching New-Onset Diabetes for Pancreatic Cancer Model in a Diverse and Integrated Healthcare Setting. Dig Dis Sci, 66(1):78-87.
Publisher | Google Scholor - Boursi B, Finkelman B, Giantonio BJ, Haynes K, Rustgi AK, Rhim AD, Mamtani R, Yang YX. (2017). A Clinical Prediction Model to Assess Risk for Pancreatic Cancer Among Patients with New-Onset Diabetes. Gastroenterology,152(4):840-850.
Publisher | Google Scholor - Bauer TM, El-Rayes BF, Li X, Hammad N, Philip PA, Shields AF, Zalupski MM, Bekaii-Saab T. (2013). Carbohydrate antigen 19-9 is a prognostic and predictive biomarker in patients with advanced pancreatic cancer who receive gemcitabine-containing chemotherapy: a pooled analysis of 6 prospective trials. Cancer, 119(2):285-292.
Publisher | Google Scholor - Choe JW, Kim HJ, Kim JS, Cha J, Joo MK, Lee BJ, Park JJ, Bak YT. (2018). Usefulness of CA 19-9 for pancreatic cancer screening in patients with new-onset diabetes. Hepatobiliary Pancreat Dis Int, 17(3):263-268.
Publisher | Google Scholor - Yamada T, Minami T, Yamada M, Terauchi Y. (2023). Proposed carbohydrate antigen 19-9 (CA19-9) cut-off values for the detection of pancreatic cancer in patients with poorly controlled diabetes: a real-world study. Endocr J.
Publisher | Google Scholor - Morris-Stiff G, Teli M, Jardine N, Puntis MC. (2009). CA19-9 antigen levels can distinguish between benign and malignant pancreaticobiliary disease. Hepatobiliary Pancreat Dis Int, 8(6):620-626.
Publisher | Google Scholor - Shipman CM, Croucher PI. (2003). Osteoprotegerin is a soluble decoy receptor for tumor necrosis factor-related apoptosis-inducing ligand/Apo2 ligand and can function as a paracrine survival factor for human myeloma cells. Cancer Res, 63(5):912-916.
Publisher | Google Scholor - Shi W, Qiu W, Wang W, Zhou X, Zhong X, Tian G, Deng A. (2014). Osteoprotegerin is up-regulated in pancreatic cancers and correlates with cancer-associated new-onset diabetes. Biosci Trends, 8(6):322-326.
Publisher | Google Scholor - Grote VA, Kaaks R, Nieters A, Tjønneland A, Halkjær J, Overvad K, Skjelbo Nielsen MR, Boutron-Ruault MC, Clavel-Chapelon F, Racine A, Teucher B, Becker S, Pischon T, Boeing H, Trichopoulou A, Cassapa C, Stratigakou V, Palli D, Krogh V, Tumino R, Vineis P, Panico S, Rodríguez L, Duell EJ, Sánchez MJ, Dorronsoro M, Navarro C, Gurrea AB, Siersema PD, Peeters PH, Ye W, Sund M, Lindkvist B, Johansen D, Khaw KT, Wareham N, Allen NE, Travis RC, Fedirko V, Jenab M, Michaud DS, Chuang SC, Romaguera D, Bueno-de-Mesquita HB, Rohrmann S. (2012). Inflammation marker and risk of pancreatic cancer: a nested case-control study within the EPIC cohort. Br J Cancer, 106(11):1866-1874.
Publisher | Google Scholor - Tavano F, Fontana A, Mazza T, Gioffreda D, Biagini T, Palumbo O, Carella M, Andriulli A. (2020). Early-Onset Diabetes as Risk Factor for Pancreatic Cancer: miRNA Expression Profiling in Plasma Uncovers a Role for miR-20b-5p, miR-29a, and miR-18a-5p in Diabetes of Recent Diagnosis. Front Oncol, 10:1567.
