Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jan 4.
Published in final edited form as: Diabetes Res Clin Pract. 2011 Oct 14;95(1):1–9. doi: 10.1016/j.diabres.2011.09.022

Metformin effects revisited

P Andújar-Plata a,*, X Pi-Sunyer b, B Laferrère b
PMCID: PMC5209790  NIHMSID: NIHMS835593  PMID: 22000494

Abstract

Metformin is a cornerstone in the treatment of type 2 diabetes. Although its mechanism of action is not well understood, there is new evidence about its possible role in cancer. A Pubmed search from 1990 to 2011 was done using the terms metformin, cancer, mechanism of action, diabetes treatment and prevention. We found more than one thousand articles and reviewed studies that had assessed the efficacy of metformin in treatment and prevention of type 2 diabetes and its mechanisms of actions, as well as articles on its antitumoral effects. We found that the United Kingdom Prospective Diabetes Study and the Diabetes Prevention Program have demonstrated the efficacy of metformin in terms of treatment and prevention of type 2 diabetes; metformin is safe, cost effective and remains the first line of diabetes therapy with diet and exercise. The mechanisms of action include a decrease of hepatic insulin resistance, change in bile acids metabolism, incretins release and decreased amyloid deposits. The AMP-activated protein kinase seems to be an important target for these effects. Epidemiological retrospective studies point out a possible association between metformin and decreased cancer risk, data supported by in vitro and animal studies. These data should trigger randomized controlled trials to prove or disprove this additional benefit of metformin.

Keywords: Metformin, Biguanides, Cancer, Type 2 diabetes

1. Introduction

The prevalence of type 2 diabetes (T2DM) is rapidly increasing worldwide [1], mostly due to obesity, so cheap and effective treatments are essential. Metformin is one of the most used oral glucose-lowering drugs for the treatment of T2DM [2]. The two main biguanides, metformin and phenformin were introduced for the first time in the late 1950s, but phenformin had to be withdrawn because of a strong association with lactic acidosis. Metformin can increase lactate oxidation but not change the release of lactate from muscle or plasma lactate concentration [3,4], so its association with lactic acidosis is very rare.

In the U.S.A., metformin became available in the 1990s. Previously, metformin was shown to lower blood glucose in randomized placebo-controlled studies [5], specifically by lowering fasting plasma glucose by decreasing hepatic glucose output and improving muscle sensitivity to insulin [3].

Before metformin came on the scene, sulfonylureas were the main oral drugs prescribed in T2DM with a well demonstrated efficacy, so at the time metformin was introduced there was a debate about what drug should be the first-line treatment in T2DM. In 1995, Campbell and Howlett did a meta-analysis of published prospective, controlled and randomized trials of metformin compared to sulfonylureas [6]. They found 11 acceptable studies involving 656 patients, and reported that both medications achieved a similar glycemic control, decreasing glycosylated hemoglobin (HbA1c) by 1.2%, but there was a weight loss benefit with metformin. Bailey and Turner, in a review in 1996 [7] also concluded that metformin could achieve a similar glycemic control than sulfonylureas, but metformin did not cause weight gain, hypoglycemia or increase insulin concentrations. Up to the present, the United Kingdom Prospective Diabetes Study (UKPDS) [8] is the most important study assessing metformin compared with other treatments; and the Diabetes Prevention Program (DPP) the most relevant trial in terms of diabetes prevention [9].

So actually, although there are many different medications to treat T2DM, metformin is still a cornerstone in the T2DM therapy. In fact, a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes, recommend metformin as the first step in T2DM treatment, if there is not a contraindication [10]. Despite the length of its use, continuing research on metformin’s mechanisms of action and particularly its potential role in cancer has restored its popularity.

2. United Kingdom Prospective Diabetes Study (UKPDS)

The UKPDS is a randomized, prospective and multicenter intervention trial which enrolled newly diagnosed T2DM patients between 1977 and 1991. The goal of the UKPDS was to study the effect of glycemic control on the prevention of complications, associated morbidity and mortality in non-insulin dependent diabetes, and compare diet alone with sulfonylureas, insulin and metformin [8].

Patients with fasting plasma glucose (FPG) at entry between 108 and 270 mg/dL, were randomized to sulfonylurea, insulin, metformin (only in overweight patients defined as >120% ideal body weight) or diet. At three years metformin achieved a similar reduction in FPG and glycated hemoglobin (HbA1c) as did sulfonylureas or insulin, but in addition it reduced fasting plasma insulin and did not induce weight gain [11]. Metformin was associated with fewer hypoglycemic episodes than sulfonylureas or insulin, but more than diet [12].

It was difficult to maintain an optimal glycemic control with monotherapy in the long term; in overweight patients on metformin only 39% could maintain FPG less than 140 mg/dL at three years and 34% had HbA1c < 7% at six years [13], similar to the other drug groups.

A 10-year analysis [14] showed that metformin produced a comparable glycemic control as did the other groups, without significant differences in weight but with lower fasting plasma insulin and lower hypoglycemic episodes than sulfonylureas and insulin.

2.1. Effect on complications

Drug therapy was associated with a lower risk of microvascular complications and reduced (not significantly) the risk of myocardial infarction. The metformin group had a lower risk of any diabetes-related endpoint (32%), diabetes related-death (42%), all-cause mortality (36%), myocardial infarction (39%) and all macrovascular diseases (30%) than did the diet group. The risk of retinopathy progression was also lower in the metformin group than in the conventional group, without differences in renal failure. Compared with the sulfonylurea and insulin groups, only the risk of diabetes-related endpoint and all-cause mortality was significantly lower in the metformin group [14].

On the other hand, in 1990 patients on sulfonylureas (overweight and non-overweight) who had FPG between 109.8 and 270 mg/dL were randomized to continue sulfonylureas alone or have metformin added to their regimen to know whether this addition had any advantage [14]. When metformin was added there was a decrease in FPG compared with the group on sulfonylureas alone; HbA1c values also decreased initially with addition of metformin but at three years were similar than in the sulfonylurea group. Surprisingly, it was found that the addition of metformin increased the risk of diabetes-related death and of any cause of death, maybe because the patients who were on sulfonylureas previously were older, more hyperglycemic and had been followed-up for a shorter time. However, a subsequent UKPDS publication [15] reported an increased life expectancy in overweight T2DM patients.

