Skip to main content

Diabetes and further risk of cancer: a nationwide population-based study

Abstract

Background

Individuals with diabetes have a significantly higher risk of developing various forms of cancer, and the potential biological links between these two diseases are not completely understood.

Methods

This was a longitudinal retrospective nationwide cohort study, a study design that allows us to examine the natural course of cancer development over an extended period of time with a large sample size. Initially, 3,111,975 and 22,208,395 eligible patients aged ≥ 20 years with and without diabetes, respectively, were matched by age, sex, and the Charlson comorbidity index. Ultimately, 1,751,457 patients were selected from each group. Stratified populations for diabetic retinopathy (DR) (n = 380,822) and without DR (n = 380,822) as well as proliferative DR (PDR) (n = 141,150) and non-proliferative DR (NPDR) (n = 141,150) were analyzed in this study. The main outcome measure was the first-time diagnosis of cancer during the follow-up period.

Results

We observed a 20% higher risk of total cancer incidence [hazard ratios (HR), 1.20; p < 0.001] in the diabetes cohort compared to the non-diabetes cohort. The highest HR was observed for liver and pancreas cancers. Moderately increased risks were observed for oral, colon, gallbladder, reproductive (female), kidney, and brain cancer. Furthermore, there was a borderline significantly increased risk of stomach, skin, soft tissue, female breast, and urinary tract (except kidney) cancers and lymphatic and hematopoietic malignancies. The stratified analysis revealed that the total cancer incidence was significantly higher in the DR cohort compared to the non-DR cohort (HR, 1.31; p < 0.001), and there was a borderline increased risk in the PDR cohort compared to the NPDR cohort (HR, 1.13; p = 0.001).

Conclusions

This study provides large-scale, nationwide, population-based evidence that diabetes is independently associated with an increased risk of subsequent development of total cancer and cancer at specific sites. Notably, this risk may further increase when DR develops.

Peer Review reports

Background

The increasing global prevalence of diabetes and cancer has significant global health implications. Epidemiological evidence suggests that people with diabetes are at a significantly higher risk of various cancers, including hepatic, pancreatic, endometrial, colorectal, bladder, and breast cancers, whereas male patients with diabetes have a lower prevalence of prostate cancer than those without diabetes [1]. Clinical evidence has indicated a positive association between cancer and concomitant abnormalities in glucose metabolism. However, the potential biological links between these diseases are not completely understood.

Diabetic retinopathy (DR) is the most common microvascular complication in patients with diabetes and the leading global cause of vision loss in working middle-aged adults [2]. The pathological processes of DR include hyperglycemia and the polyol pathway, advanced glycation end-product formation, protein kinase C activation, hexosamine pathway flux, and poly (ADP-ribose) polymerase activation, which share similar pathogenic features with cancer initiation and progression [3,4,5,6,7]. Furthermore, oxidative stress, inflammation, vascular abnormalities, and angiogenesis are closely associated with pathological changes in the progression of DR, which are also involved in pathophysiological conditions for cancer development [8,9,10]. These findings suggest that DR and cancer may share similar pathogenic features and that improving diabetes control may further reduce the risk of cancer development.

Given the similarities in the pathogenesis and global impact of and mortality caused by both diseases, additional large-scale longitudinal studies that stratify diabetes into DR and non-DR subtypes and focus on the relationship between cancer and diabetes may help clarify the potential biological links between the two diseases. Therefore, this retrospective nationwide cohort study used data from the Taiwan National Health Insurance Research Database (NHIRD) to investigate the relationship between cancer and diabetes or DR. To achieve this goal, Cox proportional hazards regression analyses were performed using two cohorts with propensity-matching by age, sex, and comorbidities, which minimized confounding variables arising from the use of observational data.

Methods

Data sources

This nationwide, 1:1 matched, retrospective cohort study was conducted between January 2007 and December 2018. The database contains all registry files and details regarding original claims data obtained from the NHIRD, the Taiwan Cancer Registry (TCR), and the National Death Registry (NDR) of Taiwan. Taiwan launched a single-payer National Health Insurance program on March 1, 1995. As of 2014, 99.9% of Taiwan’s population were enrolled. The database of this program contains registration files and original claim data for reimbursement. Starting in 2002, Taiwan’s National Health Research Institutes established and continue to maintain NHIRD for public research purposes. The NHIRD, collecting data from almost all medical facilities in Taiwan, is a large, powerful data source for approved medical research [11]. Approximately 27.22 million individuals were included in this registry. All data in the database were encrypted to protect the privacy of individuals. The database provides detailed outpatient and inpatient claims data, including patient identification number, birth date, sex, treatment information, dates of admission and discharge, date of death, and diagnostic codes according to the International Classification of Diseases, 9th Revision, Clinical Modification (ICD-9-CM) codes before 2016 and ICD-10-CM codes (10th revision) since 2016. Each patient has a unique encrypted identifier that can be linked to the TCR and NDR. All datasets were interlinked using patient identification numbers.

