Open access

Diabetes and Cancer

Written By

Subhashini Yaturu

Submitted: 14 May 2012 Published: 26 June 2013

DOI: 10.5772/56419

From the Edited Volume

Type 2 Diabetes

Edited by Kazuko Masuo

Chapter metrics overview

2,040 Chapter Downloads

View Full Metrics

1. Introduction

Diabetes and cancer are two common conditions. Many epidemiological studies suggest frequent co-occurrence of diabetes and cancer. Meta-analyses and the summary of recommendations from the American Diabetes Association (ADA) and American Cancer Society suggest association of cancer and diabetes including liver, pancreas, endometrial, colorectal, breast and bladder cancers.[1] Diabetes appears to protect against prostate cancer based on decreased incidence of prostate cancer in subjects with diabetes. Lung cancer appears not to be associated with diabetes, and data is inconclusive for renal cell cancer and lymphoma.[1] Most of the association data is on the relation of cancer to type 2 diabetes. The major concern is that type 2 diabetes is associated with three of the five leading causes of cancer mortality such as carcinoma of the colon [2], pancreas [3] and breast (postmenopausal) [4]. The excess risk for each cancer is ~30% (colon), ~50% (pancreas) and ~20% (breast). The majority of the epidemiological data on cancer incidence and mortality had been obtained in type 2 diabetic patients. A cohort study to examine cancer incidence among 29,187 patients in Sweden who were hospitalized for type 1 diabetes from 1965 through 1999, observed 355 incident cases of cancer and which corresponded to a 20% increase in overall cancer incidence among type 1 diabetes patients (RR:1.2; CI: 1.0 to 1.3) [5]. Patients with type 1 diabetes had elevated risks of cancers of the stomach (RR: 2.3; CI: 1.1 to 4.1), cervix (R: 1.6; CI: 1.1 to 2.2), and endometrium (RR: 2.7; CI: 1.4 to 4.7) [5]. Hyperinsulinemia most likely favors cancer in diabetic patients as insulin is a growth factor with pre-eminent metabolic as well as mitogenic effects. Insulin action in malignant cells is favored by mechanisms acting at both the receptor and post-receptor level. Obesity, hyperglycemia, and increased oxidative stress may also contribute to increased cancer risk in diabetes [6]. There are reports of concern of hypoglycemic therapies on cancer risk, especially with insulin analogue-Glargine. A growing body of evidence suggests that metformin potentially reduces the risk of cancer. Aspirin and non-aspirin non-steroidal anti-inflammatory drugs appear to reduce recurrence of adenomas and incidence of advanced adenomas in individuals with an increased risk of colorectal adenomas and colorectal cancer, and calcium may reduce recurrence of adenomas [7, 8]. In this chapter we will include epidemiological evidence of association of diabetes and cancer, possible mechanisms and the effect of hypoglycemic agents in relation to cancer.

Advertisement

2. Epidemiology of diabetes and cancer risk

The Centers for Disease Control and Prevention (CDC) reports that 25.8 million people (8.3% of the U.S. population) have diabetes. Among them, 18.8 million people have diagnosed diabetes and 7.0 million people have undiagnosed diabetes. http://www.cdc.gov/diabetes/pubs/estimates11.htm#1. Three hundred and forty six million people worldwide have diabetes. Type 2 diabetes comprises 90% of people with diabetes around the world, and is largely the result of excess body weight and physical inactivity. http://www.who.int/mediacentre/factsheets/fs312/en/index.html. The incidence of diabetes is increasing globally. The estimated incidence of 12.7 million new cancer cases in 2008 will rise to 21.4 million by 2030, with nearly two thirds of all cancer diagnoses occurring in low- and middle-income countries. http://www.who.int/nmh/publications/ncd_report_chapter1.pdf. A series of recent studies and meta-analyses confirm that the risk for several solid and hematologic malignancies (including liver, pancreas, colorectal, kidney, bladder, endometrial and breast cancers, and non-Hodgkin's lymphoma) is increased in patients with diabetes [6]. Here the discussion follows on each malignancy with increased frequency in diabetes.

2.1. Pancreatic cancer

Type 2 diabetes mellitus is considered to be the third modifiable risk factor for pancreatic cancer after cigarette smoking and obesity. Based on the meta-analysis of 36 case–control and cohort studies, Everhart and Wright reported that the age and sex adjusted odds ratio for the development of pancreatic cancer in people with diabetes was 1.8 (CI of 1.7–1.9) [9]. They also noted that increased frequency of pancreatic cancer occurs with long-standing diabetes, especially those with the duration of at least 5 years with a RR of 2.0 with CI of 1.2 to 3.2 [9]. Gallo et al [10] from Italy reported that 40.2% of patients with pancreatic cancer and diabetes were diagnosed concomitantly or 15.9% were diagnosed within two years prior to diagnosis of cancer. Based on the data, the authors concluded increased prevalence of diabetes is related to pancreatic cancer and the diabetes is caused by the tumor [10]. A causal relationship between diabetes and pancreatic cancer is also supported by findings from pre-diagnostic evaluations of glucose and insulin levels in prospective studies. Data show that up to 80% of patients with pancreatic cancer are either hyperglycemic or diabetic. Diabetes has been shown to improve after pancreatic-cancer resection, suggesting that diabetes is caused by the cancer [11]. Pannala et al suggest new-onset diabetes may indicate subclinical pancreatic cancer, and patients with new-onset diabetes may constitute a population in whom pancreatic cancer can be detected early [11]. A meta-analysis of three cohorts and six case-control studies revealed even a two-fold risk in type 1 diabetes patients [12]. A meta-analysis of 36 studies was carried out by Huxley and associates that included17 case-control and 19 cohort or nested case-control studies with information on 9220 individuals with pancreatic cancer [3]. They noted that individuals with recent diagnosis of diabetes (< 4 years) had a 50% greater risk of the malignancy compared with individuals who had diabetes for ≥ 5 years (OR 2.1 vs. 1.5; p = 0.005).