Another important issue is that metformin allows for cost-savings in overweight T2DM patients [15]. This is partly due to a lesser number of hospital days and lower cost of hospital stays in patients on metformin compared with the conventional treatment group [15].

So, UKPDS showed that metformin is as effective as sulfonylureas and insulin in glycemic control, but with no weight gain and a lower risk of hypoglycemia. However, the benefits of metformin were evaluated solely in overweight patients. An observational and retrospective study examining a database of patients on metformin or on sulfonylureas for at least for 6 months [16], concludes that metformin could be as effective as sulfonylureas in obese and non-obese patients after analyzing glycemic control and diabetes-related complications.

Ten years after the end of the UKPDS study, metformin in overweight participants was still associated with a statistically significant reduction in the risk of any diabetes-related endpoint (21%), diabetes-related death (30%), myocardial infarction (33%) and all-cause mortality (27%), compared with diet, although glycemic control was not significantly different; no differences in risk of microvascular disease were found [17].

As seen in UKPDS, metformin is a safe, cost-effective drug that achieves a good glucose control and ameliorates macrovascular risk in overweight patients with T2DM.

3. Diabetes Prevention Program (DPP)

Diabetes prevention is extremely important, particularly with the increased global prevalence of obesity, as more and more people are at risk of developing the disease and its associated complications, frequently present at diagnosis. DPP is the largest trial performed to assess strategies to prevent diabetes. It is a randomized trial involving more than 3000 people from 27 centers in the USA that are at high risk for diabetes: body mass index (BMI) > 24 or > 22 kg/m2 in Asians; FPG: 95–125 mg/dL; 2 h glucose after 75-g oral glucose load: 140–190 mg/dL. Participants were randomized to one of three groups: standard lifestyle recommendations plus placebo, standard lifestyle recommendations plus metformin and intensive lifestyle intervention (ILS). After an average follow up of 2.8 years, diabetes incidence was 58% lower in the ILS group and 31% lower in the metformin group compared with placebo [9], showing that treatment with metformin is a good strategy for diabetes prevention (in both genders and different ethnic groups), as well as being cost-effective [18]. Metformin was more effective in persons with higher BMI [19], higher FPG [20] and in women with previous gestational diabetes [21], and improved insulin levels and weight compared with placebo. But, although metformin reduced anthropometric measurements at one year, weight, BMI and waist circumference did not predict diabetes development in the metformin group [20].

Metformin use also predicted regression from impaired fasting glucose (IFG) and impaired glucose tolerance (IGT) to isolated IGT, but not to normal glucose tolerance [22], and improved insulin sensitivity at one year compared with placebo [23].

Metformin reduced the incidence of the metabolic syndrome in 17% of the patients, compared to placebo, an effect that was surprisingly not seen in women [24]. Although C-reactive protein and fibrinogen, possible cardiovascular risk factors, were lower in the metformin group compared to placebo [25], there were no significant differences in cardiovascular events among groups [26].

Long-term data from the DPP, obtained 10 years after randomization [27], showed that ILS and metformin were still associated with a lower diabetes incidence (34% and 18% respectively) compared with placebo. Therefore, metformin is also a good long-term strategy to prevent diabetes.

4. Safety

The most frequent adverse events of metformin are gastrointestinal symptoms, occurred in 22% of patients, but they usually are mild and transient [4]. Lactic acidosis is really very rare (4.3 cases per 100,000 patient-years) [28], probably due to the fact that metformin does not change plasma lactate levels [3,4,28]. Metformin can interfere with vitamin B12 levels, but anemia is not frequent [4]. Finally, in terms of hypoglycemia, metformin has demonstrated its safety [12].

5. Mechanisms

Metformin has been used clinically for over 50 years, but its molecular mechanism of action is not well understood. The glucose-lowering action is largely due to the improvement in hepatic insulin resistance leading to a reduction in hepatic glucose output, mainly as result of reduction in gluconeogenesis [3,29]. Metformin also increases glucose uptake in muscle, without extra lactate production, and raises insulin binding to insulin receptors (IR), while increasing the phosphorylation and tyrosine kinase activity of the IR [7]. In human hepatic cells, metformin increases insulin receptor activation independently of insulin, acting predominantly through insulin receptor substrate 2 [30].

A possible important target for metformin is AMP-activated protein kinase (AMPK), a cellular energy sensor activated under metabolic stress. The activation of AMPK inhibits glucose production by the liver, improvesinsulin sensitivity and glucose uptake by the muscle and induces fatty acid oxidation. Threonine 172 phosphorylation is necessary for this activation and the tumor suppressor gene LKB1 is the main responsible kinase. The major downstream target of AMPK is the mammalian target of rapamycin (mTOR), a kinase whose activity is very important in cellular growth processes and is inhibited by AMPK, leading to reduction of protein synthesis [31].

AMPK can be activated by exercise, hormones, cytokines and drugs [31]. Metformin activates AMPK in a dose and time dependent manner, which results in inhibition of hepatic glucose production, increased glucose uptake by muscle, inhibition of acetyl-CoA carboxylase and decreased fatty acid synthase expression in rat hepatocytes [32] and, possibly, increased glucose utilization in liver via an independent-insulin pathway [33]. The activation of AMPK by metformin seems to be independent of changes in AMP/ATP ratio [33,34], through inhibition of mitochondrial respiratory chain complex I and increase of reactive nitrogen species (RNS) [34,35]. However, another study in mice suggests that metformin can inhibit gluconeogenesis in an AMPK-independent pathway, through changes in AMP/ATP ratio [36].

Other hypotheses regarding the mechanisms of action of metformin are discussed below.

5.1. Incretins

The action of metformin on the incretin pathways has been proposed as another possible mechanism to explain its glucose-lowering effect. Maida et al. [37] observed that metformin acutely increased GLP-1 levels in mice without concomitant glucose administration, but didnot affect other gut peptides like peptide YY or GIP. Metformin also reduced the rate of gastric emptying and improved oral glucose tolerance, but this action was not mediated byGLP-1 [37]. No effect of metformin has been observed on DDP-4 activity [37]. Additionally, metformin induced upregulation of mRNA transcription of the GLP-1 receptor in islets of metformin-treated mice through an AMPK-independent pathway and involving PPARα [37].

