v
Search
Advanced Search

Publications > Journals > Cancer Screening and Prevention > Article Full Text

  • OPEN ACCESS

Diabetes Mellitus and Early Detection of Pancreatic Cancer

  • Yi-Bo Fu1,#,
  • Gang Yang1,#,
  • Jiang-Dong Qiu1 and
  • Tai-Ping Zhang1,2,* 
 Author information
Cancer Screening and Prevention   2022;1(1):64-71

doi: 10.14218/CSP.2022.00013

Abstract

Pancreatic ductal adenocarcinoma (PDAC) is most often detected at an advanced stage due to a lack of symptoms at the first stage. PDAC is relatively uncommon, and screening of the asymptomatic population is not feasible or cost-effective. Therefore, screening of individuals in high-risk groups is recommended. Diabetes mellitus (DM) is associated with PDAC, and patients with DM are recognized as a high-risk group for PDAC. Here, we review the complex relationship between pancreatic cancer and DM, including the role of diabetes as a risk factor for pancreatic cancer and its role in inducing the destruction of islet β cells and insulin resistance. We also review the current study about discriminating DM with pancreatic cancer from normal DM and the model for early screening of pancreatic cancer in DM.

Keywords

Pancreatic cancer, Diabetes mellitus, Early screening, Insulin resistance, Antidiabetic drugs

Pancreatic cancer and diabetes

Diabetes

Diabetes mellitus (DM) is a general term for heterogeneous disturbances of metabolism with the performance of chronic hyperglycemia.1 Hyperglycemia results from defects in insulin secretion, insulin action, or both. Classification of DM depends on etiology and clinical manifestations. DM can be divided into three types: type 1 diabetes (T1DM), type 2 diabetes (T2DM), and other specific types of diabetes.2

T1DM, once called insulin-dependent diabetes or juvenile-onset diabetes, is characterized as cellular-mediated autoimmune destruction of the β-cells in the pancreas. As β-cells are destructed, insulin is commonly deficient in T1DM patients. Different types of exogenous insulin are the primary treatment.3 T1DM can occur at any age but usually in childhood and adolescence. Classic symptoms of T1DM (caused by insulin deficiency and hyperglycemia) usually start rapidly (days to weeks), including polyuria, polydipsia, weight loss, and ketoacidosis.4

T2DM accounts for the vast majority of those with diabetes, encompassing patients with insulin resistance and relative insulin deficiency.5 So, T2DM patients do not need insulin treatment to control blood glucose levels for the first time.6 Most patients with T2BD have obesity, which leads to some degree of insulin resistance.7 Unlike T1DM, ketoacidosis seldom occurs spontaneously in this type of diabetes.

Other specific types of diabetes cannot be classified as T1DM or T2DM, including DM caused by genetic defects of β-cells, diseases of the exocrine pancreas (e.g., pancreatitis, trauma, infection, pancreatectomy, and pancreatic cancer), hormones imbalance (e.g., growth hormone, cortisol, epinephrine, and glucagon), infection (for instance, congenital rubella that can destruct β-cells in pancreas), and some drugs inducing DM.2

Pancreatic cancer

Pancreatic cancer is one of the most lethal malignancies, of which the main pathological component is pancreatic ductal adenocarcinoma (PDAC).8 Because of the latency and lack of symptoms at an early stage, pancreatic cancer is usually detected at an advanced stage, and most treatment regimens are ineffective.9 Therefore, early detection of pancreatic cancer is essential to improve prognosis. The cause of pancreatic cancer is complex and multifactorial. However, some factors have gradually been proven as risk factors for PDAC, for example, cigarettes and pancreatic cancer family history.10 In recent years, researchers generally revealed that DM had a close association with PDAC, and early detection of PDAC depending on DM seems possible.11 This review will discuss the relationship between DM and PC in detail and analyze the role of DM in the early detection of pancreatic cancer.

DM and the risk of PDAC

The relationship between DM and pancreatic cancer has been studied for years. Plenty of evidence supports that type 2 diabetes is a risk factor for developing pancreatic cancer. A meta-analysis involving 26 cohort studies raised that DM was associated with an increased risk of PDAC (relative risk [RR] = 3.69; 95% confidence interval [CI] = 3.12–4.37, p < 0.05).12 Multiple mechanisms may explain the role of diabetes in promoting cancer initiation progression. On the one hand, a pathological state of diabetes leads to the initiation and progression of PDAC. On the other hand, some antidiabetics are also shown to be associated with the risk of PDAC.

Diabetes: a risk factor of PDAC

Hyperglycemia and the risk of PDAC incidence

Hyperglycemia is a risk factor of PDAC. A prospective, nested case-control study involving 449 case patients and 982 control subjects showed that a higher level of HbA1c was associated with an increased risk for pancreatic cancer (highest vs. lowest quintiles of HbA1c, odds ratio [OR] = 1.14, 95%CI = 1.17 to 2.72, p = 0.04).13 Another nested case-control study of 278 cases and 826 matched controls found that long-term patterns of higher fasting blood glucose (FBG) are associated with a higher risk of PDAC (individuals with long-standing hyperglycemia vs. consistently normoglycemic individuals, OR = 2.02, 95%CI = 1.24–3.31, p < 0.05).14 A dose-response meta-analysis evaluating blood glucose concentration and risk of pancreatic cancer showed that a strong linear dose-response association between fasting blood glucose concentration and the rate of pancreatic cancer was detected (per 0.56 mmol/L, RR = 1.14, 95%CI = 1.06–1.22; p < 0.001).15 Preconditioning PDAC cells with glucose medium showed that a high-glucose environment increased cell viability and sphere formation.16

Many theories explain its role as a carcinogen, including initiation, progression, and metastasis. Chen et al. reported that hyperglycemia promotes PDAC initiation and progression by activating the wingless/integrated (Wnt)/β—catenin signaling pathway.17 Sato et al. found that hyperglycemia in PDAC cells could strengthen the STAT3 phosphorylation and MYC expression, aggravating cancer progression.16 Apart from tumor initiation and progression, hyperglycemia can also promote epithelial-mesenchymal transition (EMT) of PDAC. A study showed that hyperglycemia could stimulate HIF-1α expression in PDAC, leading to hypoxia in the pancreatic parenchyma and promoting the metastatic ability in PDAC.18 Another study raised that hyperglycemia could promote the EMT of PDAC via stimulating hydrogen peroxide, as hyperglycemic mice contain a higher plasma hydrogen peroxide level, while hydrogen peroxide scavenger could reverse hyperglycemia-induced tumor metastasis.19 In addition, hyperglycemia promotes the acquisition of pancreatic cancer stem cells (CSCs), essential for EMT.20