Publisher | Google Scholor - Permert J, Larsson J, Westermark GT, Herrington MK, Christmanson L, Pour PM, Westermark P, Adrian TE. Islet amyloid polypeptide in patients with pancreatic cancer and diabetes. N Engl J Med. 1994 Feb 3;330(5):313-8
Publisher | Google Scholor - Liao WC, Huang BS, Yu YH, Yang HH, Chen PR, Huang CC, Huang HY, Wu MS, Chow LP. (2019). Galectin-3 and S100A9: Novel Diabetogenic Factors Mediating Pancreatic Cancer-Associated Diabetes. Diabetes Care, 42(9):1752-1759.
Publisher | Google Scholor - Jenkinson C, Elliott VL, Evans A, Oldfield L, Jenkins RE, O'Brien DP, Apostolidou S, GentryMaharaj A, Fourkala EO, Jacobs IJ, Menon U, Cox T, Campbell F, Pereira SP, Tuveson DA, Park BK, Greenhalf W, Sutton R, Timms JF, Neoptolemos JP, Costello E. (2016). Decreased Serum Thrombospondin-1 Levels in Pancreatic Cancer Patients Up to 24 Months Prior to Clinical Diagnosis: Association with Diabetes Mellitus. Clin Cancer Res, 22:1734-1743.
Publisher | Google Scholor - Kang M, Qin W, Buya M, Dong X, Zheng W, Lu W, Chen J, Guo Q, Wu Y. (2016). VNN1, a potential biomarker for pancreatic cancer-associated new-onset diabetes, aggravates paraneoplastic islet dysfunction by increasing oxidative stress. Cancer Lett, 373(2):241-250.
Publisher | Google Scholor - Huang H, Dong X, Kang MX, Xu B, Chen Y, Zhang B, Chen J, Xie QP, Wu YL. (2010). Novel blood biomarkers of pancreatic cancer-associated diabetes mellitus identified by peripheral blood-based gene expression profiles. Am J Gastroenterol, 105:1661-1669.
Publisher | Google Scholor - Dai X, Pang W, Zhou Y, Yao W, Xia L, Wang C, Chen X, Zen K, Zhang CY, Yuan Y. (2016). Altered profile of serum microRNAs in pancreatic cancer-associated new-onset diabetes mellitus. J Diabetes, 8(3):422-433.
Publisher | Google Scholor - Sud A, Kinnersley B, Houlston RS. (2017). Genome-wide association studies of cancer: current insights and future perspectives. Nat Rev Cancer, 17(11):692-704.
Publisher | Google Scholor - Kozłowska M, Śliwińska A. (2023). The Link between Diabetes, Pancreatic Tumors, and miRNAs-New Players for Diagnosis and Therapy?. Int J Mol Sci, 24(12):10252.
Publisher | Google Scholor - Španinger E, Potočnik U, Bren U. (2020). Molecular Dynamics Simulations Predict That rSNP Located in the HNF-1a Gene Promotor Region Linked with MODY3 and Hepatocellular Carcinoma Promotes Stronger Binding of the HNF-4a Transcription Factor. Biomolecules, 10(12):1700.
Publisher | Google Scholor - Pierce BL, Austin MA, Ahsan H. (2011). Association study of type 2 diabetes genetic susceptibility variants and risk of pancreatic cancer: an analysis of PanScan-I data. Cancer Causes Control, 22(6):877-883.
Publisher | Google Scholor - Hu Y, Zeng N, Ge Y, et al. (2022). Identification of the Shared Gene Signatures and Biological Mechanism in Type 2 Diabetes and Pancreatic Cancer. Front Endocrinol (Lausanne), 13:847760.
Publisher | Google Scholor - Zhang R, Kan JB, Lu S, Tong P, Yang J, Xi L, et al. (2019). S100a16-Induced Adipogenesis Is Associated with Up-Regulation Of 11 β-Hydroxysteroid Dehydrogenase Type 1 (11β-Hsd1). Biosci Rep, 39.