5.2. Bile acids

Metformin alters bile acid metabolism, which is demonstrated by an increase of breath CO14 and fecal C14 in T2DM patients after the administration of C14-glycocholate [38]. This action seems to be due to a decrease of bile salt absorption in the ileum as seen in animal studies [39], by a reduction of active transport, independently of Na-K ATPase [40]. This could explain gastrointestinal effects of metformin and its hypolipidemic properties and it may be involved in glucose control because bile acids activate G-protein coupled receptor TGR5, which can stimulate GLP1 release by L-cells [41]. Furthermore, Patti et al. [42] found that patients who have undergone gastric bypass have greater bile acid levels compared with overweight/obese patients and they are correlated positively with postprandial peak of GLP1 and negatively with glucose levels, so this support the idea that bile acids may stimulate GLP1.

5.3. Islet amyloid

Metformin also could act on glucose control through changes in islet amyloid deposits. It is known that amyloid deposits lead to a decrease in β cell mass, which is usual in T2DM. In a study with transgenic mice (expressing peptide islet amyloid), metformin, although it did not prevent amyloid formation, reduced the prevalence and severity of islet amyloid. It was found that this effect could be related to changes in body fat mass and insulin secretion [43].

Further research is clearly needed to elucidate the link between the different hypotheses regarding the mechanism of action of metformin.

6. Metformin and cancer

There have been reports regarding the association between T2DM and a higher risk of cancer and a worse prognosis [4446]. This might be explained by hyperinsulinemia and insulin resistance, characteristics of T2DM, because insulin promotes growth and has mitogenic effects. Not too much is known about the role of anti-diabetic drugs in this relationship so there is an increasing interest about the anti-cancer effect of metformin, mostly after some population studies have suggested that metformin might reduce the risk of cancer in diabetic patients. Evans et al. [47] in a case–control study involving patients with newly diagnosed T2DM, found that patients on metformin had a 23% reduced risk of cancer compared to patients on sulfonylureas, even after adjusting for BMI. In another epidemiological study, involving new users of antidiabetic drugs, Bowker et al. [48] showed that the cancer-related mortality rate was significantly lower in the metformin group compared to sulfonylurea users.

Since these epidemiological reports, there have been many studies on the role of metformin in different types of cancer.

6.1. Population studies

It is known that hyperinsulinemia and T2DM are associated with a higher risk of breast cancer incidence and with a worse prognosis. Most of the clinical studies (Table 1) on the association of metformin with cancer are retrospective and therefore must be interpreted with caution because of confounding factors. Nevertheless, they suggest an interesting association that merits further investigation in prospective studies.

Table 1.

Summary of epidemiological studies on the association of metformin with cancer, including some possible confounding factors.

Reference Year Subjects Age (years) Duration of diabetes (years) Glycemic control (Hb A1c) Other therapies Outcomes
Libby et al. 2009 New users of metformin ≥35 7.9 ± 1 (metformin users)
7.2 ± 1.2 (comparators)
Sulfonylureas
Insulin
Cancer incidence in new users of metformin during 10 of years follow-up was 7.3% compared with 11.6% in patients on other treatments. This group also presented a lower mortality.
Li et al. 2009 Pancreatic cancer patients vs control subjects The higher proportion between 61 and 70 years: 39.2% (cases) vs 33.6% (controls) ≤2 years: 13.5% (cases) vs 3.4% control ≤7 in 57.1% of metformin ever users vs 62% of metformin never users Insulin secretagogues
Thiazolidinediones
Insulin
Metformin is associated with a significant reduction in risk of pancreatic cancer.
Jiralerspong et al. 2009 Invasive breast cancer patients receiving neoadjuvant chemotherapy Median: 49 Median: 7.3 (metformin group) vs 7.8 (nonmetformin group) Thiazolidinediones
Insulin
Metformin is an independent predictor of pathologic complete response (pCR) in diabetic patients with breast cancer receiving neoadjuvant chemotherapy. pCR in metformin group: 24%; in nonmetformin group: 8%: in nondiabetic group: 16%.
Wright et al. 2009 Prostate cancer patients vs control subjects ≥75% of patients: 55–74 Insulin
Sulfonylureas
Thiazolidinediones
Meglitinides
Metformin is associated with reduction in risk of prostate cancer in diabetic caucasian men.
Landman et al. 2010 T2DM patients 67.3 ± 10.6 (metformin group) vs 68 ± 12 (nonmetformin group) Median: 4.9 (metformin group) vs 7.1 (nonmetformin group) 7.7 ± 1.1 (metformin group) vs 7.4 ± 1.3 (nonmetformin group) Insulin
Sulfonylureas
Lower standardized mortality ratio for cancer mortality in patients on metformin, without changes after adjustment for confounder factors.
Bodmer et al. 2010 T2DM women 67.5 ± 10.5 ≥2: 63.3% (cases) vs 64.5% (controls) <6.5: 22.6% (cases) vs 20.7% (controls) Insulin
Sulfonylureas
Long-term use of metformin was associated with lower risk of breast cancer. The adjusted OR was 0.44 (95% CI 0.24–0.82) in metformin users compared with non-metformin users.
Hosono et al. 2010 Patients with aberrant crypt foci (no diabetic) 69.1 ± 5.95 (metformin group) vs 64.2 ± 9.07 (controls) 5.8 ± 1.01 (metformin group) vs 5.4 ± 0.56 (controls) Metformin reduces aberrant crypt foci growth and rectal epithelial cell proliferation in non-diabetic patients, without changes in insulin levels.
Monami et al. 2011 Cancer patients vs control subjects 68.9 ± 9.9 (cases) vs 68 ± 10 (controls) Median: 8.4 (cases) vs 10 (controls) Insulin
Insulin secretagogues
Metformin is associated with reduced incidence of cancer.
He et al. 2011 Diabetic prostate cancer patients 58.79%: ≤65 Thiazolidinediones
Insulin
Insulin secretagogues
Metformin is a significant predictor of improved survival.