Hyperinsulinemia, insulin resistance, and the risk of PDAC incidence

Insulin resistance and hyperinsulinemia are risk factors of PDAC, and they can be detected in most T2DM patients.21 The relationship between insulin resistance and hyperinsulinemia is complex. Traditionally it was believed that insulin resistance provoked the development of hyperinsulinemia in T2DM patients, while in recent years, some researchers came up with a reverse order and placed hyperinsulinemia upstream of insulin resistance.21

Hyperinsulinemia is associated with a higher risk of PDAC. A prospective, nested case-control study involving 449 case patients and 982 control subjects revealed that hyperinsulinemia was associated with an increased risk for pancreatic cancer (highest vs. lowest quintiles of insulin, OR = 1.57, 95%CI=1.08–2.30, p = 0.002).13 A case-cohort prospective study of 29,133 male Finnish smokers aged 50 to 69 years showed that higher insulin concentrations and insulin resistance might lead to a higher risk of pancreatic cancer (insulin: highest vs. lowest quartile, HR = 2.90; 95%CI = 1.22–6.92; p = 0.005; insulin resistance: highest vs. lowest quartile, HR = 2.71; 95%CI = 1.22–6.92; p = 0.006).22

Insulin is thought to promote carcinogenesis in many kinds of cancer, including prostate, colorectal, breast, and pancreatic cancer, by stimulating insulin receptors (IRs) and insulin-like growth factors (IGFs) receptors and then stimulating the mitogen-activated protein kinase (MAPK) pathway.23,24 Yang et al. reported that insulin could enhance cell proliferation and fibrosing responses in cultured activated pancreatic stellate cells by stimulating IR/IGF-1R and then Akt/mechanistic target of rapamycin (mTOR)/p70S6K signaling pathway.25

Obesity and the risk of PDAC incidence

Obesity is closely associated with T2DM. Obesity is believed to be a promoter of T2DM that can stimulate excessive fat accumulation and obesity.26 The most commonly used criterion for determining obesity is the body mass index (BMI). According to the World Health Organization (WHO), obesity is defined as a BMI of 30 kg/m2 or greater. Clinical studies have revealed that obesity could increase the risk of PDAC. A study analyzing the National Cancer Institute Pancreatic Cancer Cohort Consortium (PanScan) reported a positive association between BMI and pancreatic cancer risk (the highest vs. lowest quartile of BMI, OR = 1.33, 95%CI = 1.04–1.69; p < 0.03).27 Similar conclusion was drawn from many other studies and meta-analyses.28–30

Multiple molecular mechanisms and signaling pathways seem to be involved in the association of obesity with PDAC.

Leptin is a kind of adipokines synthesized primarily in white adipose tissue. Obesity is often associated with leptin resistance and overexpressed leptin.31 Epidemiology studies suggested that leptin may be associated with the incidence of PDAC in the obesity group. A pool analysis from three cohorts revealed an association between increasing leptin concentration and pancreatic cancer (quintile 5 vs. quintile 1: OR = 2.55, 95%CI = 1.23–5.27, p = 0.004).32 However, a meta-analysis involving 17 articles drew an opposite conclusion that circulating leptin levels were significantly lower in patients with PC than in those without PC (SMD = 0.830, 95%CI = 0.497–1.164, p < 0.001), although they reported that the level of AdipoQ (another kind of adipokines) was significantly higher in PC patients in the same time.33 Furthermore, the article explored the possible reasons for the unusual decrease of leptin in PC patients and found that PC patients with cachexia showed a significantly lower leptin level compared with those without cachexia, suggesting that cachexia may lead to the consumption of leptin.33

Many studies have proved that leptin showed oncogenic roles at the cell level. Xu et al. found that in PDAC cells, leptin could promote cell proliferation and increase glucose uptake by stimulating the ATK signal pathway, as the AKT inhibitor significantly counteracted the effects of leptin stimulation.34 In addition, researchers found that leptin treatment promoted the expression of the Notch signaling pathway, which plays a critical role in cell proliferation, suggesting that the leptin-Notch axis may also play an important part.35

Studies proved that obesity promoted pro-inflammatory response (including increased expression of IL-1β, IL-6, TNF-α, IL-12, or CXCL1) and immune cell infiltration in PDACs.36 Furthermore, obesity-induced IL-1β mediated PSC activation and PDAC progression.36 In addition, the study also revealed that obesity could induce a steatotic and fibrotic microenvironment in PDACs, which could also be enhanced by inflammation, and then augmented desmoplasia and tumor growth.

VEGF also seems to play a role in obesity-induced tumor initiation. Obesity was reported to be associated with a higher level of vascular endothelial growth factor (VEGF). Research showed that obesity could induce local hypoxia and then the overexpression of VEGF.37 In PDAC, VEGF stimulation led to hypoxia-inducible factor-α (HIF-α) up-regulation, which promoted glycolysis and PDAC progression.38

Antidiabetics and the risk of PDAC incidence

At present, there are various kinds of antidiabetic drugs, including biguanides, sulphonylureas, thiazolidinediones, sodium-glucose co-transporter-2 (SGLT-2) inhibitors, glucagon-like peptide-1 receptor agonists (GLP1-RA), dipeptidyl peptidase-4 (DPP-4) inhibitors, and alpha-glucosidase inhibitors.39 Studies have shown that multiple antidiabetic drugs are associated with the incidence of PDAC.

Metformin and the risk of PDAC

Metformin is the recommended first-line treatment for T2DM and is widely prescribed worldwide.40 Metformin traditionally functions as an antidiabetic drug by reducing hepatic gluconeogenesis.41 In recent years, various epidemiologic studies revealed that metformin might have a relationship with a reduced risk of PDAC. For example, a case-control study involving 973 patients with pancreatic adenocarcinoma and 863 controls showed that people with diabetes taking metformin had a significantly lower risk of pancreatic cancer than those without metformin (OR = 0.38; 95% CI = 0.22–0.69; p = 0.001).42 Similar conclusion was drawn by another case-control study in 2015 (OR = 1.46; 95% CI = 0.85–2.52).43 A meta-analysis in 2013 containing 37 studies concluded that metformin use led to a 46% risk reduction of PDAC.44 Some mechanisms may explain the anticancer effect of metformin. For example, Ren et al. reported that metformin might activate the STING/IRF3/IFN-β pathway by inhibiting AKT signaling in PDAC cells and then promote the infiltration of immune cells in the tumor microenvironment (TME).45 Zhao et al. found that metformin could induce the expression of pro-apoptosis proteins caspase-3 and Bax and reduce the expression of anti-apoptosis protein Bcl-2 through the MTOR signaling pathway, leading to the suppression of PDAC.46 In conclusion, metformin has shown its function as an anticancer drug in PDAC patients.