2.2. Colorectal cancer

Increasing evidence suggests that a history of diabetes mellitus (DM) may be associated with an increased risk of colorectal cancer (CRC). In 2005, meta-analyses of 15 studies (six case-control and nine cohort studies) in the USA and Europe, including 2 593 935 participants, Larsson and associates found that diabetes was associated with an increased risk of colorectal cancer, compared with no diabetes (RR: 1.30; CI: 1.20–1.40] [2]. These results were said to be consistent between case-control and cohort studies and across the United States and Europe. The association did not differ significantly by sex, or by cancer sub-site. Diabetes was positively associated with colorectal cancer mortality. Results from a meta analysis of 41 cohorts was reported to support that diabetes was associated with an increased incidence of CRC (RR:1.27;1.21-1.34) [13]. In a recent systematic review and meta-analysis, twenty-four studies including eight case-control and 16 cohort studies, with a total of 3,659,341 participants were included [14]. Meta-analysis of the 24 included studies indicated that diabetes was associated with an increased risk of colorectal cancer, compared with no diabetes (RR: 1.26; CI: 1.20-1.31), without heterogeneity between studies (p (heterogeneity) = 0.296). Sub-group analyses found that these results were consistent between case-control and cohort studies and among studies conducted in different areas. The association between diabetes and colorectal cancer incidence did not differ significantly by sex and sub-sites. Insulin therapy was also positively associated with risk of colorectal cancer (RR = 1.61; 1.18-1.35), with evidence of heterogeneity between studies (p (heterogeneity) = 0.014).

2.3. COX-2 and colon cancer

Although COX-2, the inducible isoform, is regularly expressed at low levels in colonic mucosa, its activity increases dramatically following mutation of the adenomatous polyposis coli (APC) gene, suggesting that β-catenin/T cell factor-mediated Wnt signaling activity may regulate COX-2 gene expression. In addition, hypoxic conditions and sodium butyrate exposure may also contribute to COX-2 gene transcription in human cancers [15]. Because of its role in carcinogenesis, apoptosis, and angiogenesis, it is an excellent target for developing new drugs with selectivity for prevention and/or treatment of human cancers [16].

2.4. Breast cancer

Meta-analyses of 20 studies (5 case-control and 15 cohort studies) by Larsson and associates found that women with diabetes had a statistically significant 20% increased risk of breast cancer (RR, 1.20; CI:1.12-1.28) compared with no diabetes [4]. The summary estimates were similar for case-control studies (RR, 1.18; CI: 1.05-1.32) and cohort studies (RR, 1.20; CI: 1.11-1.30). Meta-analysis of 5 cohort studies on diabetes and mortality from breast cancer yielded a summary RR of 1.24 and CI of 0.95-1.62 for women with versus without diabetes. Findings from this meta-analysis indicate that diabetes is associated with an increased risk of breast cancer [4]. In the Nurses' Health Study, a total of 87,143 postmenopausal women, aged 30 to 55 years and free of cancer, were followed up for up to 26 years (1976-2002) and evaluated for the incidence of invasive breast cancer with increase in weight of at least 25.0 kg or more since age 18 years. Eliassen and associates noted an increased risk of breast cancer (RR: 1.45; CI: 1.27-1.66; p <.001), with a stronger association among women who have never taken postmenopausal hormones (RR, 1.98; CI: 1.55-2.53). Data suggest that weight gain during adult life, specifically since menopause, increases the risk of breast cancer among postmenopausal women, whereas weight loss after menopause is associated with a decreased risk of breast cancer [17].

1. Non-modifiable risk factors:
a. Age
b. Sex
c. Ethnicity
2. Modifiable risk factors:
a. Overweight/obesity
b. Physical activity
c. Diet
3. Biological links:
a. Hyperglycemia
b. Insulin
c. IGF-1
d. Estrogen and androgen bioavailability
e. Cytokines

Table 1.

Risk factors common to both diabetes and cancer

Advertisement

3. Pathophysiology

3.1. Hyperinsulinemia and cancer

Hyperinsulinemia most likely favors cancer in diabetic patients as insulin is a growth factor with pre-eminent metabolic but also mitogenic effects, and its action in malignant cells is favored by mechanisms acting at both the receptor and post-receptor level. Obesity, hyperglycemia, and increased oxidative stress may also contribute to increased cancer risk in diabetes [6].

3.2. Insulin

Insulin resistance and hyperinsulinemia are important factors in the development of type 2 diabetes. Insulin is known to stimulate cell proliferation and injection of insulin in rats promoted carcinogen-induced colon cancer [18]. Insulin/insulin-like growth factor 1(IGF-1) receptors and G protein-coupled receptors (GPCR) signaling systems are implicated in autocrine-paracrine stimulation of a variety of malignancies, including ductal adenocarcinoma of the pancreas. Metformin, the most widely used drug in the treatment of type 2 diabetes, activates AMP kinase (AMPK), which negatively regulates mammalian target of rapamycin (mTOR) complex 1 (mTORC1) [19]. Metformin was shown to significantly decrease the growth of pancreatic cancer cells xenografted into the flank of nude mice by interrupting the G protein-coupled receptor (GPCR), insulin receptor signaling by down-regulating the mTOR pathway [20]. The GPCR and insulin receptor pathways are associated with increased DNA synthesis and pancreatic cancer cell growth. By negatively regulating GPCR and insulin receptor signally, and interrupting their cross talk, metformin is shown to decrease pancreatic cancer cell growth in mice. In a meta-analysis of epidemiological studies on markers of hyperinsulinemia and cancer, Pisani reported that subjects who develop colorectal and pancreatic cancers have increased pre-diagnostic blood levels of insulin and glucose [21]. High insulin levels have also been shown to be associated with risk of endometrial cancer independent of estradiol [22]. A link between breast cancer risk and hyperinsulinemia (measured by fasting C-peptide levels) has been shown mainly in postmenopausal breast cancer. Insulin levels were positively associated with endometrial carcinoma [HR: 2.33, CI: 1.13-4.82] among women not using hormone therapy [23].