Using a national cancer registry, Libby et al. found [49] that cancer incidence during a 10 year follow-up period was 7.3% in patients with diabetes on metformin compared to 11.6% in patients on other antidiabetic treatments. Patients on metformin had a higher median time to cancer and a lower mortality. In a different nested case–control study, metformin was also associated with reduced incidence of cancer in a cohort of more than 1000 patients [50]; after adjustments the odds ratio for metformin treatment was 0.46 (95% CI 0.25–0.85).

Metformin may also improve the prognosis of cancer. Indeed, in the Zwolle Outpatient Diabetes project Integrating Available Care (ZODIAC) study, the standardized cancer mortality ratio was 0.88 for metformin users and 1.625 for nonusers (median follow-up time: 10 years); this difference did not change after adjustment for confounding factors [51].

A small prospective pilot clinical trial [52] in 26 non-diabetic patients with aberrant crypt foci (ACF), a marker of colon carcinoma, showed that metformin treatment for 1 month reduced the number of ACF and rectal epithelial cell proliferation. This is remarkable because it points to a potential antiproliferative effect in non-diabetic patients.

This protective effect of metformin has not been observed in the ADOPT-RECORD analysis done by Home et al. [53]. Cancer occurrence was not an outcome of those prospective studies, but they analyzed the malignancies reports and concluded that metformin did not have a clear advantage over rosiglitazone and sulfonylureas. However, the number of reported cancers was so limited that the study is difficult to interpret.

On the other hand, there are studies evaluating the relation between metformin and specific cancers. Breast cancer is the one most studied in relation to metformin. Jiralerspong et al. [54] studied retrospectively the percentage of pathologic complete response (pCR) in patients with invasive breast cancer treated with neoadjuvant chemotherapy. The proportion of pCR was significantly higher in the metformin group compared to non-metformin treated patients with diabetes, without significant differences in chemotherapy doses or in insulin treatment between groups. However, there was no difference in survival. So, after adjustments, metformin use during neoadjuvant chemotherapy in invasive breast cancer was found to be an independent predictor of pCR. In another retrospective study, using data derived from the U.K.-based General Practice Research Database, long-term use of metformin (about 5 years) was associated with a lower risk of breast cancer [55], in patients with diabetes. The adjusted OR was 0.44 in metformin users compared with non-metformin users but with a small number of cases and controls being a limitation.

The use of metformin in patients with diabetes was also associated with a significant reduction in risk of pancreatic cancer (OR = 0.38; 95% CI, 0.21–0.67) in a hospital-based study, of almost 1000 patients with pancreatic adenocarcinoma [56].

Although prostate cancer is inversely associated with T2DM, hyperglycemia and hyperinsulinemia are risk factors of higher mortality. A retrospective study done by Wright and Stanford [57] suggests that Caucasian men with diabetes on metformin have a lower risk of prostate cancer. In another study performed in more than 200 patients with diabetes and prostate cancer [58], metformin was a significant predictor of improved survival.

6.2. In vitro studies

Metformin can inhibit breast cancer cell growth and this effect is greater when there is overexpression of the human epidermal growth factor receptor 2 (HER2). Metformin reduces HER2 oncoprotein expression in breast cancer cells, mostly acting through AMPK and downregulation of mTOR effector p70S6K1. However, metformin may act by other mechanisms because it still has some effect when an AMPK inhibitor is used [59]. Alimova et al. [60] showed that metformin reduces proliferation and colony formation in four breast cancer lines by inducing cell cycle arrest at G1. The drug also decreases erbB2 tyrosine kinase activity, but in this study, the mTOR effect was less important.

Metformin action on breast cancer has been studied in combination with chemotherapic agents. Hirsch et al. [61] observed that the combination of metformin and doxorubicin killed stem and non-stem cancer cells and achieved a greater reduction and remission of the tumor than either drug alone. This is of interest as stem cells may be linked to relapse after chemotherapy treatment due to self-renewal capacity.

Another possible action of metformin is on the epithelial–mesenchymal transition (EMT) status, which seems to have an important role in metastatic breast cancer cells. An in vitro study [62] has shown that metformin prevents the generation of a breast cancer stem cell phenotype by repressing transcriptionally the TGFβ axis.

Metformin also can inhibit the growth of cells of triple negative (TN) breast cancer, an aggressive subtype. The drug induces G1 cell cycle arrest, but also produces apoptosis and reduces levels of epidermal growth factor receptor, frequently overexpressed in TN breast cancer. Otherwise, this growth inhibition has been observed in breast cancer cells xenografted into non-diabetic mice [63], so it is possible that metformin could be useful in both diabetic and non-diabetic conditions.

Endometrial cancer is also associated with obesity and T2DM [64]. As in breast cancer, metformin [65] inhibits growth of endometrial cancer cells and causes G1 cell cycle arrest. This is largely due to AMPK activation and at high doses it can induce apoptosis. Furthermore, metformin decreases hTERT mRNA expression in endometrial cancer cells, which is a rate-limiting determinant of telomerase activity. Moreover, treatment of sera from polycystic ovary syndrome (PCOS) women with metformin seems to decrease significantly the in vitro invasion of endometrial cancer cells [66].

Metformin has also demonstrated its anti-proliferative effect in ovarian cancer cell lines, inhibiting their growth (including chemoresistant lines) and causing G1 cell cycle arrest [67]. In these types of cells, metformin activates AMPK through LKB and leads to decreased phosphorylation of mTOR and protein–lipid synthesis. However, it has a lower impact in more aggressive cancer cells and, more interestingly, there is no a significant effect over primary ovarian cells. In another study [68] metformin was observed to induce apoptosis in ovarian cancer cells (AMPK independent manner) and induce changes in Bcl-2 proteins, increasing the relation pro-apoptotic/anti-apoptotic, suggesting that several mechanisms are involved in metformin action on cancer.

Metformin acts also on pancreatic cancer cells, blocking the crosstalk between insulin and G protein-coupled receptors (GPCR) that leads to a major GPCR-induced signaling. Metformin reduces DNA synthesis and cell proliferation induced by insulin or GPCR agonist and this action is through AMPK. Moreover, it inhibits the growth of human pancreatic cancer cells xenografted into mice [69].