Publisher | Google Scholor - Kan J, Zhao C, Lu S, Shen G, Yang J, Tong P, et al. (2019). S100a16, A Novel Lipogenesis Promoting Factor in Livers Of Mice And Hepatocytes in Vitro. J Cell Physiol, 234:21395-21406.
Publisher | Google Scholor - Allgöwer C, Kretz AL, Von Karstedt S, Wittau M, Henne-Bruns D, Lemke J. (2020). Friend Or Foe: S100 Proteins in Cancer. Cancers (Basel), 12(8).
Publisher | Google Scholor - Prizment AE, Gross M, Rasmussen-Torvik L, Peacock JM, Anderson KE. (2012). Genes related to diabetes may be associated with pancreatic cancer in a population-based case-control study in Minnesota. Pancreas, 41(1):50-53.
Publisher | Google Scholor - Saito T, Mizukami H, Umetsu S, et al. (2017). Worsened outcome in patients with pancreatic ductal carcinoma on long-term diabetes: association with E-cadherin1 (CDH1) promoter methylation. Sci Rep, 7(1):18056.
Publisher | Google Scholor - Vincent EE, Yaghootkar H. (2020). Using genetics to decipher the link between type 2 diabetes and cancer: shared aetiology or downstream consequence?. Diabetologia, 63(9):1706-1717.
Publisher | Google Scholor - Wu L, Rabe KG, Petersen GM. (2015). Do variants associated with susceptibility to pancreatic cancer and type 2 diabetes reciprocally affect risk?. PLoS One, 10(2):0117230.
Publisher | Google Scholor - Tan J, You Y, Guo F, Xu J, Dai H, Bie P. (2017). Association of elevated risk of pancreatic cancer in diabetic patients: A systematic review and meta-analysis. Oncol Lett,13(3):1247-1255.
Publisher | Google Scholor - Kautzky-Willer A, Thurner S, Klimek P. Use of statins offsets insulin-related cancer risk. J Intern Med. 2017;281(2):206-216.
Publisher | Google Scholor - Currie CJ, Poole CD, Gale EA. (2009). The influence of glucose-lowering therapies on cancer risk in type 2 diabetes. Diabetologia, 52(9):1766-1777.
Publisher | Google Scholor - Bosetti C, Rosato V, Li D, et al. (2014). Diabetes, antidiabetic medications, and pancreatic cancer risk: an analysis from the International Pancreatic Cancer Case-Control Consortium. Ann Oncol, 25(10):2065-2072.
Publisher | Google Scholor - Wang G, Yin L, Peng Y, et al. (2019). Insulin promotes invasion and migration of KRASG12D mutant HPNE cells by upregulating MMP-2 gelatinolytic activity via ERK- and PI3K-dependent signalling. Cell Prolif, 52(3):12575.
Publisher | Google Scholor - Bridges HR, Jones AJ, Pollak MN, Hirst J. (2014). Effects of metformin and other biguanides on oxidative phosphorylation in mitochondria. Biochem J, 462(3):475-487.
Publisher | Google Scholor - Hu J, Fan HD, Gong JP, Mao QS. (2023). The relationship between the use of metformin and the risk of pancreatic cancer in patients with diabetes: a systematic review and meta-analysis. BMC Gastroenterol, 23(1):50.
Publisher | Google Scholor - Wang Z, Lai ST, Xie L, et al. (2014). Metformin is associated with reduced risk of pancreatic cancer in patients with type 2 diabetes mellitus: a systematic review and meta-analysis. Diabetes Res Clin Pract, 106(1):19-26.
Publisher | Google Scholor - Sadeghi, Navid, et al. (2012).
Publisher | Google Scholor - Ren D, Qin G, Zhao J, et al. (2020). Metformin activates the STING/IRF3/IFN-β pathway by inhibiting AKT phosphorylation in pancreatic cancer. Am J Cancer Res, 10(9):2851-2864.