Study cohort and patient selection

This study was approved by the Institutional Review Board of Tzu Chi Hospital, Hualien (TCHIRB109-108-C). For this retrospective study, informed consent was waived in accordance with the institutional guidelines. Among a total of 27,228,099 patients in the NHIRD between January 1, 2007, and December 31, 2018, those with unknown sex (n = 1,867,827) and age (n = 39,302) were excluded; the exclusion of these cases was based on the rationale that sex and age are two major variables for propensity matching in this study. Overall, 3,111,975 patients with primary diabetes (ICD-9-CM 250 or ICD-10-CM E10, E11) and 22,208,395 patients without diabetes were initially enrolled in the diabetes and non-diabetes groups, respectively, as the main study cohorts. Patients with secondary diabetes caused by factors that may also be independent risk factors for cancer (e.g., certain viral infections like hepatitis B virus or C virus) were excluded. Participants were further excluded from the diabetes group if they had cancer before the diagnosis of diabetes (n = 170,398) or if they were aged < 20 years (n = 16,651). To avoid confounding effects of patients’ characteristics and comorbidities, the resulting available patients with and without diabetes were further matched in a 1:1 ratio by age, sex, and Charlson index comorbidity (CCI). Finally, 2,068,075 patients from each group were included in this study. The index date was defined as the date of diabetes onset. After excluding the end date before the index date (n = 138,339) and follow-up of less than 1 year (n = 178,279), 1,751,457 paired study participants in each group were obtained. In addition to the main study cohort, stratified populations for diabetes with DR (ICD-9-CM 362.0X or ICD-10-CM E10.3X, E11.3X; n = 380,822) and without DR (n = 380,822) were obtained. Patients with DR were further stratified into proliferative DR (PDR, n = 141,150) and non-PDR (NPDR, n = 141,150) groups according to the presence (ICD-9-CM 362.02 or ICD-10-CM E10.35X, E11.35X) or absence (ICD-9-CM 362.01 or ICD-10-CM E10.32X, E10.33X, E10.34X, E11.32X, E11.33X, E11.34X) of retinal neovascularization. All stratifications were performed using similar exclusion criteria and matching procedures to those of the main cohorts. The detailed data flow of the study is shown in Fig. 1.

Fig. 1
figure 1

Study protocol and profile. A The selection flow chart and selected populations for the diabetes group and the control cohort. B The stratification flow chart and the stratified populations for diabetes with DR vs. diabetes without DR. C The stratification flow chart and the stratified populations for PDR vs. non-PDR. DM, diabetes mellitus; DR, diabetic retinopathy; PDR, proliferative diabetic retinopathy; y/o, years old

Outcomes measures

The study endpoint was the first incidence of cancer at any site during follow-up, identified according to TCR. Only the first occurrence of cancer was considered when calculating the cancer incidence. As identified by the corresponding ICD-10-CM codes of cancer, the sites were defined as follows: the lip, oral cavity, and pharynx (C00–C14); digestive organs (C15–C26), including the esophagus (C15), stomach (C16), colon (C18), liver (C22), gallbladder (C23), and pancreas (C25); respiratory and intrathoracic organs (C30–C39); the bone and articular cartilage (C40–C41); the skin (C43–C44); the soft tissue (C45–C49); the breast (C50); female genital organs (C51–C58); male genital organs (C60–C63); the urinary tract (C64–C68), including the kidney (C64) and others (C65–68); the eye (C69); the brain and other parts of the central nervous system (C70–C72); and lymphoid, hematopoietic, and related tissue (C81–C96). If a participant had lesions of different severity levels in both eyes, the grade assigned to them was that of the more severely involved eye. All outcomes were assessed during the follow-up period between the index date and December 31, 2018. Baseline comorbidities were identified using the ICD-9 codes, including CCI, hypertension (401. X–405.X, 437.2, 362.11), and hyperlipidemia (272.X).

Statistical analysis

Baseline characteristics, including age, sex, hypertension, hyperlipidemia, and CCI score were compared between two study groups using standardized mean difference (SMD). The incidence rate of cancer was calculated per 100,000 person-years, and the incidence ratio between two study groups was calculated. The Cox proportional hazards model was used to assess the adjusted hazard ratios (HR) and 99% confidence intervals (CI). The classification for the increased cancer risk was defined as follows: borderline, HR between 1.10 and 1.19; moderate, HR between 1.20 and 1.49; and high, HR ≥ 1.50. All models were adjusted for the characteristics listed in Table 1. Data analyses were performed using SAS version 9.4 for Windows (SAS Institute, Inc., Cary, NC, USA). All statistical tests were 2-sided, and a p-value < 0.01 or SMD > 0.1 was considered statistically significant.

Table 1 Characteristics of the study population

Results

The demographic characteristics and comorbidities of all cohorts are shown in Table 1.

Diabetes versus non-diabetes

During the 12-year follow-up period, the overall mean annual incidence of total cancer was higher in patients with diabetes than patients without (1309.74 per 100,000 person-years vs. 1130.13 per 100,000 person-years; incidence ratio, 1.17) (Table 2).