6.3. Animal studies

Mice with colon carcinoma on a high energy diet have greater cancer growth, insulin levels and fatty acid synthase (FASN) expression. FASN is involved in fatty acids synthesis, which are an important energy substrate for cancer cells. The effect of metformin was not seen in mice on a normal diet [70].

The diet effect was also observed in mice with lung LLC1 carcinoma. These mice on a high-energy diet have a greater cancer growth compared to mice on a normal diet, this effect is almost reversed by metformin only in mice on a high energy diet, but not in mice on a normal diet. In those mice metformin also reduces high C-peptide levels and improves insulin tolerance. Metformin activates AMPK in the tumor cells, but mice on the high-energy diet have a higher phosphorylation of insulin receptor, so it may be that metformin also acts by reducing insulin levels [71]. In mice with NNK-induced lung cancer [72], oral metformin prevents growth of tumor, with a mild effect over mTOR in the lung. However, intraperitoneal administration was more effective in preventing tumorigenesis and inhibiting mTOR and it occurred without lung-AMPK activation. One explanation would be again that metformin inhibits mTOR by decreasing insulin or IGF1 levels.

Metformin, in combination with different chemotherapeutic agents, can stop the tumor growth in mice with breast cancer and delay the relapse, mostly acting on stem cells [73], a mechanism seen in in vitro studies.

Besides the effect on tumor growth, it has been reported that metformin postpones tumor development and increases the mean life span of female spontaneously hypertensive rats when it is started in early life, but does not change tumor incidence [74].

Therefore, in vitro and in vivo studies point to an antiproliferative effect of metformin, with an interesting interaction with diet high in energy density. Theses in vitro and animal data support the epidemiological evidence that the use of metformin is associated with a lower risk of cancer.

7. Conclusion

Metformin is an oral antidiabetic drug that has been used for more than 50 years. It is cost effective and very safe clinically. Studies such as the UKPDS have clearly demonstrated its glucose-lowering efficacy, mostly in overweight patients. So, with increasing prevalence of obesity and T2DM, in addition to diet and exercise, metformin seems an appropriate first-step therapy. Moreover, it is effective in diabetes prevention in high risk populations although there are currently no guidelines for its use in this clinical setting. Its glucose-lowering effect is mostly due to its action on hepatic insulin sensitivity; its effect on bile acids, GLP-1 and amyloid deposits merit more research. There is strong epidemiological evidence, supported by in vitro and animal studies, that metformin may prevent cancer. Further clinical studies are needed to understand the mechanisms of this relationship, and whether cancer risk and/or status should be considered when prescribing antidiabetic drugs.

Footnotes

Conflict of interest

The authors declare that they have no conflict of interest.