Publisher | Google Scholor - Zhao HW, Zhou N, Jin F, Wang R, Zhao JQ. (2020). Metformin reduces pancreatic cancer cell proliferation and increases apoptosis through MTOR signaling pathway and its dose-effect relationship. Eur Rev Med Pharmacol Sci, 24(10):5336-5344.
Publisher | Google Scholor - Chaiteerakij R, Petersen GM, Bamlet WR, et al. (2016). Metformin Use and Survival of Patients with Pancreatic Cancer: A Cautionary Lesson. J Clin Oncol, 34(16):1898-1904.
Publisher | Google Scholor - Lamont BJ, Andrikopoulos S. (2014). Hope and fear for new classes of type 2 diabetes drugs: is there preclinical evidence that incretin-based therapies alter pancreatic morphology? J Endocrinol, 221:43-61.
Publisher | Google Scholor - Suryadevara V, Roy A, Sahoo J, et al. (2022). Incretin based therapy and pancreatic cancer: Realising the reality. World J Gastroenterol, 28(25):2881-2889.
Publisher | Google Scholor - Nreu B, Dicembrini I, Tinti F, Mannucci E, Monami M. (2020). Pancreatitis and Pancreatic Cancer in Patients with Type 2 Diabetes Treated with Glucagon- Like Peptide-1 Receptor Agonists: An Updated Meta-Analysis of Randomized Controlled Trials. Minerva Endocrinol.
Publisher | Google Scholor - Pinto LC, Falcetta MR, Rados DV, Leitão CB, Gross JL. (2019). Glucagon-like peptide-1 receptor agonists and pancreatic cancer: a meta-analysis with trial sequential analysis. Sci Rep, 9(1):2375.
Publisher | Google Scholor - Zhao H, Wang L, Wei R, et al. (2014). Activation of glucagon-like peptide-1 receptor inhibits tumourigenicity and metastasis of human pancreatic cancer cells via PI3K/Akt pathway. Diabetes Obes Metab. 16(9):850-860.
Publisher | Google Scholor - Zheng Y, Wu C, Yang J, et al. (2020). Insulin-like growth factor 1-induced enolase 2 deacetylation by HDAC3 promotes metastasis of pancreatic cancer. Signal Transduct Target Ther, 5(1):53.
Publisher | Google Scholor - Mentlein R, Gallwitz B, Schmidt WE. (1993). Dipeptidyl-peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon-like peptide-1(7-36) amide, peptide histidine methionine and is responsible for their degradation in human serum. Eur J Biochem, 214(3):829-835.
Publisher | Google Scholor - Azoulay L, Filion KB, Platt RW, Dahl M, Dormuth CR, Clemens KK, Durand M, Juurlink DN, Targownik LE, Turin TC, Paterson JM. (2016). Ernst P; Canadian Network for Observational Drug Effect Studies Investigators. Incretin based drugs and the risk of pancreatic cancer: international multicentre cohort study. BMJ.
Publisher | Google Scholor - Dicembrini I, Montereggi C, Nreu B, Mannucci E, Monami M. (2020). Pancreatitis and pancreatic cancer in patientes treated with Dipeptidyl Peptidase-4 inhibitors: An extensive and updated meta-analysis of randomized controlled trials. Diabetes Res Clin Pract, 159:107981.
Publisher | Google Scholor - Ferrannini E, Solini A. (2012). SGLT2 inhibition in diabetes mellitus: rationale and clinical prospects. Nat Rev Endocrinol, 8(8):495-502.
Publisher | Google Scholor - Duan X, Wang W, Pan Q, Guo L. (2021). Type 2 Diabetes Mellitus Intersects With Pancreatic Cancer Diagnosis and Development. Front Oncol, 11:730038.
Publisher | Google Scholor