Table 2 Incidence of events (100,000 person-years)

In the multivariate survival analysis, patients with diabetes (HR, 1.20; 99% CI: 1.19–1.21; p < 0.001) and CCI (HR. 1.23; 99% CI: 1.22–1.24; p < 0.001) showed moderately increased risk of subsequent total cancer development; male sex (HR, 1.19; 99% CI: 1.18–1.20; p < 0.001) and hypertension (HR, 1.10; 99% CI: 1.09–1.11; p < 0.001) both had a borderline significantly higher incidence of subsequent total cancer, except for hyperlipidemia (HR, 0.86; 99% CI: 0.85–0.87; p < 0.001), which was independently associated with a decreased risk of subsequent total cancer.

Patients with diabetes had a significantly higher incidence of subsequent liver (HR, 1.69; 99% CI: 1.63–1.74; p < 0.001) and pancreas (HR, 1.87; 99% CI: 1.73–2.02; p < 0.001) cancers. We also observed a moderately increased risk of the oral cavity and pharynx (HR, 1.30; 99% CI: 1.24–1.36; p < 0.001), colon (HR, 1.25; 99% CI: 1.21–1.29; p < 0.001), gallbladder (HR, 1.34; 99% CI: 1.20–1.50; p < 0.001), female genital organs (HR, 1.30; 99% CI: 1.22–1.37; p < 0.001), kidney (HR, 1.44; 99% CI: 1.34–1.53; p < 0.001), and brain and other parts of central nervous system cancers (HR, 1.31; 99% CI: 1.17–1.48; p < 0.001). Furthermore, there were borderline increases in the risk of stomach (HR, 1.19; 99% CI: 1.13–1.26; p < 0.001), skin (HR,1.17; 99% CI: 1.09–1.25; p < 0.001), mesothelial and soft tissue (HR, 1.18; 99% CI: 1.02–1.37; p = 0.003), female breast (HR, 1.17; 99% CI: 1.11–1.22; p < 0.001), and urinary tract cancer (except kidney) (HR, 1.17; 99% CI: 1.10–1.25; p < 0.001) and lymphatic and hematopoietic malignancies (HR, 1.19; 99% CI, 1.13–1.26; p < 0.001). Conversely, patients with diabetes had a lower risk of subsequent esophagus cancer than those without diabetes (HR, 0.83; 99% CI: 0.76–0.92; p < 0.001) (Table 3).

Table 3 Predictors of total cancer and cancer in specific sites by multivariate analysis

Diabetes with DR versus diabetes without DR

During a follow-up period of 12 years, the overall mean annual incidence of total cancer was significantly higher in diabetes patients with DR than in diabetes patients without DR (1494.33 per 100,000 person-years vs. 1151.51 per 100,000 person-years; incidence ratio, 1.32) (Table 2).

In the multivariate survival analysis, diabetes with DR was independently associated with an increased risk of subsequent total cancer development (HR, 1.31; 99% CI: 1.28–1.34; p < 0.001). Males also had a moderately higher incidence of subsequent total cancer (HR, 1.25; 99% CI: 1.23–1.28; p < 0.001), whereas hypertension and hyperlipidemia did not. Regarding cancer sites, patients with DR showed a significantly increased risk of subsequent liver, mesothelial and soft tissue, and urinary tract cancers. We also observed a moderately increased risk of lip, oral cavity and pharynx, stomach, colon, pancreas, respiratory and intrathoracic organs, skin, female breast, and lymph and hematopoietic cancers. Similarly, patients with DR showed a trend toward an increased risk of subsequent esophageal, gallbladder, bone and articular cartilage, male genitalia, and eye, but the increase did attain a statistically significant difference in the multivariate analysis (Table 3).

Development of cancer in different stages of DR

The overall mean annual incidence of total cancer was higher in PDR patients than in NPDR patients (1464.64 per 100,000 person-years vs. 1329.56 per 100,000 person-years; incidence ratio, 1.13). Meanwhile, multivariate analysis showed an increased risk of subsequent total cancer development (HR, 1.13; 99% CI: 1.10–1.17; p < 0.001) in PDR patients than in NPDR patients.

Regarding the site of cancer, PDR patients showed a moderately increased risk of stomach, liver, female genital, and urinary tract cancer and a borderline significantly increased risk of colon cancer compared to NPDR patients. Similarly, patients with PDR showed a trend toward an increased risk of subsequent cancers of the lip, oral cavity and pharynx, gallbladder, respiratory and intrathoracic organs, bone and articular cartilage, skin, and lymph and hematopoietic cancer, but the increase did attain a statistically significant difference in the multivariate analysis. In contrast, PDR patients showed a decreased risk of male genital cancer compared with NPDR patients (Table 3).