3.3. Insulin resistance

The term insulin resistance denotes that action of insulin is impaired in peripheral target tissues that include skeletal muscle, liver, and adipose tissue. Recent literature supports the hypothesis that insulin resistance is a high risk for cancers. The molecular mechanisms for this association and the role in the neoplastic transformation process are being explored. Insulin is a major anabolic hormone that can stimulate cell proliferation. Adiposity induces adverse local and systemic effects that include adipocyte intracellular lipid accumulation, endoplasmic reticulum and mitochondrial stress, and insulin resistance, with associated changes in circulating adipokines, free fatty acids, and inflammatory mediators. Insulin resistance and associated hyperglycemia, hyperinsulinemia, and inflammation have been suggested to be the underlying mechanisms contributing to development of diabetes-associated pancreatic cancer. Hyperinsulinemia, insulin resistance and proinflammatory cytokines have been linked to neoplastic proliferation of various organ cells (Fig. 1).

Figure 1.

Insulin resistance and premenopausal breast cancer. Abbreviations: SHBG, sex hormone-binding globulin; IGF, insulin-like growth factor; GH, growth hormone; HDL, high-density lipoprotein cholesterol.

In a study of the Polyp Prevention Trial of insulin and fasting glucose and risk of recurrent colorectal adenomas, Flood et al. noted the association of increased risk of adenoma recurrence and risk for recurrence of advanced adenomas with increased insulin [24].

3.4. Insulin-like Growth Factor-1 (IGF-1) and cancer

The IGF (insulin-like growth factor) system is essential for physiological growth. The IGF complex includes IGF-1 and IGF-2, their corresponding receptors (IGFR-1 and IGFR-2), IGF binding proteins 1–6 (IGFBPs), insulin receptor substrate (IRS). The signaling pathway of IGF plays a critical role in cellular proliferation and inhibition of apoptosis. Though growth hormone is the primary stimulus for IGF-1 production in the liver and insulin can increase the IGF-1 production by up-regulating growth hormone receptors in the liver, hyperinsulinemia can also increase IGF-1 bioavailability by decreasing hepatic secretion of IGF-binding protein (IGFBP)-1 and −2 [25]. IGF-R, a tyrosine kinase receptor for IGF-I and IGF-II is said to play a role in malignant transformation, progression, protection from apoptosis, and metastasis as documented in cell culture, animal and human studies [26]. Since the expression of IGF-1 receptors occurs in several cancers, the effects of insulin on cancer cell proliferation in vivo may involve IGF-1 stimulation and indirectly stimulate cancers. The IGF signaling pathway is involved in cell proliferation and differentiation and inhibits apoptosis. Increased expression of IGF-1, IGF-2, IGF-1R, or combinations have been documented in various malignancies including glioblastomas, neuroblastomas, meningiomas, medulloblastomas, carcinomas of the breast, malignancies of the gastrointestinal tract, such as colorectal and pancreatic carcinomas, and ovarian cancer [27]. Higher IGF-1 levels were reported to be associated with increased colorectal adenoma risk (ORs = 1.58; 1.16-2.16),[28] and inversely associated with endometrial carcinoma (HR: 0.53; 0.31-0.90) [23].

3.5. Adiponectin and cancer

Adiponectin, which is also referred to as ACP30 (Acrp30), is secreted predominantly by white adipose tissue [29]. Circulating concentrations of adiponectin are reduced in obesity and type 2 diabetes [30-32]. Adiponectin is considered to have beneficial antineoplastic effects, which are believed to be due to anti-proliferative, anti-inflammatory effects, along with antagonizing insulin resistance [33]. Adiponectin has been found to be an important negative regulator of hematopoiesis and the immune system as adiponectin was shown to suppress the growth of myelomonocyte cell lines in vitro by inducing apoptosis in myelomonocytic progenitor cells (leukaemia lines) and modulating expression of apoptosis-related genes and down regulating Bcl-2 gene expression [34]. Epidemilogical data have also shown a link between low adiponectin levels and renal cell cancer [35, 36]; especially large tumor size [37, 38]. Adiponectin was inversely associated with non-Hodgkin lymphoma and acute myeloblastic leukaemia (OR: 0.56; 0.34-0.94), but not with acute lymphoblastic leukaemia of B or T cell [39]. In a number of epidemiological studies, adiponectin levels have been linked to breast cancer and are believed to inhibit breast cancer cell proliferation in vivo. This effect may be due to adiponectin-triggered cellular apoptosis in MDA-MB-231 breast cancer cells in the presence of 17β-estradiol. These findings may suggest that a cross-talk between adiponectin and estrogen receptor signaling exists in breast cancer cells and that adiponectin effects on the growth and apoptosis of breast cancer cells in vitro are dependent on the presence of 17β-estradiol [40]. Low serum adiponectin is associated with colon, prostate and breast cancer [41]. In a recent study, plasma adiponectin level was associated with decreased colorectal cancer risk [42].

Adiponectin and colorectal cancer: Adiponectin was shown to act on preneoplastic colon epithelial cells to regulate cell growth by inducing autocrine IL-6 production and trans-IL-6 signaling. In a prospective case control study, men with low plasma adiponectin levels were said to have a higher risk of colorectal cancer than men with higher levels [43]. Meta analysis of 13 studies in patients with colorectal cancer and adenoma, though there was significant heterogeneity among studies, noted that there was a negative dose response relationship between levels of adiponectin and the risk of colorectal neoplasm in men [44].

Adiponectin and Endometrial cancer: Circulating adiponectin concentrations are inversely correlated with the incidence of endometrial carcinoma in epidemiological studies. In a study that investigated the direct effects of adiponectin on two endometrial carcinoma cell lines, HEC-1-A and RL95–2, adiponectin treatment led to suppression of cell proliferation in both cell types, which was primarily believed to be due to the significant increase of cell populations at G1/G0 phase and secondary to the induction of apoptosis [45].