GLP1-RAs and the risk of PDAC

Glucagon-like peptide-1 (GLP-1) is an incretin secretory molecule secreted after eating, which can lower glucose concentrations by stimulating insulin secretion and suppressing glucagon release.47

Apart from the function as a kind of antidiabetic drug, GLP-RAs (GLP-receptor agonists) have been associated with PDAC in recent years. A study reported that using GLP-RAs was linked to a higher risk of PDAC.48 Koehler et al. studied the effect of GLP1 in mice and found that GLP1 could activate the growth and survival of PDAC. In addition, they reported that pancreatic cancer cells expressed GLP1 receptors.49 However, some later studies drew a contradictory conclusion that incretin use was not associated with pancreatic cancer.50 A meta-analysis containing twelve trials revealed that GLP1-RA did not increase the risk of PDAC compared to other treatments (OR 1.06; 95% CI = 0.67–1.67; I2 14%).51 One limitation of current studies is that the follow-up duration may be insufficient to draw a convincing conclusion. More studies with long-term follow-up are needed to ensure the safety of incretin therapy.

DPP4-inhibitor and the risk of PDAC

Dipeptidyl peptidase-4 (DPP4) is a serine protease that promotes the degradation of GLP-1.52 Therefore, DPP4-inhibitor (DDP4i) can suppress the degradation of GLP-1 and be used as a kind of hypoglycemic agent.

A cohort in 2014 with a six-month follow-up showed that the risk of pancreatic cancer with DPP-4 inhibitor treatment was not higher than with other antidiabetic treatments.53 A cohort study in 2019, including 33,208 patients newly diagnosed with type 2 diabetes who were treated with antidiabetic drugs, revealed that DPP4i use is associated with increased risks of pancreatic cancer (HR 1.81, 95% CI = 1.16–2.82; p = 0.009).54 So far, no studies have explained the relationship. However, a meta-analysis in 2020 involving 157 trials suggested that DDP4i were not associated with an increased risk of pancreatic cancer (OR = 0.86, 95% CI = 0.60–1.24).55 Therefore, more large-scale RCT studies are needed to clarify the association between DD4Pi and PDAC.

In conclusion, diabetes can induce the incidence of pancreatic cancer in many ways.

PDAC and the risk of DM

As the review mentioned in section 1.1, specific types of DM contain diabetes caused by diseases of the exocrine pancreas, for instance, pancreatic cancer. The relationship between pancreatic cancer and the incidence of DM is far more complex, which can not be explained simply by the destruction of pancreatic β-cells due to PDAC. This section will discuss the role of PDAC as a risk factor for DM and the possible function of DM in the early detection of pancreatic cancer.

PDAC as a risk factor of diabetes

While type 2 diabetes has been recognized as a risk factor for pancreatic cancer, shreds of evidence showed that diabetes might be a manifestation of pancreatic cancer.56 Diabetes is present in about 80% of patients with PDAC.57 A study on the prevalence of diabetes mellitus in pancreatic cancer found that patients with PDAC had a significantly higher prevalence (68%, p < 0.0001) of DM compared with patients with other kinds of cancers and noncancer controls (23.5%).58 Another study comparing PC patients and controlled people found that DM was more prevalent (47% vs. 7%; p < 0.001) among cases compared with control, and the DM was predominantly new onset DM.59

PDAC and β cells dysfunction

Patients with PDAC are usually accompanied by a decrease in the size of islets and a reduction in the number of β cells.60 The β cell response (including response to an oral glucose load, hyperglycaemic clamp, or glucagon stimulation) was also impaired in PDAC patients.61 Using a mouse model of KrasG12D-driven PDAC, Parajuli et al. reported that PDAC progression led to an increased expression of TGF-β signaling pathway, and then caused erosion of β-cell mass through apoptosis.62 Exosomes from conditioned media of pancreatic cancer could enter β cells and inhibit insulin secretion, suggesting that PC causes paraneoplastic β-cell dysfunction by improving exosomes into circulation that inhibit insulin secretion.63 Exosomes mediated adrenomedullin (AM) production, which was delivered to the β-cell, induced endoplasmic reticulum (ER) stress, a failure of the unfolded protein response (UPR), and increased β-cell dysfunction and death.

PDAC and insulin resistance

A study reported that PDAC might produce substances that induce insulin resistance. Insulin resistance was relieved after subtotal pancreatectomy for pancreatic cancer.64 According to skeletal muscle biopsies from pancreatic cancer patients with or without diabetes, insulin receptor (IR) binding, IR mRNA, and IR substrate-1 content were all normal in PC patients. However, the fractional velocity of glycogen synthase was decreased in the diabetic pancreatic cancer group, suggesting that the PC-related insulin resistance was associated with a post-IR defect.65

Further research found that PC- derived-exosomes inhibit insulin-PI3K-Akt signaling and thereby interfere with GLUT4 uptake, which may explain why PC could induce insulin resistance in skeletal muscle.66 In addition, Sagar et al. reported that pancreatic cancer exosomes were readily internalized into adipocytes, leading to lipolysis in adipose tissue.67 This process was mediated by exosomal adrenomedullin, which interacted with its receptor on the adipocytes. It activated p38 and extracellular signal-regulated (ERK1/2) mitogen-activated protein kinases and promoted lipolysis by phosphorylating hormone-sensitive lipase. Meanwhile, adipocyte lipolysis is associated with insulin resistance in T2DM.68

New-onset DM as an early sign of pancreatic cancer

As pancreatic cancer is a risk factor of DM, new-onset DM may be a vital early sign of PC. A cohort study of 48 995 African Americans and Latinos examining the relationships between new-onset diabetes and PC incidence showed that diabetes was associated with a higher risk of PC (HRage75 = 2.39, 95%CI = 1.91–2.98). The association for recent-onset diabetes (three or fewer years) was 2.3-fold greater (HR = 3.71, 95%CI = 2.83–4.88) than for long-standing diabetes (more than three years; HR = 1.61, 95%CI = 1.18–2.21).69 The researchers conducted a population-based cohort study of different races, which showed that individuals who received a recent diagnosis of diabetes had an almost 7-fold increase in the risk of pancreatic cancer (RR = 6.91; 95%CI = 5.76–8.30).56