Discussion

In this study, patients with diabetes were associated with a 20% higher risk of the subsequent development of total cancer incidence compared to patients without diabetes. Notably, we firstly observed that patients with DR had a 32% higher cancer incidence than those without. Furthermore, patients with PDR have a 13% higher risk of cancer in comparison to patients with NPDR. Our study encompasses a nationwide cohort, and the findings contribute to the body of evidence on the relationship between diabetes and cancer risk, confirming prior research on the association between diabetes and cancer risk [12,13,14,15,16,17,18]. DR is the most common microvascular complication in patients with diabetes. The propensity to develop DR is directly proportional to patient age, diabetes duration, and poor glycemic control [2]. A meta-analysis including nineteen studies that compared persons with high versus low levels of serum glucose (cut-off > 6.1 mmol/L) showed a positive association between serum glucose and risk of cancer with a pooled RR of 1.32 (95% CI: 1.20–1.45) [19]. Furthermore, sudden variations of blood glucose may play an important role in DR; therefore, glycemic variability (GV) may be useful in predicting complications of diabetes such as DR [20]. Our findings are aligned with the results of a recent prospective cohort study that included 15,286 participants, which indicated that high GV was associated with increased risk of all-site, breast, liver cancer, and cancer-specific death in diabetes [21].

The exact link between diabetes and cancer development remains unclear. Although the analysis of claims data is not designed with biological conclusions in mind, these results raise the hypothesis that DR and cancer may share some possible similar pathogenic features. DR patients have significantly higher levels of serum vascular endothelial growth factor (VEGF) and angiopoietin-2 (Ang-2) than non-DR individuals [22, 23]. Interestingly, both tumorigenesis and DR involve VEGF- and Ang-2-mediated pathways, and pharmaceutical agents targeting these factors have been effective in treating both diseases [24,25,26]. Moreover, VEGF and Ang-2 promote endothelial cell expression of intercellular adhesion molecule 1, leading to leukocyte activation and cytokine release, thereby causing further increases in VEGF expression and amplifying the inflammatory response [22, 27].

In addition to VEGF and Ang-2, several pathophysiological features have been observed in DR and cancer. First, pericyte loss is the earliest clinical sign of DR; the possible mechanisms linking pericyte apoptosis in DR include increased oxidative stress and nuclear factor-κB (NF-kB) activation [28]. Similarly, pericytes are also implicated as mediators of several processes associated with cancer pathophysiology, including tumor angiogenesis and metastasis [29]. Additionally, NF-κB is the most important molecule linking chronic inflammation to cancer; its activation occurs in cancer cells and tumor microenvironments in most solid cancers and hematopoietic malignancies [30]. Furthermore, platelet-derived growth factors (PDGFs) are growth factors that regulate cell growth and division. Increased PDGF levels, which are the main pathological characteristic of DR, especially the impairment of endothelial migration and proliferation by the inflammatory and angiogenic effects of PDGFs [31]. Intriguingly, PDGF signaling overactivity is associated with the development of numerous types of malignancies [32].

Systemic inflammation is an intrinsic response to diabetes and can promote or increase the risk of many different cancers, including liver, pancreatic, colon, breast, and other malignancies [33]. Several inflammatory mediators play roles in both DR and cancer. Our results revealed that patients with diabetes tended to have a greater cancer risk than their matched controls, and this trend intensified when DR developed. DR development refers to the breakdown of the blood-retinal barrier. Increasing evidence supports the role of proinflammatory cytokines, chemokines, and other inflammatory mediators in the pathogenesis of DR [34,35,36,37], leading to persistent low-grade inflammation. The inflammatory mediators released in DR may further trigger cancer pathogenesis, thereby increasing the likelihood of cancer.

The natural history of DR has been divided into two stages based on the proliferative status of the retinal vasculature: early NPDR and advanced PDR; our findings showed that patients with PDR had a higher overall mean annual cancer incidence than those with NPDR. PDR patients exhibit significantly elevated levels of serum interleukin (IL)-1β, tumor necrosis factor α, IL-6, VEGF, and matrix metalloproteinases (MMPs) than NPDR patients [38, 39]. Several studies have demonstrated a positive correlation between MMPs expression and the invasive and metastatic potential of malignant tumor [40]. Furthermore, transforming growth factor ꞵ1 (TGFꞵ1) is a pro-inflammatory cytokine implicated in the pathogenesis of DR, particularly in the late phase of the disease. TGFβ released and activated within the tumor microenvironment promotes cancer progression. Enhanced TGFβ signaling promotes cancer cell invasion, dissemination, and suppresses the sensitivity to anticancer drugs [41]. Our study findings are also supported by the fact that PDR patients have more severe pathology and inflammation than NPDR patients. In addition, cancer-associated retinopathy is a rare paraneoplastic disorder with loss of visual acuity caused by circulating antibodies formed against the retinal proteins in the presence of systemic cancer [42]. Since our patients had DR first and subsequently developed cancer, these two types of retinopathy have different etiological bases.

Except for female breast cancer, we found a significantly lower incidence of all-site cancer in patients with hyperlipidemia than in those without hyperlipidemia. Most epidemiological studies have reported inconsistent results regarding the association between hyperlipidemia and cancer incidence [43,44,45,46,47,48,49,50,51,52,53]. Cancer is known to have a protean physiological effect, which might include metabolic depression of blood cholesterol [54] or competing risks, and patients showing high total serum cholesterol (TSC) levels are more likely to be censored owing to cardiovascular mortality before they are diagnosed with cancer [55]. However, some studies found inverse associations with a time lag of ≥ 4 years between baseline cholesterol level and cancer diagnosis [48, 55, 56]; thus, the possibility of a direct effect of cholesterol on cancer still cannot be completely ruled out. In our study, hyperlipidemia was positively associated with breast cancer in women. Previous animal models have implied that increased plasma cholesterol levels might accelerate breast cancer development and exacerbate their aggressiveness [57]. Our results are also consistent with findings from a prospective large longitudinal cohort study in Korean adults [50], which showed that high TSC levels were positively associated with breast cancer risk in women.