3.6. Obesity and cancer

Accumulating epidemiologic evidence shows that obesity is associated with an increased risk of several common adult cancers. The risk of diabetes increases linearly with BMI; the prevalence of diabetes increased from 2% in those with a BMI of 25 to 29.9 kg/m2, to 8% in those with a BMI of 30 to 34.9 kg/m2, and finally to 13% in those with a BMI greater than 35 kg/m2 [46]. Similarly, an association between obesity or an incremental increase in body mass index (BMI) and an increased cancer risk have been reported for colon cancer (men and women) and rectal cancer (men only),[47] colon cancer,[48] liver cancer,[49] gall bladder cancer,[50] multiple myeloma, non-Hodgkin's lymphoma,[51] pancreatic cancer,[52] leukemia,[53] ovarian cancer,[54] breast cancer,[55] and endometrial cancer [56, 57]. In a population based prospective study of more than 900,000 U.S. adults, the reported relative risk of cancers in overweight and obesity was 1.52 for men and 1.62 for women [18]. A study from the United Kingdom showed that increasing BMI was associated with an increased incidence of endometrial cancer (RR:2.89, CI: 2.62–3.18), adenocarcinoma of the esophagus (RR:2.38;CI: 1.59–3.56), kidney cancer (RR:1.53, CI: 1.27–1.84), leukemia (RR:1.50, CI:1.23–1.83), multiple myeloma (RR:1.31;CI: 1.04–1.65), pancreatic cancer (RR:1.24;CI: 1.03–1.48), non-Hodgkin's lymphoma (RR:1.17; CI: 1.03–1.34), ovarian cancer (RR: 1.14; CI: 1.03–1.27), all cancers combined (RR:1.12; CI:1.09–1.14), breast cancer in postmenopausal women (RR: 1.40; CI: 1.31–1.49), and colorectal cancer in premenopausal women (RR:1.61; CI: 1.05–2.48) [58]. It is not surprising to note that increased adiposity may have a negative effect on treatment outcome and ultimate survival, because obesity has been found to be a negative prognostic factor for breast cancer[59] and colon cancer [60, 61].

3.7. Cyclooxygenase and cancers

Cyclooxygenase-2 (COX-2) over expression has been found in several types of human cancers, such as colon, breast, prostate, and pancreas, and appears to control many cellular processes. The contribution of COX-2 to carcinogenesis and the malignant phenotype of tumor cells have been thought to be related to its abilities to: (1) increase production of prostaglandins, (2) convert procarcinogens to carcinogens, (3) inhibit apoptosis, (4) promote angiogenesis, (5) modulate inflammation and immune function, and (6) increase tumor cell invasiveness [62].

3.8. Proinflammatory cytokines

Adipocytes secrete a number of proinflammatory cytokines. These cytokines are known to promote insulin resistance and increase circulating TG, features of the metabolic syndrome. Several cytokines, reactive oxygen species (ROS), and mediators of the inflammatory pathway, such as activation of nuclear factor-κB (NF-κB) and COX-2, lead to an increase in cell proliferation, survival, and inhibition of the proapoptotic pathway, ultimately resulting in tumor angiogenesis, invasion, and metastasis [16]. Proinflammatory cytokines implicated in carcinogenesis include IL-1, IL-6, IL-15, colony-stimulating factors, TNF-α, and the macrophage migration inhibitory factor.

Macrophage inhibitory cytokine-1 (MIC-1), also known as prostate-derived factor (PDF), is a molecule of the TGF-β super family and has been associated with the progression of various types of diseases including prostate cancer [63]. Collectively, cytokines are considered as a linker between inflammation and cancer. Cytokines, ROS, and mediators of the inflammatory pathway (e.g., NF-κB and COX-2) have been shown to increase cell cycling, cause loss of tumor suppressor function, and stimulate oncogene expression and lead to cancers. Positive feedback mechanisms between estrogens and inflammatory factors may exist in the breast and contribute to hormone-dependent breast cancer growth and progression [64]. Prostaglandin E synthase (PTGES) is also up-regulated by the proinflammatory cytokines TNF-α or IL-1β. Cytokines can enhance estrogen receptor (ER) activity and PTGES expression through the NF-κB pathway and cytokines can act to up-regulate aromatase expression as well as 17β-hydroxysteroid dehydrogenase activity in breast tissue, thereby leading to a further increase in E2 production [64].

Advertisement

4. Diabetes therapies and cancer

Diabetes is associated with increased risks of bladder, breast, colorectal, endometrial, kidney, liver and pancreatic cancer and a lower risk of prostate cancer. Diabetes treatments may influence the risk of cancer independently of their effect on glycemia. This may complicate the issues in investigation of the association between diabetes and cancer. Epidemiologic studies have suggested a protective role for metformin. On the other hand, Glargine, the most widely used long-acting insulin analogue, may confer a greater risk than other insulin preparations, particularly for breast cancer. In general, diabetes therapies that are said to be associated with increased risk of cancer include, use of insulin, sulfonyl urea and DPP4 inhibitors. Diabetes therapies that are shown benefit by decreasing the cancer risk include use of metformin and thiazolidinediones. Here we will discuss association of cancer risk with each of the diabetes therapeutic agents.

4.1. Thiazolidinedione (TZD) and cancer risk in type 2 diabetes

TZDs are reported to decrease in cancer both in clinical data series and in vitro studies. In addition pioglitazone, one of the TZDs, was shown to increase the risk of bladder cancer in those with the use for more than 24 months. Based on the randomized clinical trials of rosiglitazone with duration of >24 weeks that includes eighty trials enrolling 16,332 and 12,522 patients in the rosiglitazone and comparator groups, Monami and associates reported that the incidence of malignancies was significantly lower with the use of rosiglitazone than in control groups (RR: 0.23; CI: 0.19–0.26) vs. RR of 0.44(CI:0.34–0.58) cases/100 patient-years; p < 0.05) [65]. In a study using the diabetes registry of Hong Kong, Yang and associates reported that TZD usage was associated with 83% reduction in cancer risk in Chinese patients with type 2 diabetes in a dose–response manner [66]. Using the Taiwan National Health Insurance claims database, significantly lower risk of liver cancer incidence was found for any use of rosiglitazone or pioglitazone; use of rosiglitazone but not pioglitazone was associated with decreased incidence of colorectal cancer [67].

4.2. Pioglitazone and bladder cancer

Using the Kaiser Permanente Northern California diabetes registry with 193,099 patients who were ≥40 years of age between 1997 and 2002, excluding those with prior bladder cancer, 30,173 patients treated with pioglitazone were identified. In this cohort of patients with diabetes, short-term use of pioglitazone was not associated with an increased incidence of bladder cancer, but use for more than 2 years was weakly associated with increased risk [68].

Using the general practice research database in the United Kingdom, use of pioglitazone more than 24 months was reported to be associated with an increased risk of incident bladder cancer among people with type 2 diabetes [69. Using data from the French national health insurance information system, in a population based study, pioglitazone use of more than 24 months was reported significantly associated with increased risk of bladder cancer [70].

4.3. Metformin

Studies of patients with T2DM on metformin have demonstrated a lower risk of cancer [71-74]. In a recent study of the influence of treatment with metformin on survival after cancer diagnosis by Currie and associates, metformin was shown to be associated with survival benefit both in comparison with other treatments for diabetes and in comparison with a non-diabetic population [75]. An observational cohort study from the United Kingdom suggested that metformin use may be associated with a reduced risk of cancer (HR:0.63 (0.53-0.75) [72]. The study noted that the reduced risk was after adjusting for sex, age, BMI, A1C, smoking, and other drug use [72]. In a different database study from UK general practices, metformin use was reported to be associated with lower risk of cancer of the colon or pancreas, but did not affect the risk of breast or prostate cancer [71]. Metformin use was associated with survival benefit in comparison with other treatments for diabetes and also in comparison with a non-diabetic population [75]. Metformin has been associated with reduced risk of pancreatic cancer in diabetics and recognized as an antitumor agent with the potential to prevent and treat this cancer [76]. A retrospective cohort study to investigate the survival benefit of metformin in patients with diabetes and pancreatic malignancy, from the MD Anderson Cancer Center from 2000 to 2009, reported that metformin users have a significant survival benefit compared to non-users (the median survival 16.6 vs. 11.5 months; p = 0.0044) [77]. They also report a 33% decrease risk of death in patients who used metformin (HR: 0.67; p = 0.005) [77]. This implies that metformin may have some beneficial effects on slowing the progression of pancreatic malignancy. However, specific pathogenesis is unclear and would have to be further explored. In an interesting finding from a data base study from Hong Kong, nonusers of metformin with low HDL cholesterol had an adjusted hazard ratio of 5.75 (CI: 3.03-10.90) compared with HDL cholesterol ≥ 1.0 mmol/L plus use of metformin [78]. The reduction in cancer risk with the use of metformin in patients with type 2 diabetes is said to be dose related [74]. In a Canadian population-based cohort study, Bowker and associates noted that patients with type 2 diabetes exposed to sulfonylureas and exogenous insulin had a significantly increased risk of cancer-related mortality compared with patients exposed to metformin [79]. In addition they also noted that, the sulfonylurea cohort had greater cancer-related mortality compared with the metformin cohort after multivariate adjustment [79]. There are several in vitro and in vivo studies from cell lines and animal models support the benefit of metformin against cancer. There are several ongoing trials to examine the clinical outcomes.

4.3.1. Metformin and individual cancers

Long term use of metformin was shown to decrease risk of breast cancer in female patients with type 2 diabetes [80]. In a case-control study using the U.K.-based General Practice Research Database, metformin use was associated with an adjusted odds ratio of 0.44 (CI: 0.24-0.82) for developing breast cancer compared with no use of metformin [81]. In a similar study from the UK, long-term use of ≥ 40 prescriptions (>5 years) of metformin, based on 17 exposed case patients and 120 exposed control patients, was associated with an adjusted odds ratio of 0.44 (95% CI 0.24-0.82) for developing breast cancer compared with no use of metformin [80]. A meta-analysis of 17 case-control studies and 32 cohort studies of diabetes and hepatocellular carcinoma, metformin treatment was potentially protective [82]. In a meta- analysis of five studies comprising 108,161 patients with type 2 diabetes, metformin therapy appears to be associated with a significantly lower risk of colorectal cancer in patients with type 2 diabetes [82].

4.3.2. Mechanism of metformin in reducing cancer

It has been postulated that the effect of metformin on cancer development and progression may be a result of decreased levels of insulin [83] and insulin resistance. However, the possible anticancer effect of metformin is believed to be mediated by the inhibition of mitochondrial oxidative phosphorylation leading to activated AMPK pathway or independent of non-AMPK pathway. Human breast cancer cells treated with metformin demonstrate inhibited proliferation and colony formation and increased cell cycle arrest [84]. Studies have shown that metformin also has a direct effect on tumor cell proliferation [85]. As stated previously, metformin activates AMPK. The AMPK/mTOR axis is modulated by liver kinase B1 (LKB1). LKB1 is a tumor suppressor that activates AMPK, leading to mTOR inhibition, resulting in inhibited cell growth [85]. In vitro studies have shown that treatment with metformin inhibits cancer cell lines such as breast cancer breast cancer cells,[86] prostate cancer cell lines,[87, 88] glioma,[89] and fibro sarcoma cell lines [90].

4.4. Therapeutic consideration

4.4.1. Therapeutic considerations in general

Therapeutic considerations need to focus on reduction of the risk factors. Various therapeutic interventions for weight reduction and healthy life style have been linked to a reduced cancer risk in the general population. Therapeutic strategies to decrease chronic hyperinsulinemia and insulin resistance may offer a general approach to prevention of cancer. Metformin is the insulin sensitizer used primarily in the treatment of type 2 diabetes mellitus.

In a retrospective study of long term benefits of bariatric surgery, a significant decrease in mortality from cancer-related deaths in the bariatric surgery group compared both with all subjects and matched subjects with a decrease of 60% for cancer at mean follow-up of 7.1 years. Anticytokine vaccines, inhibitors of proinflammatory NF-κB and COX-2 pathways, thiazolidinediones, and antioxidants are potentially useful for the prevention or treatment of pancreatic cancer. Similarly epidemiologic studies have documented a 40–50% reduction in the incidence of colorectal cancer in individuals taking nonsteroidal anti-inflammatory drugs (NSAIDs). The long-term use of COX-2-selective inhibitors has, unfortunately, demonstrated cardiovascular toxicity, so their use in cancer prevention and therapy is currently questionable. However, there is evidence suggesting that further development of novel COX-2-selective agents is needed for the prevention and/or treatment of human cancers, especially pancreatic cancer. Targeting PGE2 signaling by EP receptor antagonists holds promise for the development of targeted therapy for the treatment of cancer [91]. PPARs also play a role in the regulation of cancer cell growth. Recent evidence suggests that PPAR modulators may have beneficial effects as chemopreventive agents [92].

Recent clinical studies with IGF-1R inhibitors have revealed several obstacles to successful use in cancer therapy. Strategies to inhibit IGF-1R signaling such as tyrosine kinase inhibitors also disrupt IR signaling, resulting in hyperglycemia and hyperinsulinemia. Several strategies being considered are based on biomarkers that could identify subpopulations most likely to be responsive to IGF targeting. The combination therapies with other targeted drugs could maximize the therapeutic effects of IGF inhibitors [93].

4.4.2. Chemoprevention of colorectal cancer

Of cancers affecting both men and women, colorectal cancer (cancer of the colon and rectum) is the second leading cancer killer in the United States and the incidence increases with age. The U.S. Preventive Services Task Force report on the effectiveness of aspirin (ASA), non-aspirin non steroidal anti-inflammatory drugs (non-ASA NSAIDs), and cyclooxygenase-2 inhibitors (COX-2 inhibitors) for the chemoprevention of colorectal cancer indicate that aspirin and non-ASA NSAIDs appear to be effective at reducing the incidence of CRAs and CRC [7]. The report also stated that more information is required to clarify the optimal dose, starting age, and duration of use of ASA since observational studies suggest that higher doses and prolonged use improve chemo-preventative efficacy. In a recent systematic review and meta-analysis for randomized controlled trials (RCTs) from United Kingdom, Cooper and associates identified 44 relevant RCTs and six ongoing studies [8]. They reported that there was a statistically significant 21% reduction in risk of adenoma recurrence (RR: 0.79; CI: 0.68 to 0.92) in an analysis of aspirin versus no aspirin in individuals with a history of adenomas or CRC. In the general population, a significant 26% reduction in CRC incidence was demonstrated in studies with a 23-year follow-up (RR: 0.74; CI: 0.57 to 0.97). In individuals with a history of adenomas there was a statistically significant 34% reduction in adenoma recurrence risk (RR: 0.66; CI: 0.60 to 0.72) and a statistically significant 55% reduction in advanced adenoma incidence (RR 0.45; CI: 0.35 to 0.58). No studies assessed the effect of non-aspirin NSAIDs in the general population. There was said to be no significant effect of folic acid versus placebo on adenoma recurrence (RR: 1.16; CI: 0.97 to 1.39) or advanced adenoma incidence in individuals with a history of adenomas. In the general population there was said to be no significant effect of folic acid on risk of colorectal cancer (RR: 1.13; CI: CI: 0.77 to 1.64), although studies were of relatively short duration. Calcium use by familial adenomatous polyposis (FAP) patients was reported to have no significant reduction in polyp number or disease progression. In individuals with a history of adenomas there was said to be a statistically significant 18% reduction in risk of adenoma recurrence (RR: 0.82; CI: 0.69 to 0.98) and a non-significant reduction in risk of advanced adenomas (RR: 0.77; CI: 0.50 to 1.17). Though these studies are not selective for subjects with diabetes, prevention of colorectal cancer in subjects with diabetes is important and relevant.

Advertisement

Acknowledgments

The author receives salary support from Veterans Health Administration. Appreciate Barbara Youngberg for her efforts in an excellent proof reading.

References

  1. 1. GiovannucciEet alDiabetes and cancer: a consensus report. Diabetes Care, 201016741685
  2. 2. LarssonS. CNOrsiniand AWolkDiabetes mellitus and risk of colorectal cancer: a meta-analysis. J Natl Cancer Inst, 200516791687
  3. 3. HuxleyRet alType-II diabetes and pancreatic cancer: a meta-analysis of 36 studies. Br J Cancer, 200520762083
  4. 4. LarssonS. CC. SMantzorosand AWolkDiabetes mellitus and risk of breast cancer: a meta-analysis. Int J Cancer, 2007856862
  5. 5. ZendehdelKet alCancer incidence in patients with type 1 diabetes mellitus: a population-based cohort study in Sweden. J Natl Cancer Inst, 200317971800
  6. 6. VigneriPet alDiabetes and cancer. Endocr Relat Cancer, 200911031123
  7. 7. RostomACDubeand GLewin2007
  8. 8. CooperKet alChemoprevention of colorectal cancer: systematic review and economic evaluation. Health Technol Assess, 20101206
  9. 9. EverhartJand DWrightDiabetes mellitus as a risk factor for pancreatic cancer. A meta-analysis. JAMA, 199516051609
  10. 10. GulloLRPezzilliand A. MMorselli-labateDiabetes and the risk of pancreatic cancer. N Engl J Med, 19948184
  11. 11. PannalaRet alNew-onset diabetes: a potential clue to the early diagnosis of pancreatic cancer. Lancet Oncol, 20098895
  12. 12. StevensR. JA. WRoddamand VBeralPancreatic cancer in type 1 and young-onset diabetes: systematic review and meta-analysis. Br J Cancer, 2007507509
  13. 13. JiangYet alDiabetes mellitus and incidence and mortality of colorectal cancer: a systematic review and meta-analysis of cohort studies. Eur J Epidemiol, 2011863876
  14. 14. DengLet alDiabetes mellitus and the incidence of colorectal cancer: an updated systematic review and meta-analysis. Dig Dis Sci, 201215761585
  15. 15. DempkeWet alCyclooxygenase-2: a novel target for cancer chemotherapy? J Cancer Res Clin Oncol, 2001411417
  16. 16. SarkarF. Het alBack to the future: COX-2 inhibitors for chemoprevention and cancer therapy. Mini Rev Med Chem, 2007599608
  17. 17. EliassenA. Het alAdult weight change and risk of postmenopausal breast cancer. JAMA, 2006193201
  18. 18. CalleE. Eet alOverweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med, 200316251638
  19. 19. RozengurtEJSinnett-smithand KKisfalviCrosstalk between insulin/insulin-like growth factor-1 receptors and G protein-coupled receptor signaling systems: a novel target for the antidiabetic drug metformin in pancreatic cancer. Clin Cancer Res, 201025052511
  20. 20. KisfalviKet alMetformin disrupts crosstalk between G protein-coupled receptor and insulin receptor signaling systems and inhibits pancreatic cancer growth. Cancer Res, 200965396545
  21. 21. PisaniPHyper-insulinaemia and cancer, meta-analyses of epidemiological studies. Arch Physiol Biochem, 20086370
  22. 22. LaceyJ. VJr., et alInsulin-like growth factors, insulin-like growth factor-binding proteins, and endometrial cancer in postmenopausal women: results from a U.S. case-control study. Cancer Epidemiol Biomarkers Prev, 2004607612
  23. 23. GunterM. Jet alA prospective evaluation of insulin and insulin-like growth factor-I as risk factors for endometrial cancer. Cancer Epidemiol Biomarkers Prev, 2008921929
  24. 24. FloodAet alElevated serum concentrations of insulin and glucose increase risk of recurrent colorectal adenomas. Gastroenterology, 200714231429
  25. 25. CoweySand R. WHardyThe metabolic syndrome: A high-risk state for cancer? Am J Pathol, 200615051522
  26. 26. FrascaFet alThe role of insulin receptors and IGF-I receptors in cancer and other diseases. Arch Physiol Biochem, 20082337
  27. 27. SamaniA. Aet alThe role of the IGF system in cancer growth and metastasis: overview and recent insights. Endocr Rev, 20072047
  28. 28. GaoYet alSerum IGF1, IGF2 and IGFBP3 and risk of advanced colorectal adenoma. Int J Cancer, 2012E105E113
  29. 29. SchererP. Eet alA novel serum protein similar to C1q, produced exclusively in adipocytes. J Biol Chem, 19952674626749
  30. 30. YaturuSet alPlasma and urine levels of resistin and adiponectin in chronic kidney disease. Cytokine, 200715
  31. 31. YaturuSet alResistin and adiponectin levels in subjects with coronary artery disease and type 2 diabetes. Cytokine, 2006219223
  32. 32. YaturuSJ. FBridgesand D.R. Subba Reddy, Decreased levels of plasma adiponectin in prediabetes, Type 2 diabetes and coronary artery disease. Med Sci Monit, 2006CR17CR20
  33. 33. BarbDet alAdiponectin in relation to malignancies: a review of existing basic research and clinical evidence. Am J Clin Nutr, 2007s858s866
  34. 34. YokotaTet alAdiponectin, a new member of the family of soluble defense collagens, negatively regulates the growth of myelomonocytic progenitors and the functions of macrophages. Blood, 200017231732
  35. 35. HoriguchiAet alDecreased serum adiponectin levels in patients with metastatic renal cell carcinoma. Jpn J Clin Oncol, 2008106111
  36. 36. SpyridopoulosT. Net alLow adiponectin levels are associated with renal cell carcinoma: a case-control study. Int J Cancer, 200715731578
  37. 37. KapoorSRe: Jehonathan H. Pinthus, Nir Kleinmann, Britton Tisdale, et al. Lower plasma adiponectin levels are associated with larger tumor size and metastasis in clear-cell carcinoma of the kidney. Eur Urol 2008;54:866-74. Eur Urol, 2009e52e53
  38. 38. PinthusJ. Het alLower plasma adiponectin levels are associated with larger tumor size and metastasis in clear-cell carcinoma of the kidney. Eur Urol, 2008866873
  39. 39. PetridouEet alAdiponectin in relation to childhood myeloblastic leukaemia. Br J Cancer, 2006156160
  40. 40. PfeilerG. Het alAdiponectin effects on human breast cancer cells are dependent on 17-beta estradiol. Oncol Rep, 2008787793
  41. 41. FentonJ. Iet alAdiponectin blocks multiple signaling cascades associated with leptin-induced cell proliferation in Apc Min/+ colon epithelial cells. Int J Cancer, 200824372445
  42. 42. TouvierMet alPre-diagnostic levels of adiponectin and soluble vascular cell adhesion molecule-1 are associated with colorectal cancer risk. World J Gastroenterol, 201228052812
  43. 43. WeiE. Ket alLow plasma adiponectin levels and risk of colorectal cancer in men: a prospective study. J Natl Cancer Inst, 200516881694
  44. 44. XuX. Tet alMeta-analysis: circulating adiponectin levels and risk of colorectal cancer and adenoma. J Dig Dis, 2011234244
  45. 45. CongLet alHuman adiponectin inhibits cell growth and induces apoptosis in human endometrial carcinoma cells, HEC-1-A and RL95 2. Endocr Relat Cancer, 2007713720
  46. 46. HarrisM. Iet alPrevalence of diabetes, impaired fasting glucose, and impaired glucose tolerance in U.S. adults. The Third National Health and Nutrition Examination Survey, 1988-1994. Diabetes Care, 1998518524
  47. 47. LarssonS. Cand AWolkObesity and colon and rectal cancer risk: a meta-analysis of prospective studies. Am J Clin Nutr, 2007556565
  48. 48. DaiZY. CXuand LNiuObesity and colorectal cancer risk: a meta-analysis of cohort studies. World J Gastroenterol, 200741994206
  49. 49. LarssonS. Cand AWolkOverweight, obesity and risk of liver cancer: a meta-analysis of cohort studies. Br J Cancer, 200710051008
  50. 50. LarssonS. Cand AWolkObesity and the risk of gallbladder cancer: a meta-analysis. Br J Cancer, 200714571461
  51. 51. WangYet alA prospective study of waist circumference and body mass index in relation to colorectal cancer incidence. Cancer Causes Control, 2008783792
  52. 52. LarssonS. CNOrsiniand AWolkBody mass index and pancreatic cancer risk: A meta-analysis of prospective studies. Int J Cancer, 200719931998
  53. 53. LarssonS. Cand AWolkOverweight and obesity and incidence of leukemia: a meta-analysis of cohort studies. Int J Cancer, 200814181421
  54. 54. OlsenC. Met alBody size and risk of epithelial ovarian and related cancers: a population-based case-control study. Int J Cancer, 2008450456
  55. 55. JensenAet alRisk of breast cancer and gynecologic cancers in a large population of nearly 50,000 infertile Danish women. Am J Epidemiol, 20084957
  56. 56. PatelA. Vet alThe role of body weight in the relationship between physical activity and endometrial cancer: results from a large cohort of US women. Int J Cancer, 200818771882
  57. 57. MccourtC. Ket alBody mass index: relationship to clinical, pathologic and features of microsatellite instability in endometrial cancer. Gynecol Oncol, 2007535539
  58. 58. ReevesG. Ket alCancer incidence and mortality in relation to body mass index in the Million Women Study: cohort study. BMJ, 20071134
  59. 59. CarmichaelA. RObesity and prognosis of breast cancer. Obes Rev, 2006333340
  60. 60. DignamJ. Jet alBody mass index and outcomes in patients who receive adjuvant chemotherapy for colon cancer. J Natl Cancer Inst, 200616471654
  61. 61. HaydonA. Met alEffect of physical activity and body size on survival after diagnosis with colorectal cancer. Gut, 20066267
  62. 62. XuX. CCOX-2 inhibitors in cancer treatment and prevention, a recent development. Anticancer Drugs, 2002127137
  63. 63. DubeySet alInflammation-associated regulation of the macrophage inhibitory cytokine (MIC-1) gene in prostate cancer. Oncol Lett, 201211661170
  64. 64. FrasorJet alSynergistic up-regulation of prostaglandin E synthase expression in breast cancer cells by 17beta-estradiol and proinflammatory cytokines. Endocrinology, 200862726279
  65. 65. MonamiMet alRosiglitazone and risk of cancer: a meta-analysis of randomized clinical trials. Diabetes Care, 200814551460
  66. 66. YangXet alUse of thiazolidinedione and cancer risk in Type 2 diabetes: the Hong Kong diabetes registry. Diabetes Res Clin Pract, 2012e13e17
  67. 67. ChangC. Het alAssociation of thiazolidinediones with liver cancer and colorectal cancer in type 2 diabetes mellitus. Hepatology, 201214621472
  68. 68. LewisJ. Det alRisk of bladder cancer among diabetic patients treated with pioglitazone: interim report of a longitudinal cohort study. Diabetes Care, 2011916922
  69. 69. AzoulayLet alThe use of pioglitazone and the risk of bladder cancer in people with type 2 diabetes: nested case-control study. BMJ, 2012e3645
  70. 70. NeumannAet alPioglitazone and risk of bladder cancer among diabetic patients in France: a population-based cohort study. Diabetologia, 201219531962
  71. 71. CurrieC. JC. DPooleand E. AGaleThe influence of glucose-lowering therapies on cancer risk in type 2 diabetes. Diabetologia, 200917661777
  72. 72. LibbyGet alNew users of metformin are at low risk of incident cancer: a cohort study among people with type 2 diabetes. Diabetes Care, 200916201625
  73. 73. JiralerspongSet alMetformin and pathologic complete responses to neoadjuvant chemotherapy in diabetic patients with breast cancer. J Clin Oncol, 200932973302
  74. 74. EvansJ. Met alMetformin and reduced risk of cancer in diabetic patients. BMJ, 200513041305
  75. 75. CurrieC. Jet alMortality after incident cancer in people with and without type 2 diabetes: impact of metformin on survival. Diabetes Care, 2012299304
  76. 76. LiDet alAntidiabetic therapies affect risk of pancreatic cancer. Gastroenterology, 2009482488
  77. 77. SadeghiNet alMetformin use is associated with better survival of diabetic patients with pancreatic cancer. Clin Cancer Res, 201229052912
  78. 78. YangXet alLow HDL cholesterol, metformin use, and cancer risk in type 2 diabetes: the Hong Kong Diabetes Registry. Diabetes Care, 2011375380
  79. 79. BowkerS. Let alIncreased cancer-related mortality for patients with type 2 diabetes who use sulfonylureas or insulin. Diabetes Care, 2006254258
  80. 80. BodmerMet alLong-term metformin use is associated with decreased risk of breast cancer. Diabetes Care, 201013041308
  81. 81. KeatingH. Jrd, Humanizing the intensive care unit. Del Med J, 1991129
  82. 82. WangPet alDiabetes mellitus and risk of hepatocellular carcinoma: a systematic review and meta-analysis. Diabetes Metab Res Rev, 2012109122
  83. 83. BersteinL. MMetformin, insulin, breast cancer and more. Future Oncol, 2009309312
  84. 84. AlimovaI. Net alMetformin inhibits breast cancer cell growth, colony formation and induces cell cycle arrest in vitro. Cell Cycle, 2009909915
  85. 85. Ben SahraI., et al., Metformin, independent of AMPK, induces mTOR inhibition and cell-cycle arrest through REDD1. Cancer Res, 201143664372
  86. 86. ZakikhaniMet alMetformin is an AMP kinase-dependent growth inhibitor for breast cancer cells. Cancer Res, 20061026910273
  87. 87. ChenDet alActivation of AMP-Activated Protein Kinase by 3,3’-Diindolylmethane (DIM) Is Associated with Human Prostate Cancer Cell Death In Vitro and In Vivo. PLoS One, 2012e47186
  88. 88. Ben SahraI., et al., The antidiabetic drug metformin exerts an antitumoral effect in vitro and in vivo through a decrease of cyclin D1 level. Oncogene, 200835763586
  89. 89. IsakovicAet alDual antiglioma action of metformin: cell cycle arrest and mitochondria-dependent apoptosis. Cell Mol Life Sci, 200712901302
  90. 90. HwangY. Pand H. GJeongMetformin blocks migration and invasion of tumour cells by inhibition of matrix metalloproteinase-9 activation through a calcium and protein kinase Calpha-dependent pathway: phorbol-12-myristate-13-acetate-induced/extracellular signal-regulated kinase/activator protein-1. Br J Pharmacol, 201011951211
  91. 91. WuW. Ket alCyclooxygenase-2 in tumorigenesis of gastrointestinal cancers: an update on the molecular mechanisms. Cancer Lett, 2010716
  92. 92. TachibanaKet alThe Role of PPARs in Cancer. PPAR Res, 2008. 2008102737
  93. 93. SeccarecciaEand PBrodtThe role of the insulin-like growth factor-I receptor in malignancy: An update. Growth Horm IGF Res, 2012

Written By

Subhashini Yaturu

Submitted: 14 May 2012 Published: 26 June 2013