We concluded studies about pancreatic cancer and DM with different topics (Table 1).13–19,22–25,27–30,32–38,42–46,48–51,53–55,58–68

Table 1

Studies about pancreatic cancer and DM with different topics

TopicStudy
Diabetes: a risk factor of PDAC
  Hyperglycemia and the risk of PDAC incidence1319
  Hyperinsulinemia and the risk of PDAC incidence13,22
  Insulin resistance and the risk of PDAC incidence2325
  Obesity and the risk of PDAC incidence2730,3238
Antidiabetics and the risk of PDAC incidence
  Metformin4246
  GLP1-RAs4851
  DPP4-inhibitor5355
PDAC: a risk factor of diabetes
  PDAC as a risk factor of diabetes58,59
  PDAC and β cells dysfunction6063
  PDAC and insulin resistance6468

DM in the early detection of PC

In recent years, DM has gradually been regarded as a manifestation of pancreatic cancer. However, more importantly, DM appears typically 2–3 years earlier than PC, making DM important in early PC detection.

Diagnosis of PC

Numerous international guidelines recommend computed tomography (CT) as an initial measure for diagnosing suspected PC.70,71 Because of its low cost, CT is preferred over magnetic resonance imaging (MRI) as the first-line modality. In the CT set, the tumor detection rates were about 90%, and MRI shared a similar sensitivity.72 The classic CT feature of PC includes a hypoattenuating pancreatic mass, pancreatic duct dilatation, and atrophy of the upstream pancreas. Therefore, CT is also applied to screen patients at high risk for PC.73 In the early stage of PC, inhomogeneous parenchyma, interruption of the pancreatic duct, and loss of fatty marbling can be seen in the CT set, which was helpful for early detection of PC.74,75 Other measures for the diagnosis of PC include magnetic resonance cholangiopancreatography (MRCP) and endoscopic ultrasound (EUS).

Early detection of PC in DM patients

Given that the prevalence of PDAC in the general population is relatively low, population CT screening is cost-inefficient and challenging to achieve. Therefore, the identification of high-risk populations for pancreatic cancer is essential.

The key to early PC detection in DM patients is differentiating pancreatic cancer-related diabetes (PCRD) from common DM. New onset diabetes in PDAC often differs from T2DM in significant ways. For example, PCRD is usually concomitant with weight loss, while T2DM is commonly associated with weight gain.76 Besides, diabetes can be better controlled by weight loss in common DM, which is barely seen in PCRD. A cohort study using multivariate logistic regression analysis reported that BMI, the age of onset of diabetes, HBV infection, TBIL, ALT, Cr, APO-A1, and WBC are factors that could differentiate PC + DM from common DM.77

NOD is vital in the early diagnosis of pancreatic cancer. Sharma et al. developed a model called enriching new-onset diabetes for pancreatic cancer (END-PAC) to evaluate the risk of pancreatic cancer in patients with new-onset diabetes.78 The model contains the change of blood glucose, change of weight, and age at glycemically-defined new-onset diabetes. More significant changes in blood glucose, lower change of weight, and higher age lead to a higher risk of PC in patients with new-onset diabetes. Several studies have validated the END-PAC model. For example, a study by Boursi et al. focused on 5,408 patients with NOD, and according to their research comparing the high-risk group compared with the low-risk group, the sensitivity, specificity, positive predictive value, and negative predictive value of the model were 54.2%, 76.98%, 2.57%, and 99.4%, respectively.79 Similarly, a study of 13,947 NOD patients in a healthcare setting validated the END-PAC model. At the 3+ threshold, the sensitivity, specificity, PPV, and NPV were 62.6%, 78.5%, 2.0%, and 99.7%, respectively.80 In conclusion, current studies supported the robustness, generalizability, and clinical applicability of the END-PAC model. However, the original investigation and validation studies have some limitations. For instance, the definition of NOD was restricted and may exclude certain patients who truly had NOD. Besides, all the studies were retrospective. Therefore, more efforts are needed to validate the screening strategies for patients with NOD in real-world settings.

In recent years, many other metabolites have been shown to play a role in the discrimination of DM from PC+DM. With the help of 1H mass spectrometry and nuclear magnetic resonance (NMR) spectroscopy, alterations of many plasma metabolite concentrations can discriminate T2DM and PC, suggesting that some proposed plasma metabolite biomarkers can be included in the model to evaluate the risk of pancreatic cancer in patients with new-onset diabetes.81 Similarly, He et al. analyzed their data using principal components analysis (PCA) and orthogonal projection to latent structures (OPLS). They identified 62 differential metabolites that may function as individual biomarkers of PC.82 The droplet digital PCR (ddPCR) confirmed that miR-20b-5p, a kind of miRNA, showed a higher level in PC patients with new-onset diabetes, suggesting that miR-20b-5p achieved higher diagnostic accuracy than PC with new-onset diabetes.83

A study evaluated the cost-effectiveness of PDAC early detection strategy targeting high-risk new-onset diabetes patients using MRCP and positive MRI underwent EUS/fine-needle aspiration (FNA). They found out that considering a willingness to pay (WTP) threshold between $100,000 and $150,000 per quality-adjusted life-year, a minimum predicted three-year PDAC risk of 1.0% to 2.0% may be cost-effective.84 Advances in the early detection of PDAC in NOD were concluded in Table 2.78–80,84

Table 2

Advances in the early detection of PDAC in NOD

TopicStudyConclusion
END-PACSharma et al78END-PAC* can evaluate the risk of pancreatic cancer in patients with new-onset diabetes according to change in blood glucose, change of weight, and age at glycemically-defined new-onset diabetes
Boursi et al79The sensitivity, specificity, positive predictive value, and negative predictive value of the END-PAC model were 54.2%, 76.98%, 2.57%, and 99.4%, respectively.
Chen W. et al80At the 3+ threshold, the sensitivity, specificity, PPV, and NPV of the END-PAC model were 62.6%, 78.5%, 2.0%, and 99.7%.
Cost-effectivenessWang L., et al84Considering a WTP* threshold between $100,000 and $150,000 per quality-adjusted life-year, a minimum predicted 3-year PDAC risk of 1.0% to 2.0% may be cost-effective

Conclusions

Substantial studies have focused on the complex relationship between PC and DM. On the one hand, DM is a risk factor of PC, as hyperglycemia, hyperinsulinemia, insulin resistance, and obesity all improve PC initiation and progression. On the other hand, DM seems to be a manifestation of PC, considering that PC can lead to the destruction of islet β cells and insulin resistance. The incidence of PC in patients with DM is apparently higher than in normal people, suggesting that early PC screening in patients with DM is significant. The END-PAC model can help to identify people at high risk for PC in patients with new-onset diabetes. In addition, some metabolites have been shown to help discriminate PC + DM from normal DM. In conclusion, these methods can help identify the high-risk population of PC in diabetic patients and diagnose PC earlier. However, the practical application value and effect still need clinical verification.