References

  • 1.International Diabetes Federation. Diabetes atlas. 4. Brussels: International Diabetes Federation; 2009. [Google Scholar]
  • 2.Kirpichnikov D, McFarlane SI, Sowers JR. Metformin: an update. Ann Intern Med. 2002;137:25–33. doi: 10.7326/0003-4819-137-1-200207020-00009. [DOI] [PubMed] [Google Scholar]
  • 3.Stumvoll M, Nurjhan N, Perriello G, Dailey G, Gerich JE. Metabolic effects of metformin in non-insulin-dependent diabetes mellitus. N Engl J Med. 1995;333:550–4. doi: 10.1056/NEJM199508313330903. [DOI] [PubMed] [Google Scholar]
  • 4.DeFronzo RA, Goodman AM. Efficacy of metformin in patients with non-insulin-dependent diabetes mellitus. The Multicenter Metformin Study Group. N Engl J Med. 1995;333:541–9. doi: 10.1056/NEJM199508313330902. [DOI] [PubMed] [Google Scholar]
  • 5.The global challenge of diabetes. Lancet. 2008;371:1723. doi: 10.1016/S0140-6736(08)60733-3. [DOI] [PubMed] [Google Scholar]
  • 6.Campbell IW, Howlett HC. Worldwide experience of metformin as an effective glucose-lowering agent: a meta-analysis. Diabetes Metab Rev. 1995;11(Suppl 1):S57–62. doi: 10.1002/dmr.5610110509. [DOI] [PubMed] [Google Scholar]
  • 7.Bailey CJ, Turner RC. Metformin. N Engl J Med. 1996;334:574–9. doi: 10.1056/NEJM199602293340906. [DOI] [PubMed] [Google Scholar]
  • 8.UK Prospective Diabetes Study (UKPDS) VIII. Study design, progress and performance. Diabetologia. 1991;34:877–90. [PubMed] [Google Scholar]
  • 9.Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, Walker EA, et al. Reduction in the incidence of type 2 diabetes with lifestyle intervention or metformin. N Engl J Med. 2002;346:393–403. doi: 10.1056/NEJMoa012512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Nathan DM, Buse JB, Davidson MB, Ferrannini E, Holman RR, Sherwin R, et al. Medical management of hyperglycemia in type 2 diabetes: a consensus algorithm for the initiation and adjustment of therapy: a consensus statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care. 2009;32:193–203. doi: 10.2337/dc08-9025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.United Kingdom Prospective Diabetes Study (UKPDS) 13: Relative efficacy of randomly allocated diet, sulphonylurea, insulin, or metformin in patients with newly diagnosed non-insulin dependent diabetes followed for three years. BMJ. 1995;310:83–8. [PMC free article] [PubMed] [Google Scholar]
  • 12.Wright AD, Cull CA, Macleod KM, Holman RR. Hypoglycemia in Type 2 diabetic patients randomized to and maintained on monotherapy with diet, sulfonylurea, metformin, or insulin for 6 years from diagnosis: UKPDS73. J Diabetes Complications. 2006;20:395–401. doi: 10.1016/j.jdiacomp.2005.08.010. [DOI] [PubMed] [Google Scholar]
  • 13.Turner RC, Cull CA, Frighi V, Holman RR. Glycemic control with diet, sulfonylurea, metformin, or insulin in patients with type 2 diabetes mellitus: progressive requirement for multiple therapies (UKPDS 49). UK Prospective Diabetes Study (UKPDS) Group. JAMA. 1999;281:2005–12. doi: 10.1001/jama.281.21.2005. [DOI] [PubMed] [Google Scholar]
  • 14.Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34) UK Prospective Diabetes Study (UKPDS) Group. Lancet. 1998;352:854–65. [PubMed] [Google Scholar]
  • 15.Clarke P, Gray A, Adler A, Stevens R, Raikou M, Cull C, et al. Cost-effectiveness analysis of intensive blood-glucose control with metformin in overweight patients with type II diabetes (UKPDS No. 51) Diabetologia. 2001;44:298–304. doi: 10.1007/s001250051617. [DOI] [PubMed] [Google Scholar]
  • 16.Ong CR, Molyneaux LM, Constantino MI, Twigg SM, Yue DK. Long-term efficacy of metformin therapy in nonobese individuals with type 2 diabetes. Diabetes Care. 2006;29:2361–4. doi: 10.2337/dc06-0827. [DOI] [PubMed] [Google Scholar]
  • 17.Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med. 2008;359:1577–89. doi: 10.1056/NEJMoa0806470. [DOI] [PubMed] [Google Scholar]
  • 18.Diabetes Prevention Program Research Group. Within-trial cost-effectiveness of lifestyle intervention or metformin for the primary prevention of type 2 diabetes. Diabetes Care. 2003;26:2518–23. doi: 10.2337/diacare.26.9.2518. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Fujimoto WY, Jablonski KA, Bray GA, Kriska A, Barrett-Connor E, Haffner S, et al. Body size and shape changes and the risk of diabetes in the Diabetes Prevention Program. Diabetes. 2007;56:1680–5. doi: 10.2337/db07-0009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Lachin JM, Christophi CA, Edelstein SL, Ehrmann DA, Hamman RF, Kahn SE, et al. Factors associated with diabetes onset during metformin versus placebo therapy in the Diabetes Prevention Program. Diabetes. 2007;56:1153–9. doi: 10.2337/db06-0918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Ratner RE, Christophi CA, Metzger BE, Dabelea D, Bennett PH, Pi-Sunyer X, et al. Prevention of diabetes in women with a history of gestational diabetes: effects of metformin and lifestyle interventions. J Clin Endocrinol Metab. 2008;93:4774–9. doi: 10.1210/jc.2008-0772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Perreault L, Kahn SE, Christophi CA, Knowler WC, Hamman RF. Regression from pre-diabetes to normal glucose regulation in the Diabetes Prevention Program. Diabetes Care. 2009;32:1583–8. doi: 10.2337/dc09-0523. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Kitabchi AE, Temprosa M, Knowler WC, Kahn SE, Fowler SE, Haffner SM, et al. Role of insulin secretion and sensitivity in the evolution of type 2 diabetes in the Diabetes Prevention Program: effects of lifestyle intervention and metformin. Diabetes. 2005;54:2404–14. doi: 10.2337/diabetes.54.8.2404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Orchard TJ, Temprosa M, Goldberg R, Haffner S, Ratner R, Marcovina S, et al. The effect of metformin and intensive lifestyle intervention on the metabolic syndrome: the Diabetes Prevention Program randomized trial. Ann Intern Med. 2005;142:611–9. doi: 10.7326/0003-4819-142-8-200504190-00009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Haffner S, Temprosa M, Crandall J, Fowler S, Goldberg R, Horton E, et al. Intensive lifestyle intervention or metformin on inflammation and coagulation in participants with impaired glucose tolerance. Diabetes. 2005;54:1566–72. doi: 10.2337/diabetes.54.5.1566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Ratner R, Goldberg R, Haffner S, Marcovina S, Orchard T, Fowler S, et al. Impact of intensive lifestyle and metformin therapy on cardiovascular disease risk factors in the Diabetes Prevention Program. Diabetes Care. 2005;28:888–94. doi: 10.2337/diacare.28.4.888. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Knowler WC, Fowler SE, Hamman RF, Christophi CA, Hoffman HJ, Brenneman AT, et al. 10-year follow-up of diabetes incidence and weight loss in the Diabetes Prevention Program Outcomes Study. Lancet. 2009;374:1677–86. doi: 10.1016/S0140-6736(09)61457-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Salpeter SR, Greyber E, Pasternak GA, Salpeter EE. Risk of fatal and nonfatal lactic acidosis with metformin use in type 2 diabetes mellitus. Cochrane Database Syst Rev. 2010:CD002967. doi: 10.1002/14651858.CD002967.pub3. [DOI] [PubMed] [Google Scholar]