This study has several strengths. First, the NHIRD contains all claims data recorded electronically, ensuring accuracy and avoiding recall biases. Second, data from the NHIRD provide population-based and representative claims information for insured people in Taiwan and reduce the likelihood of selection bias. Third, the large dataset size and longitudinal study design provided considerable statistical power, enabling the effective detection of differences between the cancer and control cohorts. This type of longitudinal cohort study has advantages over cross-sectional or case–control studies because the design allows the researchers to examine the natural course of cancer development over an extended period of time. However, some shortcomings of this type of design such as death as the competing risk for the event should be considered.

Study limitations

First, the NHIRD is an administrative database lacking laboratory results, such as HbA1c, and it cannot differentiate between the subtypes of hyperlipidemia (hypercholesterolemia, hypertriglyceridemia, or both in combined hyperlipidemia). Some important risk factors for cancer are not available in the NHIRD, such as education level, drinking and smoking habits, body mass index, physical activity, and family history of cancer, which might have confounded our results. Second, although the current retrospective cohort study was more efficient than a prospective study, some potential risk factors could not be obtained owing to the retrospective nature of the study. Third, early cancer might have been asymptomatic, and individuals with early cancer might have been undiagnosed, which could have led to misclassification bias. This non-differential misclassification could bias our results toward the null hypothesis and dilute the real difference in cancer incidence between the two cohorts. Fourth, early-stage diabetes might also have been underdiagnosed, which could have resulted in group misclassification. Fifth, this study lacked information on the patients’ use of cholesterol-lowering drugs. However, previous studies have not suggested a strong link between drug use and the incidence of cancer [58, 59]. Sixth, most of our study participants were ethnically Chinese from Taiwan, which might affect the generalizability of our results to other ethnic groups. Finally, this epidemiological study only supports the concept that there is a correlation between diabetes and tumorigenesis. Direct evidence to prove the causal relationship between these two diseases is not feasible to obtain in human studies considering the long follow-up period to identify the event (cancer). Perhaps, findings from relevant animal models may provide novel evidence to support this relationship. Analyzing serum biomarkers related to cancer development may help to provide some indirect evidence to support the concept. However, NHIRD does not contain laboratory data from patients, and clinical tests that analyze patients’ samples are not part of the scope of this study.

Conclusions

This nationwide population-based study provides evidence reinforcing an association between diabetes and the overall cancer incidence. To our knowledge, we first explored the association between DR and cancer. The result contributes novel insights by unveiling that patient with DR were at a significantly greater risk of subsequent cancer development at specific sites than their matched controls. These results raise the possibility that diabetes and DR may share common pathogenic features with cancer, and strict blood glucose control to prevent DR in patients with diabetes may further reduce cancer development. Further studies are required to better understand the underlying processes.

Availability of data and materials

Data are available from the National Health Insurance Research Database (NHIRD) published by the Taiwan National Health Insurance (NHI) Bureau. Due to the legal restrictions imposed by the government of Taiwan in relation to the “Personal Information Protection Act,” data cannot be made publicly available. Requests for data can be sent as formal proposals to the NHIRD (http://nhird.nhri.org.tw).

Abbreviations

Ang-2:

Angiopoietin-2

CCI:

Charlson index comorbidity

CI:

Confidence intervals

DR:

Diabetic retinopathy

GV:

Glycemic variability

HR:

Hazard ratios

ICD-9-CM:

International Classification of Diseases, 9th Revision, Clinical Modification

ICD-10-CM:

International Classification of Diseases, 10th Revision, Clinical Modification

IL:

Interleukin

MMPs:

Matrix metalloproteinases

NDR:

National Death Registry

NF-κB:

Nuclear factor-κB

NHIRD:

National Health Insurance Research Database

NPDR:

Non-proliferative diabetic retinopathy

PDGFs:

Platelet-derived growth factors

PDR:

Proliferative diabetic retinopathy

SMD:

Standardized mean difference

TCR:

Taiwan Cancer Registry

TGF β:

Transforming growth factor β

TSC:

Total serum cholesterol

VEGF:

Vascular endothelial growth factor

References

  1. Bonagiri PR, Shubrook JH. Review of associations between type 2 diabetes and cancer. Clin Diabetes. 2020;38(3):256–65.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Solomon SD, Chew E, Duh EJ, Sobrin L, Sun JK, VanderBeek BL, et al. Diabetic retinopathy: a position statement by the American Diabetes Association. Diabetes Care. 2017;40(3):412–8.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Schwab A, Siddiqui A, Vazakidou ME, Napoli F, Böttcher M, Menchicchi B, et al. Polyol pathway links glucose metabolism to the aggressiveness of cancer cells. Cancer Res. 2018;78(7):1604–18.

    Article  CAS  PubMed  Google Scholar 

  4. Schröter D, Höhn A. Role of advanced glycation end products in carcinogenesis and their therapeutic implications. Curr Pharm Des. 2018;24(44):5245–51.

    Article  PubMed  Google Scholar 

  5. Garg R, Benedetti LG, Abera MB, Wang H, Abba M, Kazanietz MG. Protein kinase C and cancer: what we know and what we do not. Oncogene. 2014;33(45):5225–37.

    Article  CAS  PubMed  Google Scholar 

  6. Akella NM, Ciraku L, Reginato MJ. Fueling the fire: emerging role of the hexosamine biosynthetic pathway in cancer. BMC Biol. 2019;17(1):52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Martin-Oliva D, Aguilar-Quesada R, O’valle F, Muñoz-Gámez JA, Martínez-Romero R, García Del Moral R, et al. Inhibition of poly(ADP-ribose) polymerase modulates tumor-related gene expression, including hypoxia-inducible factor-1 activation, during skin carcinogenesis. Cancer Res. 2006;66(11):5744–56.

    Article  CAS  PubMed  Google Scholar 

  8. Hayes JD, Dinkova-Kostova AT, Tew KD. Oxidative stress in cancer. Cancer Cell. 2020;38(2):167–97.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420(6917):860–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lugano R, Ramachandran M, Dimberg A. Tumor angiogenesis: causes, consequences, challenges and opportunities. Cell Mol Life Sci. 2020;77(9):1745–70.

    Article  CAS  PubMed  Google Scholar 

  11. Lin LY, Warren-Gash C, Smeeth L, Chen PC. Data resource profile: the National Health Insurance Research Database (NHIRD). Epidemiol Health. 2018;40:e2018062.

    Article  PubMed  PubMed Central  Google Scholar 

  12. Ling S, Brown K, Miksza JK, Howells L, Morrison A, Issa E, et al. 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. 2020;43(9):2313–22.

    Article  PubMed  Google Scholar 

  13. Inoue M, Iwasaki M, Otani T, Sasazuki S, Noda M, Tsugane S. Diabetes mellitus and the risk of cancer: results from a large-scale population-based cohort study in Japan. Arch Intern Med. 2006;166(17):1871–7.

    Article  PubMed  Google Scholar 

  14. Ballotari P, Vicentini M, Manicardi V, Gallo M, Chiatamone Ranieri S, Greci M, et al. Diabetes and risk of cancer incidence: results from a population-based cohort study in northern Italy. BMC Cancer. 2017;17(1):703.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Kuriki K, Hirose K, Tajima K. Diabetes and cancer risk for all and specific sites among Japanese men and women. Eur J Cancer Prev. 2007;16(1):83–9.

    Article  PubMed  Google Scholar 

  16. Bjornsdottir HH, Rawshani A, Rawshani A, Franzén S, Svensson AM, Sattar N, et al. A national observation study of cancer incidence and mortality risks in type 2 diabetes compared to the background population over time. Sci Rep. 2020;10(1):17376.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Harding JL, Shaw JE, Peeters A, Cartensen B, Magliano DJ. Cancer risk among people with type 1 and type 2 diabetes: disentangling true associations, detection bias, and reverse causation. Diabetes Care. 2015;38(2):264–70.

    Article  PubMed  Google Scholar 

  18. Zhang PH, Chen ZW, Lv D, Xu YY, Gu WL, Zhang XH, et al. Increased risk of cancer in patients with type 2 diabetes mellitus: a retrospective cohort study in China. BMC Public Health. 2012;12:567.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Crawley DJ, Holmberg L, Melvin JC, Loda M, Chowdhury S, Rudman SM, et al. Serum glucose and risk of cancer: a meta-analysis. BMC Cancer. 2014;14:985.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Chatziralli IP. The role of glycemic control and variability in diabetic retinopathy. Diabetes Ther. 2018;9(1):431–4.

    Article  PubMed  Google Scholar 

  21. Mao D, Lau ESH, Wu H, Yang A, Shi M, Fan B, et al. Risk associations of long-term HbA1c variability and obesity on cancer events and cancer-specific death in 15,286 patients with diabetes - a prospective cohort study. Lancet Reg Health West Pac. 2021;18:100315.

    PubMed  PubMed Central  Google Scholar 

  22. Tang J, Kern TS. Inflammation in diabetic retinopathy. Prog Retin Eye Res. 2011;30(5):343–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Rasul S, Reiter MH, Ilhan A, Lampichler K, Wagner L, Kautzky-Willer A. Circulating angiopoietin-2 and soluble tie-2 in type 2 diabetes mellitus: a cross-sectional study. Cardiovasc Diabetol. 2011;10:55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Whitehead M, Osborne A, Widdowson PS, Yu-Wai-Man P, Martin KR. Angiopoietins in diabetic retinopathy: current understanding and therapeutic potential. J Diabetes Res. 2019;2019:5140521.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Hu B, Cheng SY. Angiopoietin-2: development of inhibitors for cancer therapy. Curr Oncol Rep. 2009;11(2):111–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Meadows KL, Hurwitz HI. Anti-VEGF therapies in the clinic. Cold Spring Harb Perspect Med. 2012;2(10):a006577.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Fiedler U, Reiss Y, Scharpfenecker M, Grunow V, Koidl S, Thurston G, et al. Angiopoietin-2 sensitizes endothelial cells to TNF-alpha and has a crucial role in the induction of inflammation. Nat Med. 2006;12(2):235–9.

    Article  CAS  PubMed  Google Scholar 

  28. Romeo G, Liu WH, Asnaghi V, Kern TS, Lorenzi M. Activation of nuclear factor- kappaB induced by diabetes and high glucose regulates a proapoptotic program in retinal pericytes. Diabetes. 2002;51(7):2241–8.

    Article  CAS  PubMed  Google Scholar 

  29. Raza A, Franklin MJ, Dudek AZ. Pericytes and vessel maturation during tumor angiogenesis and metastasis. Am J Hematol. 2010;85(8):593–8.

    Article  CAS  PubMed  Google Scholar 

  30. Taniguchi K, Karin M. NF-κB, inflammation, immunity and cancer: coming of age. Nat Rev Immunol. 2018;18(5):309–24.

    Article  CAS  PubMed  Google Scholar 

  31. Shen S, Wang F, Fernandez A, Hu W. Role of platelet-derived growth factor in type II diabetes mellitus and its complications. Diab Vasc Dis Res. 2020;17(7):1479164120942119.

    PubMed  PubMed Central  Google Scholar 

  32. Heldin CH. Targeting the PDGF signaling pathway in tumor treatment. Cell Commun Signal. 2013;11:97.

    Article  PubMed  PubMed Central  Google Scholar 

  33. Roberts EW, Ruhland MK, Cai E, Mujal AM, Marchuk K, Beppler C, et al. Tumors exploit dedicated intracellular vesicles to program T cell responses. bioRxiv. 2019:691873. https://doi.org/10.1101/691873.

  34. Doganay S, Evereklioglu C, Er H, Türköz Y, Sevinç A, Mehmet N, et al. Comparison of serum NO, TNF-alpha, IL-1beta, sIL-2R, IL-6 and IL-8 levels with grades of retinopathy in patients with diabetes mellitus. Eye (Lond). 2002;16(2):163–70.

    Article  CAS  PubMed  Google Scholar 

  35. Wang W, Lo ACY. Diabetic retinopathy: pathophysiology and treatments. Int J Mol Sci. 2018;19(6):1816.

    Article  PubMed  PubMed Central  Google Scholar 

  36. Piemonti L, Calori G, Lattuada G, Mercalli A, Ragogna F, Garancini MP, et al. Association between plasma monocyte chemoattractant protein-1 concentration and cardiovascular disease mortality in middle-aged diabetic and nondiabetic individuals. Diabetes Care. 2009;32(11):2105–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Clausen P, Jacobsen P, Rossing K, Jensen JS, Parving HH, Feldt-Rasmussen B. Plasma concentrations of VCAM-1 and ICAM-1 are elevated in patients with type 1 diabetes mellitus with microalbuminuria and overt nephropathy. Diabet Med. 2000;17(9):644–9.

    Article  CAS  PubMed  Google Scholar 

  38. Abu El-Asrar AM, Mohammad G, Nawaz MI, Siddiquei MM, Van den Eynde K, Mousa A, et al. Relationship between vitreous levels of matrix metalloproteinases and vascular endothelial growth factor in proliferative diabetic retinopathy. PLoS One. 2013;8(12):e85857.

    Article  PubMed  PubMed Central  Google Scholar 

  39. Koleva-Georgieva DN, Sivkova NP, Terzieva D. Serum inflammatory cytokines IL-1beta, IL-6, TNF-alpha and VEGF have influence on the development of diabetic retinopathy. Folia Med (Plovdiv). 2011;53(2):44–50.

    PubMed  Google Scholar 

  40. Reunanen N, Kähäri VM. Matrix Metalloproteinases in Cancer Cell Invasion. In: Madame Curie Bioscience Database [Internet]. Austin: Landes Bioscience; 2000-2013. https://www.ncbi.nlm.nih.gov/books/NBK6598/.

  41. Derynck R, Turley SJ, Akhurst RJ. TGFβ biology in cancer progression and immunotherapy. Nat Rev Clin Oncol. 2021;18(1):9–34.

    Article  PubMed  Google Scholar 

  42. Naramala S, Ahmad J, Adapa S, Gavini F, Konala VM. Case series of cancer- associated retinopathy (CAR). Cureus. 2019;11(6):e4872.

    PubMed  PubMed Central  Google Scholar 

  43. Iso H, Ikeda A, Inoue M, Sato S, Tsugane S, JPHC Study Group. Serum cholesterol levels in relation to the incidence of cancer: the JPHC study cohorts. Int J Cancer. 2009;125(11):2679–86.

    Article  CAS  PubMed  Google Scholar 

  44. Melvin JC, Holmberg L, Rohrmann S, Loda M, Van Hemelrijck M. Serum lipid profiles and cancer risk in the context of obesity: four meta-analyses. J Cancer Epidemiol. 2013;2013:823849.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Morris DL, Borhani NO, Fitzsimons E, Hardy RJ, Hawkins CM, Kraus JF, et al. Serum cholesterol and cancer in the hypertension detection and follow-up Program. Cancer. 1983;52(9):1754–9.

    Article  CAS  PubMed  Google Scholar 

  46. Kark JD, Smith AH, Hames CG. Serum retinol and the inverse relationship between serum cholesterol and cancer. Br Med J (Clin Res Ed). 1982;284(6310):152–4.

    Article  CAS  PubMed  Google Scholar 

  47. Williams RR, Sorlie PD, Feinleib M, McNamara PM, Kannel WB, Dawber TR. Cancer incidence by levels of cholesterol. JAMA. 1981;245(3):247–52.

    Article  CAS  PubMed  Google Scholar 

  48. Schatzkin A, Hoover RN, Taylor PR, Ziegler RG, Carter CL, Larson DB, et al. Serum cholesterol and cancer in the NHANES I epidemiologic followup study. National health and nutrition examination survey. Lancet. 1987;2(8554):298–301.

    Article  CAS  PubMed  Google Scholar 

  49. Strohmaier S, Edlinger M, Manjer J, Stocks T, Bjørge T, Borena W, et al. Total serum cholesterol and cancer incidence in the Metabolic syndrome and Cancer Project (Me-Can). PLoS ONE. 2013;8(1):e54242.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kitahara CM, Berrington de González A, Freedman ND, Huxley R, Mok Y, Jee SH, et al. Total cholesterol and cancer risk in a large prospective study in Korea. J Clin Oncol. 2011;29(12):1592–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Benn M, Tybjærg-Hansen A, Stender S, Frikke-Schmidt R, Nordestgaard BG. Low-density lipoprotein cholesterol and the risk of cancer: a mendelian randomization study. J Natl Cancer Inst. 2011;103(6):508–19.

    Article  CAS  PubMed  Google Scholar 

  52. Strasak AM, Pfeiffer RM, Brant LJ, Rapp K, Hilbe W, Oberaigner W, et al. Time-dependent association of total serum cholesterol and cancer incidence in a cohort of 172,210 men and women: a prospective 19-year follow-up study. Ann Oncol. 2009;20(6):1113–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Nago N, Ishikawa S, Goto T, Kayaba K. Low cholesterol is associated with mortality from stroke, heart disease, and cancer: the Jichi Medical School Cohort Study. J Epidemiol. 2011;21(1):67–74.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Vitols S, Gahrton G, Björkholm M, Peterson C. Hypocholesterolaemia in malignancy due to elevated low-density-lipoprotein-receptor activity in tumour cells: evidence from studies in patients with leukaemia. Lancet. 1985;2(8465):1150–4.

    Article  CAS  PubMed  Google Scholar 

  55. Schatzkin A, Hoover RN, Taylor PR, Ziegler RG, Carter CL, Albanes D, et al. Site-specific analysis of total serum cholesterol and incident cancer in the National health and nutrition examination survey i epidemiologic follow-up study. Cancer Res. 1988;48(2):452–8.

    CAS  PubMed  Google Scholar 

  56. Isles CG, Hole DJ, Gillis CR, Hawthorne VM, Lever AF. Plasma cholesterol, coronary heart disease, and cancer in the Renfrew and Paisley survey. BMJ. 1989;298(6678):920–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Llaverias G, Danilo C, Mercier I, Daumer K, Capozza F, Williams TM, et al. Role of cholesterol in the development and progression of breast cancer. Am J Pathol. 2011;178(1):402–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Boudreau DM, Yu O, Johnson J. Statin use and cancer risk: a comprehensive review. Expert Opin Drug Saf. 2010;9(4):603–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Dale KM, Coleman CI, Henyan NN, Kluger J, White CM. Statins and cancer risk: a meta-analysis. JAMA. 2006;295(1):74–80.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to thank Professor Yu Ru Kou, who provided critical suggestions for revising this manuscript.

Funding

This study was supported by a research grant from the Ministry of Health and Welfare, Taiwan (H109163), and Buddhist Tzu Chi General Hospital, Hualien, Taiwan (TCRD110-70).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: MSH. Methodology: WCC, TCH, MSH. Data collection: WCC, TCH, WLH, FLC, HRT. Statistical analysis: WCC, TCH. Visualization: WCC, TCH, WLH, FLC, HRT. Supervision: MSH. Writing—original draft: WCC, MSH. Writing—review, and editing: WCC, MSH. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Ming-Shan He.

Ethics declarations

Ethics approval and consent to participate

This study was approved by the Institutional Review Board of Tzu Chi Hospital, Hualien (TCHIRB109-108-C). For this retrospective study, informed consent was waived in accordance with the institutional guidelines.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chang, WC., Hsieh, TC., Hsu, WL. et al. Diabetes and further risk of cancer: a nationwide population-based study. BMC Med 22, 214 (2024). https://doi.org/10.1186/s12916-024-03430-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12916-024-03430-y

Keywords