Abbreviations

AM: 

adrenomedullin

BMI: 

body mass index

CSCs: 

cancer stem cells

CT: 

computed tomography

ddPCR: 

droplet digital PCR

DM: 

diabetes mellitus

DPP-4: 

dipeptidyl peptidase-4

EMT: 

epithelial-mesenchymal transition

END-PAC: 

enriching new-onset diabetes for pancreatic cancer

ER: 

endoplasmic reticulum

FNA: 

fine-needle aspiration

GLP1-RA: 

glucagon-like peptide-1 receptor agonists

HIF-α: 

hypoxia-inducible factor-α

IGFs: 

insulin-like growth factors

IR: 

insulin receptor

MAPK: 

mitogen-activated protein kinase

MRI: 

magnetic resonance imaging

mTOR: 

mechanistic target of rapamycin

NMR: 

nuclear magnetic resonance

NOD: 

new-onset diabetes

OPLS: 

orthogonal projection to latent structures

PCA: 

principal components analysis

PCRD: 

pancreatic cancer-related diabetes

PDAC: 

pancreatic ductal adenocarcinoma

SGLT-2: 

sodium-glucose co-transporter-2

T1DM: 

type 1 diabetes

T2DM: 

type 2 diabetes

TME: 

tumor microenvironment

UPR: 

unfolded protein response

VEGF: 

vascular endothelial growth factor

WHO: 

world health organization

WTP: 

willingness to pay

Declarations

Acknowledgement

None.

Funding

This study was supported by grants from the National Key R&D Program of China (2018YFE0118600), the National Natural Science Foundation of China (No.81972258), the National Multidisciplinary Cooperative Diagnosis and Treatment Capacity Building Project for Major Diseases, China Postdoctoral Science Foundation (2021T140071 and 2021M690462), and National Natural Science Foundation of China (No. 81772639, No.81802475, No.81972258, No. 82103016).

Conflict of interest

TPZ has been an associate editor of Cancer Screening and Prevention since March 2022. The authors declare no other competing interests.

Authors’ contributions

YBF performed literature searches, drafted the literature review, and revised and finalized the manuscript. GY revised the manuscripts. JDQ revised the manuscripts. TPZ conceptualized, revised, and finalized the manuscript. All authors have read and agreed to the published version of the manuscript.

References

  1. Kerner W, Brückel J, German Diabetes Association. Definition, classification and diagnosis of diabetes mellitus. Exp Clin Endocrinol Diabetes 2014;122(7):384-386 View Article PubMed/NCBI
  2. American Diabetes Association. Diagnosis and classification of diabetes mellitus. Diabetes Care 2013;36(Suppl 1):S67-S74 View Article PubMed/NCBI
  3. Li W, Huang E, Gao S. Type 1 Diabetes Mellitus and Cognitive Impairments: A Systematic Review. J Alzheimers Dis 2017;57(1):29-36 View Article PubMed/NCBI
  4. Katsarou A, Gudbjörnsdottir S, Rawshani A, Dabelea D, Bonifacio E, Anderson BJ, et al. Type 1 diabetes mellitus. Nat Rev Dis Primers 2017;3:17016 View Article PubMed/NCBI
  5. Kao KT, Sabin MA. Type 2 diabetes mellitus in children and adolescents. Aust Fam Physician 2016;45(6):401-406 PubMed/NCBI
  6. Xu L, Li Y, Dai Y, Peng J. Natural products for the treatment of type 2 diabetes mellitus: Pharmacology and mechanisms. Pharmacol Res 2018;130:451-465 View Article PubMed/NCBI
  7. Al-Sulaiti H, Diboun I, Agha MV, Mohamed FFS, Atkin S, Dömling AS, et al. Metabolic signature of obesity-associated insulin resistance and type 2 diabetes. J Transl Med 2019;17(1):348 View Article PubMed/NCBI
  8. Vincent A, Herman J, Schulick R, Hruban RH, Goggins M. Pancreatic cancer. Lancet 2011;378(9791):607-620 View Article PubMed/NCBI
  9. Niederhuber JE, Brennan MF, Menck HR. The National Cancer Data Base report on pancreatic cancer. Cancer 1995;76(9):1671-1677 View Article PubMed/NCBI
  10. Blackford A, Parmigiani G, Kensler TW, Wolfgang C, Jones S, Zhang X, et al. Genetic mutations associated with cigarette smoking in pancreatic cancer. Cancer Res 2009;69(8):3681-3688 View Article PubMed/NCBI
  11. Risch HA. Diabetes and Pancreatic Cancer: Both Cause and Effect. J Natl Cancer Inst 2019;111(1):1-2 View Article PubMed/NCBI
  12. Zhang JJ, Jia JP, Shao Q, Wang YK. Diabetes mellitus and risk of pancreatic cancer in China: A meta-analysis based on 26 case-control studies. Prim Care Diabetes 2019;13(3):276-282 View Article PubMed/NCBI
  13. Wolpin BM, Bao Y, Qian ZR, Wu C, Kraft P, Ogino S, et al. Hyperglycemia, insulin resistance, impaired pancreatic β-cell function, and risk of pancreatic cancer. J Natl Cancer Inst 2013;105(14):1027-1035 View Article PubMed/NCBI
  14. Keum N, Ha KH, Bao Y, Chung MJ, Kim HC, Giovannucci EL. Long-term patterns of fasting blood glucose levels and pancreatic cancer incidence. Cancer Causes Control 2018;29(1):135-142 View Article PubMed/NCBI
  15. Liao WC, Tu YK, Wu MS, Lin JT, Wang HP, Chien KL. Blood glucose concentration and risk of pancreatic cancer: systematic review and dose-response meta-analysis. BMJ 2015;350:g7371 View Article PubMed/NCBI
  16. Sato K, Hikita H, Myojin Y, Fukumoto K, Murai K, Sakane S, et al. Hyperglycemia enhances pancreatic cancer progression accompanied by elevations in phosphorylated STAT3 and MYC levels. PLoS One 2020;15(7):e0235573 View Article PubMed/NCBI
  17. Chen H, Zhao J, Jiang N, Wang Z, Liu C. Hyperglycemia Promotes Pancreatic Cancer Initiation and Progression by Activating the Wnt/β-Catenin Signaling Pathway. Anticancer Agents Med Chem 2021;21(18):2592-2602 View Article PubMed/NCBI
  18. Li W, Liu H, Qian W, Cheng L, Yan B, Han L, et al. Hyperglycemia aggravates microenvironment hypoxia and promotes the metastatic ability of pancreatic cancer. Comput Struct Biotechnol J 2018;16:479-487 View Article PubMed/NCBI
  19. Li W, Zhang L, Chen X, Jiang Z, Zong L, Ma Q. Hyperglycemia Promotes the Epithelial-Mesenchymal Transition of Pancreatic Cancer via Hydrogen Peroxide. Oxid Med Cell Longev 2016;2016:5190314 View Article PubMed/NCBI
  20. Rahn S, Zimmermann V, Viol F, Knaack H, Stemmer K, Peters L, et al. Diabetes as risk factor for pancreatic cancer: Hyperglycemia promotes epithelial-mesenchymal-transition and stem cell properties in pancreatic ductal epithelial cells. Cancer Lett 2018;415:129-150 View Article PubMed/NCBI
  21. Janssen JAMJL. Hyperinsulinemia and Its Pivotal Role in Aging, Obesity, Type 2 Diabetes, Cardiovascular Disease and Cancer. Int J Mol Sci 2021;22(15):7797 View Article PubMed/NCBI
  22. Stolzenberg-Solomon RZ, Graubard BI, Chari S, Limburg P, Taylor PR, Virtamo J, et al. Insulin, glucose, insulin resistance, and pancreatic cancer in male smokers. JAMA 2005;294(22):2872-2878 View Article PubMed/NCBI
  23. Avgerinos KI, Spyrou N, Mantzoros CS, Dalamaga M. Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metabolism 2019;92:121-135 View Article PubMed/NCBI
  24. Pollak M. Insulin and insulin-like growth factor signalling in neoplasia. Nat Rev Cancer 2008;8(12):915-928 View Article PubMed/NCBI
  25. Yang J, Waldron RT, Su HY, Moro A, Chang HH, Eibl G, et al. Insulin promotes proliferation and fibrosing responses in activated pancreatic stellate cells. Am J Physiol Gastrointest Liver Physiol 2016;311(4):G675-G687 View Article PubMed/NCBI
  26. Malone JI, Hansen BC. Does obesity cause type 2 diabetes mellitus (T2DM)? Or is it the opposite?. Pediatr Diabetes 2019;20(1):5-9 View Article PubMed/NCBI
  27. Arslan AA, Helzlsouer KJ, Kooperberg C, Shu XO, Steplowski E, Bueno-de-Mesquita HB, et al. Anthropometric measures, body mass index, and pancreatic cancer: a pooled analysis from the Pancreatic Cancer Cohort Consortium (PanScan). Arch Intern Med 2010;170(9):791-802 View Article PubMed/NCBI
  28. Genkinger JM, Spiegelman D, Anderson KE, Bernstein L, van den Brandt PA, Calle EE, et al. A pooled analysis of 14 cohort studies of anthropometric factors and pancreatic cancer risk. Int J Cancer 2011;129(7):1708-1717 View Article PubMed/NCBI
  29. Jiao L, Berrington de Gonzalez A, Hartge P, Pfeiffer RM, Park Y, Freedman DM, et al. Body mass index, effect modifiers, and risk of pancreatic cancer: a pooled study of seven prospective cohorts. Cancer Causes Control 2010;21(8):1305-1314 View Article PubMed/NCBI
  30. Pang Y, Holmes MV, Kartsonaki C, Guo Y, Yang L, Bian Z, et al. Young adulthood and adulthood adiposity in relation to incidence of pancreatic cancer: a prospective study of 0.5 million Chinese adults and a meta-analysis. J Epidemiol Community Health 2017;71(11):1059-1067 View Article PubMed/NCBI
  31. Obradovic M, Sudar-Milovanovic E, Soskic S, Essack M, Arya S, Stewart AJ, et al. Leptin and Obesity: Role and Clinical Implication. Front Endocrinol (Lausanne) 2021;12:585887 View Article PubMed/NCBI
  32. Stolzenberg-Solomon RZ, Newton CC, Silverman DT, Pollak M, Nogueira LM, Weinstein SJ, et al. Circulating Leptin and Risk of Pancreatic Cancer: A Pooled Analysis From 3 Cohorts. Am J Epidemiol 2015;182(3):187-197 View Article PubMed/NCBI
  33. Yuan QH, Zhang LL, Xu Y, Chen X, Zhang B, Li LX, et al. Circulating leptin and adiponectin levels in patients with pancreatic cancer. Chin Med J (Engl) 2021;134(17):2134-2136 View Article PubMed/NCBI
  34. Xu Y, Tan M, Tian X, Zhang J, Zhang J, Chen J, et al. Leptin receptor mediates the proliferation and glucose metabolism of pancreatic cancer cells via AKT pathway activation. Mol Med Rep 2020;21(2):945-952 View Article PubMed/NCBI
  35. Harbuzariu A, Rampoldi A, Daley-Brown DS, Candelaria P, Harmon TL, Lipsey CC, et al. Leptin-Notch signaling axis is involved in pancreatic cancer progression. Oncotarget 2017;8(5):7740-7752 View Article PubMed/NCBI
  36. Incio J, Liu H, Suboj P, Chin SM, Chen IX, Pinter M, et al. Obesity-Induced Inflammation and Desmoplasia Promote Pancreatic Cancer Progression and Resistance to Chemotherapy. Cancer Discov 2016;6(8):852-869 View Article PubMed/NCBI
  37. Loebig M, Klement J, Schmoller A, Betz S, Heuck N, Schweiger U, et al. Evidence for a relationship between VEGF and BMI independent of insulin sensitivity by glucose clamp procedure in a homogenous group healthy young men. PLoS One 2010;5(9):e12610 View Article PubMed/NCBI
  38. Shi S, Xu J, Zhang B, Ji S, Xu W, Liu J, et al. VEGF Promotes Glycolysis in Pancreatic Cancer via HIF1α Up-Regulation. Curr Mol Med 2016;16(4):394-403 View Article PubMed/NCBI
  39. Bang C, Mortensen MB, Lauridsen KG, Bruun JM. Trends in antidiabetic drug utilization and expenditure in Denmark: A 22-year nationwide study. Diabetes Obes Metab 2020;22(2):167-172 View Article PubMed/NCBI
  40. Sanchez-Rangel E, Inzucchi SE. Metformin: clinical use in type 2 diabetes. Diabetologia 2017;60(9):1586-1593 View Article PubMed/NCBI
  41. Bridges HR, Jones AJ, Pollak MN, Hirst J. Effects of metformin and other biguanides on oxidative phosphorylation in mitochondria. Biochem J 2014;462(3):475-487 View Article PubMed/NCBI
  42. Li D, Yeung SC, Hassan MM, Konopleva M, Abbruzzese JL. Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology 2009;137(2):482-488 View Article PubMed/NCBI
  43. Lu Y, García Rodríguez LA, Malgerud L, González-Pérez A, Martín-Pérez M, Lagergren J, et al. New-onset type 2 diabetes, elevated HbA1c, anti-diabetic medications, and risk of pancreatic cancer. Br J Cancer 2015;113(11):1607-1614 View Article PubMed/NCBI
  44. Zhang P, Li H, Tan X, Chen L, Wang S. Association of metformin use with cancer incidence and mortality: a meta-analysis. Cancer Epidemiol 2013;37(3):207-218 View Article PubMed/NCBI
  45. Ren D, Qin G, Zhao J, Sun Y, Zhang B, Li D, et al. Metformin activates the STING/IRF3/IFN-β pathway by inhibiting AKT phosphorylation in pancreatic cancer. Am J Cancer Res 2020;10(9):2851-2864 PubMed/NCBI
  46. Zhao HW, Zhou N, Jin F, Wang R, Zhao JQ. Metformin reduces pancreatic cancer cell proliferation and increases apoptosis through MTOR signaling pathway and its dose-effect relationship. Eur Rev Med Pharmacol Sci 2020;24(10):5336-5344 View Article PubMed/NCBI
  47. Smith NK, Hackett TA, Galli A, Flynn CR. GLP-1: Molecular mechanisms and outcomes of a complex signaling system. Neurochem Int 2019;128:94-105 View Article PubMed/NCBI
  48. Elashoff M, Matveyenko AV, Gier B, Elashoff R, Butler PC. Pancreatitis, pancreatic, and thyroid cancer with glucagon-like peptide-1-based therapies. Gastroenterology 2011;141(1):150-156 View Article PubMed/NCBI
  49. Körner M, Stöckli M, Waser B, Reubi JC. GLP-1 receptor expression in human tumors and human normal tissues: potential for in vivo targeting. J Nucl Med 2007;48(5):736-743 View Article PubMed/NCBI
  50. Knapen LM, van Dalem J, Keulemans YC, van Erp NP, Bazelier MT, De Bruin ML, et al. Use of incretin agents and risk of pancreatic cancer: a population-based cohort study. Diabetes Obes Metab 2016;18(3):258-265 View Article PubMed/NCBI
  51. Pinto LC, Falcetta MR, Rados DV, Leitão CB, Gross JL. Glucagon-like peptide-1 receptor agonists and pancreatic cancer: a meta-analysis with trial sequential analysis. Sci Rep 2019;9(1):2375 View Article PubMed/NCBI
  52. Mentlein R, Gallwitz B, Schmidt WE. 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 1993;214(3):829-835 View Article PubMed/NCBI
  53. Gokhale M, Buse JB, Gray CL, Pate V, Marquis MA, Stürmer T. Dipeptidyl-peptidase-4 inhibitors and pancreatic cancer: a cohort study. Diabetes Obes Metab 2014;16(12):1247-1256 View Article PubMed/NCBI
  54. Lee M, Sun J, Han M, Cho Y, Lee JY, Nam CM, et al. Nationwide Trends in Pancreatitis and Pancreatic Cancer Risk Among Patients With Newly Diagnosed Type 2 Diabetes Receiving Dipeptidyl Peptidase 4 Inhibitors. Diabetes Care 2019;42(11):2057-2064 View Article PubMed/NCBI
  55. Dicembrini I, Montereggi C, Nreu B, Mannucci E, Monami M. 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 2020;159:107981 View Article PubMed/NCBI
  56. Huang BZ, Pandol SJ, Jeon CY, Chari ST, Sugar CA, Chao CR, et al. New-Onset Diabetes, Longitudinal Trends in Metabolic Markers, and Risk of Pancreatic Cancer in a Heterogeneous Population. Clin Gastroenterol Hepatol 2020;18(8):1812-1821.e7 View Article PubMed/NCBI
  57. Permert J, Ihse I, Jorfeldt L, von Schenck H, Arnqvist HJ, Larsson J. Pancreatic cancer is associated with impaired glucose metabolism. Eur J Surg 1993;159(2):101-107 PubMed/NCBI
  58. Aggarwal G, Kamada P, Chari ST. Prevalence of diabetes mellitus in pancreatic cancer compared to common cancers. Pancreas 2013;42(2):198-201 View Article PubMed/NCBI
  59. Pannala R, Leirness JB, Bamlet WR, Basu A, Petersen GM, Chari ST. Prevalence and clinical profile of pancreatic cancer-associated diabetes mellitus. Gastroenterology 2008;134(4):981-987 View Article PubMed/NCBI
  60. Nagpal SJS, Kandlakunta H, Sharma A, Sannapaneni S, Velamala P, Majumder S, et al. 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 2018;113:26-S28 View Article
  61. Cersosimo E, Pisters PW, Pesola G, McDermott K, Bajorunas D, Brennan MF. Insulin secretion and action in patients with pancreatic cancer. Cancer 1991;67(2):486-493 View Article PubMed/NCBI
  62. Parajuli P, Nguyen TL, Prunier C, Razzaque MS, Xu K, Atfi A. Pancreatic cancer triggers diabetes through TGF-β-mediated selective depletion of islet β-cells. Life Sci Alliance 2020;3(6):e201900573 View Article PubMed/NCBI
  63. Javeed N, Sagar G, Dutta SK, Smyrk TC, Lau JS, Bhattacharya S, et al. Pancreatic Cancer-Derived Exosomes Cause Paraneoplastic β-cell Dysfunction. Clin Cancer Res 2015;21(7):1722-1733 View Article PubMed/NCBI
  64. Permert J, Ihse I, Jorfeldt L, von Schenck H, Arnquist HJ, Larsson J. Improved glucose metabolism after subtotal pancreatectomy for pancreatic cancer. Br J Surg 1993;80(8):1047-1050 View Article PubMed/NCBI
  65. Liu J, Knezetic JA, Strömmer L, Permert J, Larsson J, Adrian TE. The intracellular mechanism of insulin resistance in pancreatic cancer patients. J Clin Endocrinol Metab 2000;85(3):1232-1238 View Article PubMed/NCBI
  66. Wang L, Hu L, Zhou X, Xiong Z, Zhang C, Shehada HMA, et al. Exosomes secreted by human adipose mesenchymal stem cells promote scarless cutaneous repair by regulating extracellular matrix remodelling. Sci Rep 2017;7(1):13321 View Article PubMed/NCBI
  67. Sagar G, Sah RP, Javeed N, Dutta SK, Smyrk TC, Lau JS, et al. Pathogenesis of pancreatic cancer exosome-induced lipolysis in adipose tissue. Gut 2016;65(7):1165-1174 View Article PubMed/NCBI
  68. Morigny P, Houssier M, Mouisel E, Langin D. Adipocyte lipolysis and insulin resistance. Biochimie 2016;125:259-266 View Article PubMed/NCBI
  69. Setiawan VW, Stram DO, Porcel J, Chari ST, Maskarinec G, Le Marchand L, et al. Pancreatic Cancer Following Incident Diabetes in African Americans and Latinos: The Multiethnic Cohort. J Natl Cancer Inst 2019;111(1):27-33 View Article PubMed/NCBI
  70. Tempero MA, Malafa MP, Al-Hawary M, Asbun H, Bain A, Behrman SW, et al. Pancreatic Adenocarcinoma, Version 2.2017, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2017;15(8):1028-1061 View Article PubMed/NCBI
  71. Okusaka T, Nakamura M, Yoshida M, Kitano M, Uesaka K, Ito Y, et al. Clinical Practice Guidelines for Pancreatic Cancer 2019 From the Japan Pancreas Society: A Synopsis. Pancreas 2020;49(3):326-335 View Article PubMed/NCBI
  72. Chen FM, Ni JM, Zhang ZY, Zhang L, Li B, Jiang CJ. Presurgical Evaluation of Pancreatic Cancer: A Comprehensive Imaging Comparison of CT Versus MRI. AJR Am J Roentgenol 2016;206(3):526-535 View Article PubMed/NCBI
  73. Chu LC, Goggins MG, Fishman EK. Diagnosis and Detection of Pancreatic Cancer. Cancer J 2017;23(6):333-342 View Article PubMed/NCBI
  74. Gonoi W, Hayashi TY, Okuma H, Akahane M, Nakai Y, Mizuno S, et al. Development of pancreatic cancer is predictable well in advance using contrast-enhanced CT: a case-cohort study. Eur Radiol 2017;27(12):4941-4950 View Article PubMed/NCBI
  75. Ahn SS, Kim MJ, Choi JY, Hong HS, Chung YE, Lim JS. Indicative findings of pancreatic cancer in prediagnostic CT. Eur Radiol 2009;19(10):2448-2455 View Article PubMed/NCBI
  76. Hart PA, Kamada P, Rabe KG, Srinivasan S, Basu A, Aggarwal G, et al. Weight loss precedes cancer-specific symptoms in pancreatic cancer-associated diabetes mellitus. Pancreas 2011;40(5):768-772 View Article PubMed/NCBI
  77. Dong X, Lou YB, Mu YC, Kang MX, Wu YL. Predictive Factors for Differentiating Pancreatic Cancer-Associated Diabetes Mellitus from Common Type 2 Diabetes Mellitus for the Early Detection of Pancreatic Cancer. Digestion 2018;98(4):209-216 View Article PubMed/NCBI
  78. Sharma A, Kandlakunta H, Nagpal SJS, Feng Z, Hoos W, Petersen GM, Chari ST. Model to Determine Risk of Pancreatic Cancer in Patients With New-Onset Diabetes. Gastroenterology 2018;155(3):730-739.e3 View Article PubMed/NCBI
  79. Boursi B, Patalon T, Webb M, Margalit O, Beller T, Yang YX, et al. Validation of the Enriching New-Onset Diabetes for Pancreatic Cancer Model: A Retrospective Cohort Study Using Real-World Data. Pancreas 2022;51(2):196-199 View Article PubMed/NCBI
  80. Chen W, Butler RK, Lustigova E, Chari ST, Wu BU. Validation of the Enriching New-Onset Diabetes for Pancreatic Cancer Model in a Diverse and Integrated Healthcare Setting. Dig Dis Sci 2021;66(1):78-87 View Article PubMed/NCBI
  81. Michálková L, Horník Š, Sýkora J, Habartová L, Setnička V, Bunganič B. Early Detection of Pancreatic Cancer in Type 2 Diabetes Mellitus Patients Based on 1H NMR Metabolomics. J Proteome Res 2021;20(3):1744-1753 View Article PubMed/NCBI
  82. He X, Zhong J, Wang S, Zhou Y, Wang L, Zhang Y, et al. Serum metabolomics differentiating pancreatic cancer from new-onset diabetes. Oncotarget 2017;8(17):29116-29124 View Article PubMed/NCBI
  83. Tavano F, Fontana A, Mazza T, Gioffreda D, Biagini T, Palumbo O, et al. 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 2020;10:1567 View Article PubMed/NCBI
  84. Wang L, Scott FI, Boursi B, Reiss KA, Williams S, Glick H, et al. Cost-Effectiveness of a Risk-Tailored Pancreatic Cancer Early Detection Strategy Among Patients with New-Onset Diabetes. Clin Gastroenterol Hepatol 2022;20(9):1997-2004.e7 View Article PubMed/NCBI
  • Cancer Screening and Prevention
  • pISSN 2993-6314
  • eISSN 2835-3315
Back to Top

Diabetes Mellitus and Early Detection of Pancreatic Cancer

Yi-Bo Fu, Gang Yang, Jiang-Dong Qiu, Tai-Ping Zhang
  • Reset Zoom
  • Download TIFF