  • 29.Scarpello JH, Howlett HC. Metformin therapy and clinical uses. Diab Vasc Dis Res. 2008;5:157–67. doi: 10.3132/dvdr.2008.027. [DOI] [PubMed] [Google Scholar]
  • 30.Gunton JE, Delhanty PJ, Takahashi S, Baxter RC. Metformin rapidly increases insulin receptor activation in human liver and signals preferentially through insulin-receptor substrate-2. J Clin Endocrinol Metab. 2003;88:1323–32. doi: 10.1210/jc.2002-021394. [DOI] [PubMed] [Google Scholar]
  • 31.Luo Z, Zang M, Guo W. AMPK as a metabolic tumor suppressor: control of metabolism and cell growth. Future Oncol. 2010;6:457–70. doi: 10.2217/fon.09.174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, et al. Role of AMP-activated protein kinase in mechanism of metformin action. J Clin Invest. 2001;108:1167–74. doi: 10.1172/JCI13505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yoshida T, Okuno A, Tanaka J, Takahashi K, Nakashima R, Kanda S, et al. Metformin primarily decreases plasma glucose not by gluconeogenesis suppression but by activating glucose utilization in a non-obese type 2 diabetes Goto-Kakizaki rats. Eur J Pharmacol. 2009;623:141–7. doi: 10.1016/j.ejphar.2009.09.003. [DOI] [PubMed] [Google Scholar]
  • 34.Zou MH, Kirkpatrick SS, Davis BJ, Nelson JS, Wiles WG, Schlattner U, et al. Activation of the AMP-activated protein kinase by the anti-diabetic drug metformin in vivo. Role of mitochondrial reactive nitrogen species. J Biol Chem. 2004;279:43940–51. doi: 10.1074/jbc.M404421200. [DOI] [PubMed] [Google Scholar]
  • 35.Fujita Y, Hosokawa M, Fujimoto S, Mukai E, Abudukadier A, Obara A, et al. Metformin suppresses hepatic gluconeogenesis and lowers fasting blood glucose levels through reactive nitrogen species in mice. Diabetologia. 2010;53:1472–81. doi: 10.1007/s00125-010-1729-5. [DOI] [PubMed] [Google Scholar]
  • 36.Foretz M, Hebrard S, Leclerc J, Zarrinpashneh E, Soty M, Mithieux G, et al. Metformin inhibits hepatic gluconeogenesis in mice independently of the LKB1/AMPK pathway via a decrease in hepatic energy state. J Clin Invest. 2010;120:2355–69. doi: 10.1172/JCI40671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Maida A, Lamont BJ, Cao X, Drucker DJ. Metformin regulates the incretin receptor axis via a pathway dependent on peroxisome proliferator-activated receptor-alpha in mice. Diabetologia. 2011;54:339–49. doi: 10.1007/s00125-010-1937-z. [DOI] [PubMed] [Google Scholar]
  • 38.Scarpello JH, Hodgson E, Howlett HC. Effect of metformin on bile salt circulation and intestinal motility in type 2 diabetes mellitus. Diabet Med. 1998;15:651–6. doi: 10.1002/(SICI)1096-9136(199808)15:8<651::AID-DIA628>3.0.CO;2-A. [DOI] [PubMed] [Google Scholar]
  • 39.Carter D, Howlett HC, Wiernsperger NF, Bailey CJ. Differential effects of metformin on bile salt absorption from the jejunum and ileum. Diabetes Obes Metab. 2003;5:120–5. doi: 10.1046/j.1463-1326.2003.00252.x. [DOI] [PubMed] [Google Scholar]
  • 40.Carter D, Howlett HC, Wiernsperger NF, Bailey C. Effects of metformin on bile salt transport by monolayers of human intestinal Caco-2 cells. Diabetes Obes Metab. 2002;4:424–7. doi: 10.1046/j.1463-1326.2002.00223.x. [DOI] [PubMed] [Google Scholar]
  • 41.Thomas C, Gioiello A, Noriega L, Strehle A, Oury J, Rizzo G, et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metab. 2009;10:167–77. doi: 10.1016/j.cmet.2009.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Patti ME, Houten SM, Bianco AC, Bernier R, Larsen PR, Holst JJ, et al. Serum bile acids are higher in humans with prior gastric bypass: potential contribution to improved glucose and lipid metabolism. Obesity (Silver Spring) 2009;17:1671–7. doi: 10.1038/oby.2009.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hull RL, Shen ZP, Watts MR, Kodama K, Carr DB, Utzschneider KM, et al. Long-term treatment with rosiglitazone and metformin reduces the extent of, but does not prevent, islet amyloid deposition in mice expressing the gene for human islet amyloid polypeptide. Diabetes. 2005;54:2235–44. doi: 10.2337/diabetes.54.7.2235. [DOI] [PubMed] [Google Scholar]
  • 44.Saydah SH, Loria CM, Eberhardt MS, Brancati FL. Abnormal glucose tolerance and the risk of cancer death in the United States. Am J Epidemiol. 2003;157:1092–100. doi: 10.1093/aje/kwg100. [DOI] [PubMed] [Google Scholar]
  • 45.Michels KB, Solomon CG, Hu FB, Rosner BA, Hankinson SE, Colditz GA, et al. Type 2 diabetes and subsequent incidence of breast cancer in the Nurses Health Study. Diabetes Care. 2003;26:1752–8. doi: 10.2337/diacare.26.6.1752. [DOI] [PubMed] [Google Scholar]
  • 46.La Vecchia C, Negri E, Franceschi S, D’Avanzo B, Boyle P. A case–control study of diabetes mellitus and cancer risk. Br J Cancer. 1994;70:950–3. doi: 10.1038/bjc.1994.427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metformin and reduced risk of cancer in diabetic patients. BMJ. 2005;330:1304–5. doi: 10.1136/bmj.38415.708634.F7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bowker SL, Majumdar SR, Veugelers P, Johnson JA. Increased cancer-related mortality for patients with type 2 diabetes who use sulfonylureas or insulin. Diabetes Care. 2006;29:254–8. doi: 10.2337/diacare.29.02.06.dc05-1558. [DOI] [PubMed] [Google Scholar]
  • 49.Libby G, Donnelly LA, Donnan PT, Alessi DR, Morris AD, Evans JM. New users of metformin are at low risk of incident cancer: a cohort study among people with type 2 diabetes. Diabetes Care. 2009;32:1620–5. doi: 10.2337/dc08-2175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Monami M, Colombi C, Balzi D, Dicembrini I, Giannini S, Melani C, et al. Metformin and cancer occurrence in insulin-treated type 2 diabetic patients. Diabetes Care. 2011;34:129–31. doi: 10.2337/dc10-1287. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Landman GW, Kleefstra N, van Hateren KJ, Groenier KH, Gans RO, Bilo HJ. Metformin associated with lower cancer mortality in type 2 diabetes: ZODIAC-16. Diabetes Care. 2010;33:322–6. doi: 10.2337/dc09-1380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hosono K, Endo H, Takahashi H, Sugiyama M, Sakai E, Uchiyama T, et al. Metformin suppresses colorectal aberrant crypt foci in a short-term clinical trial. Cancer Prev Res (Phila) 2010;3:1077–83. doi: 10.1158/1940-6207.CAPR-10-0186. [DOI] [PubMed] [Google Scholar]
  • 53.Home PD, Kahn SE, Jones NP, Noronha D, Beck-Nielsen H, Viberti G. Experience of malignancies with oral glucose-lowering drugs in the randomised controlled ADOPT (A Diabetes Outcome Progression Trial) and RECORD (Rosiglitazone Evaluated for Cardiovascular Outcomes and Regulation of Glycaemia in Diabetes) clinical trials. Diabetologia. 2010;53:1838–45. doi: 10.1007/s00125-010-1804-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Jiralerspong S, Palla SL, Giordano SH, Meric-Bernstam F, Liedtke C, Barnett CM, et al. 2009 Metformin and pathologic complete responses to neoadjuvant chemotherapy in diabetic patients with breast cancer. J Clin Oncol. 2009;27:3297–302. doi: 10.1200/JCO.2009.19.6410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Bodmer M, Meier C, Krahenbuhl S, Jick SS, Meier CR. Long-term metformin use is associated with decreased risk of breast cancer. Diabetes Care. 2010;33:1304–8. doi: 10.2337/dc09-1791. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Li D, Yeung SC, Hassan MM, Konopleva M, Abbruzzese JL. Antidiabetic therapies affect risk of pancreatic cancer. Gastroenterology. 2009;137:482–8. doi: 10.1053/j.gastro.2009.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Wright JL, Stanford JL. Metformin use and prostate cancer in Caucasian men: results from a population-based case–control study. Cancer Causes Control. 2009;20:1617–22. doi: 10.1007/s10552-009-9407-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.He XX, Tu SM, Lee MH, Yeung SC. Thiazolidinediones and metformin associated with improved survival of diabetic prostate cancer patients. Ann Oncol. 2011 doi: 10.1093/annonc/mdr020. Epub ahead of print. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Vazquez-Martin A, Oliveras-Ferraros C, Menendez JA. The antidiabetic drug metformin suppresses HER2 (erbB-2) oncoprotein overexpression via inhibition of the mTOR effector p70S6K1 in human breast carcinoma cells. Cell Cycle. 2009;8:88–96. doi: 10.4161/cc.8.1.7499. [DOI] [PubMed] [Google Scholar]
  • 60.Alimova IN, Liu B, Fan Z, Edgerton SM, Dillon T, Lind SE, et al. Metformin inhibits breast cancer cell growth, colony formation and induces cell cycle arrest in vitro. Cell Cycle. 2009;8:909–15. doi: 10.4161/cc.8.6.7933. [DOI] [PubMed] [Google Scholar]
  • 61.Hirsch HA, Iliopoulos D, Tsichlis PN, Struhl K. Metformin selectively targets cancer stem cells, and acts together with chemotherapy to block tumor growth and prolong remission. Cancer Res. 2009;69:7507–11. doi: 10.1158/0008-5472.CAN-09-2994. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Vazquez-Martin A, Oliveras-Ferraros C, Cufi S, Del Barco S, Martin-Castillo B, Menendez JA. Metformin regulates breast cancer stem cell ontogeny by transcriptional regulation of the epithelial–mesenchymal transition (EMT) status. Cell Cycle. 2010;9:3807–14. [PubMed] [Google Scholar]
  • 63.Liu B, Fan Z, Edgerton SM, Deng XS, Alimova IN, Lind SE, et al. Metformin induces unique biological and molecular responses in triple negative breast cancer cells. Cell Cycle. 2009;8:2031–40. doi: 10.4161/cc.8.13.8814. [DOI] [PubMed] [Google Scholar]
  • 64.Maatela J, Aromaa A, Salmi T, Pohja M, Vuento M, Gronroos M. The risk of endometrial cancer in diabetic and hypertensive patients: a nationwide record-linkage study in Finland. Ann Chir Gynaecol Suppl. 1994;208:20–4. [PubMed] [Google Scholar]
  • 65.Cantrell LA, Zhou C, Mendivil A, Malloy KM, Gehrig PA, Bae-Jump VL. Metformin is a potent inhibitor of endometrial cancer cell proliferation—implications for a novel treatment strategy. Gynecol Oncol. 2010;116:92–8. doi: 10.1016/j.ygyno.2009.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Tan BK, Adya R, Chen J, Lehnert H, Sant Cassia LJ, Randeva HS. Metformin treatment exerts antiinvasive and antimetastatic effects in human endometrial carcinoma cells. J Clin Endocrinol Metab. 2011;96:808–16. doi: 10.1210/jc.2010-1803. [DOI] [PubMed] [Google Scholar]
  • 67.Rattan R, Giri S, Hartmann LC, Shridhar V. Metformin attenuates ovarian cancer cell growth in an AMP-kinase dispensable manner. J Cell Mol Med. 2011;15:166–78. doi: 10.1111/j.1582-4934.2009.00954.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Yasmeen A, Beauchamp MC, Piura E, Segal E, Pollak M, Gotlieb WH. Induction of apoptosis by metformin in epithelial ovarian cancer: involvement of the Bcl-2 family proteins. Gynecol Oncol. 2011 doi: 10.1016/j.ygyno.2011.02.021. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 69.Kisfalvi K, Eibl G, Sinnett-Smith J, Rozengurt E. Metformin disrupts crosstalk between G protein-coupled receptor and insulin receptor signaling systems and inhibits pancreatic cancer growth. Cancer Res. 2009;69:6539–45. doi: 10.1158/0008-5472.CAN-09-0418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Algire C, Amrein L, Zakikhani M, Panasci L, Pollak M. Metformin blocks the stimulative effect of a high-energy diet on colon carcinoma growth in vivo and is associated with reduced expression of fatty acid synthase. Endocr Relat Cancer. 2010;17:351–60. doi: 10.1677/ERC-09-0252. [DOI] [PubMed] [Google Scholar]
  • 71.Algire C, Zakikhani M, Blouin MJ, Shuai JH, Pollak M. Metformin attenuates the stimulatory effect of a high-energy diet on in vivo LLC1 carcinoma growth. Endocr Relat Cancer. 2008;15:833–9. doi: 10.1677/ERC-08-0038. [DOI] [PubMed] [Google Scholar]
  • 72.Memmott RM, Mercado JR, Maier CR, Kawabata S, Fox SD, Dennis PA. Metformin prevents tobacco carcinogen—induced lung tumorigenesis. Cancer Prev Res (Phila) 2010;3:1066–76. doi: 10.1158/1940-6207.CAPR-10-0055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Iliopoulos D, Hirsch HA, Struhl K. Metformin decreases the dose of chemotherapy for prolonging tumor remission in mouse xenografts involving multiple cancer cell types. Cancer Res. 2011;71:3196–201. doi: 10.1158/0008-5472.CAN-10-3471. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Anisimov VN, Berstein LM, Popovich IG, Zabezhinski MA, Egormin PA, Piskunova TS, et al. If started early in life, metformin treatment increases life span and postpones tumors in female SHR mice. Aging (Albany NY) 2011;3:148–57. doi: 10.18632/aging.100273. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES