Open access peer-reviewed chapter

Repurposing of Metformin as a Multifaceted and Multitasking Preventative and Treatment for Cancer

Written By

Raymond Chang

Submitted: 21 September 2020 Reviewed: 19 January 2021 Published: 05 February 2021

DOI: 10.5772/intechopen.96101

From the Edited Volume

Drug Repurposing - Molecular Aspects and Therapeutic Applications

Edited by Shailendra K. Saxena

Chapter metrics overview

573 Chapter Downloads

View Full Metrics

Abstract

Metformin is a cornerstone treatment of diabetes mellitus. Since 2005 when it has been first reported to reduce the risk of cancer in diabetics, a large number of preclinical and clinical studies have implicated its potential role as a preventative and adjunct therapy for a broad range of cancers. Whereas preclinical studies demonstrate its actions on a multitude of molecular pathways involving nearly all aspects of cancer development including metabolism, angiogenesis, apoptosis, autophagy, immunity, epigenetics, inflammation and crosstalk with the microbiome, other studies demonstrate its synergism with a range of anticancer modalities including chemotherapy, radiotherapy, immunotherapy, and targeted therapies. Furthermore, an increasing number of clinical studies not only confirm its preventative properties against cancers but have extended its potential for a possible adjunctive role in the neoadjuvant, adjuvant, maintenance and salvage therapies of cancer. This article intends to summarize the basic science that allows us to understand the complex multiple mechanisms of action of this remarkable multitasking molecule as well as review the recent meta-analyses that have summarized the clinical studies assessing the therapeutic efficacy of metformin for various cancers.

Keywords

  • metformin
  • diabetes
  • repurposing
  • cancer therapy

1. Introduction

Metformin is derived from the French lilac (also known as goat’s rue or Gallega Officinalis), a medieval European medicinal herb that was first described as a diabetes treatment in a mid-17th century English treatise called Culperper’s Complete Herbal, but it was not until 1957 that the French physician Jean Sterne formally patented metformin as a drug treatment for diabetes. The efficacy of metformin for type 2 diabetes mellitus (T2DM) has since been established and it was approved by the US FDA in 1995 as a treatment for T2DM. Meanwhile by the late 1980’s, studies on the effects of metformin on insulin receptor binding on tumor cells led researchers to conceive that metformin’s effect may potentially be applied for cancer management [1]. Separately, it has long been suspected that T2DM may be a risk for cancer with its cancer promoting effects believed due to hyperinsulinemia in T2DM, since insulin was believed to exert a mitogenic effect [2], thus it was simply logical to investigate the potential benefits of an insulin lowering and hence counter-mitogenic anti-diabetic agent for its possible anti-cancer effects. By the early 2000’s, studies have already established the potential benefits of metformin on hyperinsulinemia, obesity, hyperlipemia, hypertension, fibrinolysis, and endothelial dysfunction, with the expansion of the drug’s potential applicability beyond T2DM to address weight gain, acanthosis nigricans, infertility and polycystic ovary syndrome [3]. In 2005, a landmark retrospective case control study by Evans et al. demonstrated that metformin exposure in T2DM was associated with the reduced risk of cancers [4] and further epidemiological studies also corroborated that diabetics treated with metformin have a lowered incidence of cancer than those treated with other agents, leading to increasing calls for the use of metformin to reduce the risk of cancer. In the past decade, metformin has seen over 50 million prescriptions per year in the US alone and there has been a concurrent explosion of interest in metformin’s anticancer effects with dozens of systematic reviews and meta-analyses performed and published on hundreds of cancer studies involving hundreds of thousands of patients and with hundreds of clinical trials on metformin and cancer currently actively recruiting. The development and expansion of research into metformin’s anticancer activities in the past two decades from the bench to the clinic is illustrated below in Figure 1.

Figure 1.

Research development of metformin as anticancer agent: Since early epidemiologic reports suggesting metformin use in type 2 diabetes was associated with reduced cancer incidence, research evidence that metformin may be preventive and/or therapeutic for human cancers has expanded, with most of the molecular and clinical breakthroughs in metformin and cancer have taken place during the past decades, and hundreds of clinical trials are currently exploring metformin’s potential in cancer. Source: [5], Licensed under CC BY 3.0.

Given that this review is intended as a summary of current clinical evidence for the potential uses of metformin in the prevention and treatment of cancer, we will provide only a succinct synthesis of the thousands of preclinical studies on the biological mechanisms and molecular pathways that has been performed in the past two decades and focus our attention mostly on recent clinical evidence of metformin’s efficacy as demonstrated by clinical studies.

Advertisement

2. Pleiotropic effects of metformin against cancer

The early days of laboratory research on metformin’s anti-cancer mechanisms focused mainly on its metabolic effects on cell proliferation, which naturally follows from the initial use of metformin as a treatment for T2DM as a metabolic disorder. Eventually, it became gradually apparent that unlike modern day targeted therapies, metformin’s anti-neoplastic bioactivity is broad ranged and pleiotropic, encompassing not only its established metabolic effects, but also involving antiangiogenic, anti-inflammatory, epigenetic, apoptotic and autophagic, and immunologic actions as well as effects on the microbiome and on cancer stem cells (CSCs) that all synergistically contribute to overall cancer prevention and control. Furthermore, within each category of its bioactivity, it further exerts multiple molecular actions, and it has thus become increasingly apparent that metformin could be properly conceived of as a multi-faceted multi-tasking molecule with direct and indirect actions against cancer. In summary, the anti-cancer effects of metformin is based on 1) its main action on cellular metabolism via the maintenance of plasma glucose and insulin levels, 2) targeted action against cancer cells with pleiotropic inhibitory effects on multiple pathways involved in cancer cell survival and metastasis, and 3) indirect anti-angiogenic anti-inflammatory as well as immunomodulatory effects and also its actions on the microbiome and CSCs. The complex pleiotropic nature of metformin effects on cancer is illustrated in Figure 2.

Figure 2.

Representation of some of the pleiotropic direct and indirect anticancer effects of metformin as illustrated by molecular and cellular pathways. Metformin effects key energy and metabolic processes such as the mitochondrial respiration (complex I), TCA cycle, fatty acid β-oxidation, gluconeogenesis, and glycolysis. Metformin affects the cell cycle, cell growth, immune response, autophagy, and apoptosis, angiogenesis and cancer stem cells. Abbreviations: 4EBP1, 4E-binding protein 1; ACC, acetyl-CoA carboxylase; AKT, AKT serine/threonine kinase 1; AMPK, AMP-activated protein kinase; BCL2, apoptosis regulator, BCL2; CCND1, cyclin D1; CSC, cancer stem cell; DDIT4, DNA damage inducible transcript 4; EMT, epithelial-to-mesenchymal transition; FOXO3, forkhead box O3; GPDH, glycerol-3-phosphate dehydrogenase; IPMK, inositol polyphosphate multikinase; LKB1, liver kinase B1; miRNA, micro RNA; mTORC1, target of rapamycin complex 1; SREBF1, sterol regulatory element binding transcription factor 1; STAT3, signal transducer and activator of transcription 3; TCA, tricarboxylic acid; TGFB1, transforming growth factor beta 1; VEGF, vascular endothelial growth factor. Phosphorylated molecules are indicated by a prefix p. source: [6], Licensed under CC BY 4.0.

2.1 Metformin metabolic effects

To understand the metabolic impact of metformin on cancer, we must first recognize the intimate relationship between glucose energy metabolism and cellular proliferation as well as a unique propensity of cancer cells to utilize glucose anaerobically even in the presence of oxygen in contrast to non-cancer cells which utilize oxidative phosphorylation to generate energy. This phenomenon was first noted by Otto Warburg almost a hundred years ago, and subsequently termed the “Warburg effect” [7]. This altered energy metabolism of cancer cells may underline their proliferation, invasiveness, and chemoresistance and this altered metabolic pattern in cancer is regulated by oncogenic and tumor suppressor signals such as hypoxia inducible factor 1 (HIF-1), myelocytomatosis oncogene cellular homolog (Myc), p53, and the phosphoinositide 3 kinase (PI3K)/AKT8 virus oncogene cellular homolog (Akt)/mammalian target of rapamycin (mTOR) pathways.

Metformin’s main pharmacologic action is the reducing elevated plasma glucose is largely due to the improvement in hepatic insulin resistance leading to a reduction in hepatic glucose output from gluconeogenesis, increases glucose uptake in muscle, decreased absorption of sugar from the intestines, and improved insulin sensitivity, mainly via activation of a cellular energy sensor known as AMP-activated protein kinase (AMPK). The major downstream target of AMPK is mTOR, which is very important in cellular growth processes and cancer dynamics, and mTOR is inhibited by AMPK [8]. Since glucose metabolism is at the center of the metabolic derangement that is a hallmark of cancer cells, and metformin chiefly targets glucose metabolism, it follows that the altered metabolic pathway may be a target by metformin for cancer prevention or therapy.

It is through its main effects above on metabolism and cellular energetics that metformin can attenuate cancer cell proliferation (See Figure 3). Furthermore, these metabolic effects in turn impact the immune system, epigenetics, inflammation, cellular apoptotic and autophagic pathways as well as the microbiome and CSCs which all play a role in cancer development.

Figure 3.

The effect of metformin in suppressing cancer cell growth via metabolic pathways. Metformin inhibits complex I of the electron transport chain, which leads to increased AMP/ATP ratio and activation of AMPK by LKB1. Activated AMPK subsequently inhibits mTOR and its downstream targets by the following two pathways: 1. AMPK stabilizes TSC1/2, which inhibits Rheb, an activator of mTOR; 2. AMPK inhibits mTOR binding protein raptor. Metformin directly inhibits mTOR by up-regulating REDD1 and suppressing rags. AMPK, AMP-activated protein kinase; Rheb, Ras homolog enriched in brain; LKB1, liver kinase B1; REDD1, regulated in development and DNA damage response 1; TSC, tuberous sclerosis complex; rags, rag GTPases; mTOR, mammalian target of rapamycin; 4EBP1, eukaryotic initiation factor 4E binding protein 1; S6K, S6 kinase. Source: [9], Licensed under CC BY 3.0.

2.2 Metformin immuno-modulatory effects

The immune system participates broadly in the prevention and control of cancer and interacts with biological pathways of metabolism and inflammation, and metformin again acts in a multifaceted fashion to bolster immunity against cancer with effects on almost every aspect of the immune system, especially with reference to cancer immunty (Figure 4). One of metformin’s actions is the enhancement of CD8+ T lymphocytes and rescues them from exhaustion. CD8+ T cells which is one of the key components in cellular immunity against tumors, as these cells can expand and transform into effector cytotoxic T lymphocytes (CTL) which targets cancer. This phenomenon of the rescue of exhausted CD8+ T lymphocytes has been confirmed in vitro in leukemia, melanoma, renal cell carcinoma, non–small-cell lung carcinoma (NSCLC), gastrointestinal carcinoma, and breast cancer. Also, metformin-induced activation of AMPK as one of its main metabolic actions mentioned above promotes immune check-point programmed death ligand 1 (PD-L1) degradation, which allows CTL-mediated tumor cell death [11]. Additionally, metformin can also enhance local as well as systemic cytokine responses to tumors [12]. Furthermore, metformin also has indirect effects on the immune system via its influence on the microbiome and its anti-inflammatory effects, which has been reviewed exhaustively and is briefly summarized below.

Figure 4.

Metformin effects related to anticancer immunity. Metformin indirectly increases T-cell activity by negatively regulating (a) chronic inflammation, (B) hypoxia, and (C) PD-L1 levels that inhibit T-cell activity. Metformin directly relieves T-cell exhaustion by means of metabolic reprogramming of TIL and promotes memory T-cell differentiation (D). Metformin shifts the profile of gut microbiota more favorably to T-cell immunity (TAM) tumor-associated macrophages (E); (Mφ) macrophages; (MDSC) myeloid-derived suppressor cells; (T) T-cell; (DAMPs) damage-associated molecular patterns; (APC) antigen presenting; (SCFA) short-chain fatty acid. Source: [10], CC BY-NC 3.0.

2.3 Metformin effects on the microbiome

Whereas science has become increasingly aware of the central role the gut microbiome plays in health and diseases including cancer, particularly via its effects on the immune system [13], metformin’s beneficial role on host metabolism has also been found to be in part related to the microflora in the gut. The microbiome modulates our immune system and inflammatory response and both of these are key factors in determining cancer development and are associated with inflammatory immune response [14] highlights the crosstalk between metformin effects on metabolism, immunity, inflammation and the microbiome, which in turn can modulate cancer biodyamanics, and part of the mechanisms involved in this complex interplay is illustrated in Figure 5 below.

Figure 5.

Crosstalk between metformin action and gut microbiota. GLP1: Glucagon-like peptide-1; GLP2: Glucagon-like peptide-2; LPS: Lipopolysaccharide; SCFA: Short-chain fatty acid. Source: [15], Licensed under CC BY-NC 4.0.

2.4 Metformin anti-inflammatory effects

Inflammation effects on cancer promotion is well known. In 1863, Rudolf Virchow first proposed the role of inflammation in cancer based on the observation of leukocytes in cancerous tissue. Subsequently, accumulated evidence has identified inflammation both as a cause and result of malignancy [16], with numerous studies in past decades implicating chronic inflammation in the promotion of malignancy [17] (Figure 6). Not surprisingly then, given the T2DM’s known association with chronic low-grade subclinical inflammation which is part and parcel of its the insulin resistance that is its hallmark [19], and metformin’s effects on the immune and metabolic systems, that metformin must also modulate the inflammatory response. This connection has been well demonstrated by animal experiments where metformin treated rodents reveal dampened pro-inflammatory pathways nuclear factor k B (NF-k) and Jun N-terminal kinase (JNK) and increased anti-inflammatory cytokine IL-10 [20].

Figure 6.

Inflammatory cytokines released by immune cells within the tumor microenvironment has a direct effect on pre-malignant and cancer cells by increasing their proliferation and resistance to cell death and stresses thus directly promoting tumor growth and progression. Additionally, inflammatory signals can suppress anti-tumor immunity via action of regulatory T-cells, myeloid cells and enhance other cancer promoting cells (such as fibroblasts, myeloid cells and endothelium of new blood vessels); altogether, these inflammation driven changes also significantly contribute to tumor growths and progression. TME: Tumor microenvironment, Treg: Regulatory T cells. Source: [18], Licensed under CC BY 3.0.

2.5 Metformin epigenetic effects

Epigenetics is the genomic mechanism that reversibly modulates gene expression independent of DNA sequences. Epigenetic processes which allow for the gene modulatory effect involve DNA methylation, histone modification, the readout of these modifications, chromatin remodeling and the effects of noncoding RNA all of which affects cellular activities such as growth and differentiation. Thus, epigenetics can in one sense be conceived of as a master switch of cancer biological processes. Recently, there has been growing interest in epigenetic targeting as a promising therapeutic option for cancer [21]. And since cellular metabolism is tightly linked to epigenetic modifications, it is again not surprising that metformin as a modulator of cellular metabolism may also possess significant epigenetic effects mainly via histone modification (Figure 7), which in turn is another avenue whereby metformin may exert its anti-cancer effects [9].

Figure 7.

Schematic of histone modifications via metabolic effects of metformin. Glycolysis determines the NAD+/NADH ratio, which affects the activity of histone deacetylases to reduce histone acetylation. Source: [9], Licensed under CC BY 3.0.

2.6 Metformin apoptotic and autophagic effects

Both apoptosis or programmed cell death and autophagy are important catabolic and tumor-suppressive pathways that control cell survival and cell death and are thus increasingly important therapeutic targets in cancer [22]. While apoptosis involves cellular suicide and cell death pathways, autophagy involves recycling and degradation of cellular waste which if maladapted and excessive can also lead to cell death and there is significant cross-talk between these two pathways [23]. In cancer biology, autophagy is cancer suppressive as it facilitates the degradation of oncogenic molecules thus pre-empting the development of cancers, while apoptosis leads to cellular suicide and limits the survival of cancer cells. As a result, defective or inadequate autophagy or apoptosis can both lead to cancer. The complexity of the crosstalk between the apoptosis and autophagy is illustrated in Figure 8.

Figure 8.

Complex crosstalk between autophagy and apoptosis pathways. Various proteins involved at the different points of crosstalk are shown and labeled. Lines denote interactions or processes, with solid lines corresponding to intrapathway processes and dashed lines corresponding to inter-pathway connections. Red lines denote inhibitory interactions, while lines with arrows indicate facilitating interactions. Source: [24], Licensed under CC BY 3.0.

In the case of these pathways, metformin has been shown to promote apoptosis in a variety of cancers via various biological pathways [24] while also promoting autophagy [25] as two other dimensions of its anti-cancer bioactivity.

2.7 Metformin effects on cancer stem cells

CSCs were only identified in the 1990s, and they have been hypothesized to persist in tumors as a distinct cell population capable of self-renewal and maybe responsible for cancer relapse and metastasis by giving rise to new tumors. These CSCs are also believed to be resistant to traditional chemotherapy and radiation. A complex regulatory network consisting of microRNAs and Wnt/β-catenin, Notch, and Hedgehog signaling pathways control the properties of CSCs. Therefore, the development of specific therapies targeting and its regulatory pathways is another avenue for improved cancer treatments to prevent relapse and metastases, and improve survival [26]. In this regard, metformin has been reported to target CSCs perhaps via blunting of the Warburg effect and consequently down-regulates their growth. In animal studies, it has been found that metformin exposure was associated with a ~ 2-fold reduction in ovarian CSCs and increased in chemotherapy response and translational studies completed as part of a multi-center phase 2 clinical trial was able to demonstrate a 2.4-fold CSC reduction as well as improved survival in ovarian cancer patients [27].

2.8 Metformin’s antiangiogenic effects

Angiogenesis is the process where a tumor can induce its own blood supply via neovascularization to enhance its own nutrient source as well as increase its propensity to metastasize. It follows that antiangiogenesis which involves the suppression of vascular supply to tumors may be an effective method of cancer control as initially proposed by Folkman [28]. In this regard, preclinical studies with metformin have reported that it indirectly modulates tumor angiogenesis most likely via metabolic pathways affecting proangiogenic signals. As an example, metformin is known to decrease HIF-1α stability in cancer cells, reducing the expression of HIF-1 targeted genes and thus resulting in smaller tumor vessel size, reduced microvessel density and slower tumor growth [29]. Another murine experiment analyzing angiogenesis in a matrigel plug model found that metformin treatment lead to a decrease in angiogenesis [30].

Advertisement

3. Deployment strategies for metformin in cancer

Metformin can be used tactically under various scenarios against cancer. It can be used as standalone or in combination with other agents for the primary or secondary prevention of cancer [31], as neoadjuvant or adjuvant cancer therapy [32], as maintenance therapy or salvage therapy, or to reduce chemoresistance or enhance radiosensitivity [33] as well as for the reduction of side-effects or complications [34]. Notably, metformin is usually deployed as an adjunct but not as a sole agent except in the case of primary prevention. Since it has such low toxicity and multi-faceted mechanisms of actions, it is usually integrated with other treatment agents and modalities under other scenarios besides primary prevention. The key feature of metformin that allows this combinatorial deployment is its low toxicity and its synergism with various other agents and modalities, as it has been demonstrated both in vitro and in vivo.

3.1 Metformin synergies with other anticancer agents and modalities

Notably, synergisms with metformin has been reported with numerous anticancer agents and modalities including chemotherapy [35], targeted drugs [36], and radiotherapy [37]. In the past ten years alone, metformin synergism with chemotherapies pemetrexed [38], temozolomide [39], cisplatin [40], gemcitabine [41], paclitaxel [42], 5FU [43], vincristine [44] with targeted agents erlotinib against non-small cell lung cancer [45], imatinib against colon cancer [46], gefitinib against bladder cancer [47], trastuzumab against human epidermal growth factor receptor 2 (HER2) positive breast cancer [48], celecoxib against NSCLC [49], regorafenib against liver cancer [50], with everolimus as neuroendocrine cancers [51]; and other anticancer agents such as with nelfinavir against cervical cancer [52], propranolol against breast cancer [53], 2-deoxyglucose against ovarian cancer [54], arsenic trioxide against cholangiocarcinoma [55], and with natural compounds epigallocatechin-3-gallate [56], curcumin [57], berberine [58], resveratrol [59].

What is interesting is that different biological mechanisms may be responsible for the efficacy of metformin’s combinatorial effects depending on the specific combination. For example, regulation of lipid synthesis may underlie metformin enhancement of taxanes, pro-apoptotic mechanisms could account for its synergy with cisplatin, AMPK/mTOR signaling maybe significant when combined with hormonal drugs, and suppression of HIF-1, P glycoprotein (p-gp) and multidrug resistance-associated protein 1 (MRP1) expression is thought to be responsible for metformin’s synergy with anti-metabolites [60]. In the case of targeted agents such as the epidermal growth factor receptor (EGFR) inhibitor gefitinib against NSCLC where a Chinese study on diabetic NSCLC patients on gefitinib demonstrated significantly improved response rate, disease control rate, median progression free survival (PFS) and median overall survival (OS) compared with patients controls (70.5% vs. 45.7%, P = 0.017; 97.7% vs. 80.4%, P = 0.009; 19 months vs. 8 months, P = 0.005; 32 months vs. 23 months, P = 0.002, respectively) [61]. Separately, metformin combination with m-TOR inhibitor everolimus in patients with advanced pancreatic neuroendocrine tumors showed improved median PFS of patients treated with the combination vs. control (median PFS, 20.8 months; hazard ratio, 0.49; 95% confidence interval (CI), 0.34–0.69; P < .0001), suggesting that metformin may sensitize everolimus in these patients [62]. As far as combination with antibody treatments go, a randomized phase II study of metformin plus bevacizumab-based chemotherapy in advanced or metastatic NSCLC patients resulted in a 47% (95% CI, 25%–88%) one-year PFS in patients on metformin, which is much improved over a historical control of 15%. Median overall survival of 15.9 months of metformin treated patients was also improved over control arm of 13.9 months [63]. Furthermore, metformin in combination with immune check-point inhibitors (ICI) has received much recent attention as ICI is increasingly being deployed in cancer treatments. A retrospective review of 50 NSCLC patients receiving ICIs as second or third line therapy with or without metformin showed higher overall response rate, disease control, median OS and PFS in the metformin group (41.1 vs. 30.7%, P = 0.4; 70.5 vs. 61.6%, P = 0.5; 11.5 vs. 7.6 months, P = 0.5 and 4.0 vs. 3.0 months, P = 0.6, respectively) [64]. Very recently, several significant trials have been launched to further investigate the role metformin may have in combination with ICI’s, including a metformin-nivolumab combination in patients with NSCLC (NCT03048500), a phase I trial investigating the combined effect of metformin and another anti-PD-L1 antibody durvalamab in head and neck squamous cell carcinoma (NCT03618654), a phase I trial of metformin in combination of the anti-PD-1 antibody pembrolizumab in advanced melanoma (NCT03311308), and a phase II trial combining metformin with nivolumab in stage IV colorectal cancer that has not responded to previous treatment (NCT03800602).

The use of metformin under various scenarios against cancer has been best studied clinically for primary prevention and in the neoadjuvant setting and some of the relevant data is summarized below.

3.2 Metformin for primary prevention of cancer

Cancer prevention is the earliest role that metformin was hypothesized to play in the disease as it was Evans’ original 2005 retrospective case–control study demonstrating metformin’s involvement in reducing cancer risk in T2DM that highlighted its potential for cancer [4]. Subsequently, a confirmative cohort study of T2DM with metformin followed in which the frequency of cancer was significantly lower in patients receiving metformin versus controls who had never received metformin, after adjusting for body mass index, hemoglobin A1C, smoking and the use of other drugs [65], a finding that was subsequently repeatedly confirmed. Indeed, meta-analyses have demonstrated that metformin is associated with a decreased risk of breast, colon, liver, pancreas, prostate, endometrium and lung cancer across meta-analyses [31] suggesting that people with T2DM receiving metformin demonstrate a lower risk and improved outcomes with most common cancers; more specifically one meta-analysis found that metformin-treated T2DM patients had a 31% reduction in the incidence of cancer and a 34% reduction in cancer mortality after adjusting for body mass index [66].

3.3 Metformin in neoadjuvant treatment

Neoadjuvant effects of metformin in combination or alone has been clinically explored in several cancers types. In one study of two hundred eighty-five patients with esophageal adenocarcinoma treated with concurrent chemoradiation followed by esophagectomy, complete remission (CR) was higher in T2DM patients taking metformin (34.5%) compared to those who are not (4.8%, P = 0.01) as well as non-diabetic patients who are not on the drug (19.6%, P = 0.05) and furthermore the CR rate was found to be related to metformin dose, with ≥1500 mg per day associated with a higher CR rate [67]. In a separate study of diabetic rectal cancer patients undergoing neoadjuvant chemoradiotherapy, those on metformin experienced better tumor responses (P = 0.002), pathologic complete remission (p = 0.037), and N downstaging (P < 0.001) as well as experienced improved cancer specific survival and lower risk of recurrence [68]. Separately, women with endometrial cancer on neoadjuvant metformin 850 mg twice daily for an average of 20 days between diagnosis and surgery had reduced cell proliferation per Ki-67 expression, compared to the untreated [69]. A similar biomarker based on a “window of opportunity” assessment of metformin 500 mg three times daily for a median duration of 18 days in non-diabetic breast cancer also demonstrated that short-term preoperative metformin resulted in both clinical and cellular changes including a significant decrease in the Ki-67 proliferation index from 36.5 to 33.5% (P = 0.016) [70]. Separately and perhaps more significantly, a study involving early-stage breast cancer assessing remission rates after neoadjuvant therapy among metformin vs. non-metformin users found a significant difference in CR of 24% in the metformin group, 8.0% in the non-metformin group, and 16% in the non-diabetic group, with metformin use independently predictive of response (OR 2.95; P = 0.04) after adjustment for diabetes, body mass index, age, stage, grade, receptor status, and neoadjuvant chemotherapy use by multivariate logistic regression [71].

Advertisement

4. Systematic reviews and meta-analyses on metformin clinical outcomes in various cancers

Since metformin is so versatile and has been studied in a wide variety of settings from the laboratory to bedside, and since this review is intended to focus on the clinical deployment of metformin, it is thus useful to have a summary perspective of its potential usefulness in cancer by reviewing clinical results as recently meta-analyzed for various cancers.

4.1 Bladder cancer

A review of 9 retrospective cohort studies with 1,270,179 patients did not reveal a benefit from metformin in preventing bladder cancer (Hazard ratio (HR) = 0.82, 95% CI = 0.61–1.09; P = .17). However, metformin intake was associated with an improved recurrence-free survival (HR = 0.55, 95% CI = 0.35–0.88; P = .01), progression-free survival (HR = 0.70, 95% CI = 0.51–0.96; P = .03), as well as cancer-specific survival (HR = 0.57, 95% CI = 0.40–0.81; P = .002) [72].

4.2 Breast cancer

There have been a number of studies relating to metformin’s effect on biomarkers in breast cancer patients and it has been shown that metformin therapy reduced the levels of insulin, sex hormones and sex hormone-binding globulin, Ki67, caspase-3, p-Akt, obesity, CRP, blood glucose and lipid profile overall [73]. More, in a clinical trial to examine the clinical and biological effects of neoadjuvant metformin on patients with breast cancer, non-diabetic women with untreated breast cancer given 500 mg of metformin three times daily for ≥2 weeks exhibited decreased insulin receptor expression (P = 0.04), phosphorylation status of protein kinase B /Akt, extracellular signal-regulated kinase 1/2, AMPK and acetyl coenzyme A carboxylase (P = 0.0001, P < 0.0001, P < 0.005 and P = 0.02, respectively) in tumors correlating with decreases in tumor cell proliferation and increases in apoptosis [74]. In T2DM patients with breast cancer, a 2018 meta-analysis of eleven studies of all-cause mortality found a 45% risk reduction was observed for all-cause mortality (HR = 0.55; 95% CI 0.44–0.70) and concluded that metformin may improve overall survival in this patient subset [75]. Separately in another review, 7 observational studies showed significantly reduced breast cancer risk among T2DM patients on metformin OR = 0.83 (CI 0.71–0.97) [76]. Separately, in a sub-study involving over four hundred diabetic patients in the large phase 3 ALTTL trial of Her2+ breast cancer patients, Her2+ and estrogen receptor positive breast cancer cases on metformin experienced had improved disease free survival, metastasis free disease survival and overall survival over those patients not on metformin over a median of four and a half years [77]. However despite the vast amount of preclinical and epidemiologic data on its benefits in breast cancer, there are no trials in non-diabetic breast cancer patients to date which have unequivocally demonstrated a clinical benefit of metformin.

4.3 Colon cancer

Ng et al. from Singapore found 58 studies that provided incidences of colorectal adenoma and cancer and cancer survival outcomes and found that metformin significantly lowered the risk of colorectal adenoma (RR 0.77, CI 0.67–0.88, P < 0.001), advanced adenoma (0.61, CI 0.42–0.88, P = 0.008) and colorectal cancer (RR 0.76, CI 0.69–0.84, P < 0.001) respectively. Overall cancer survival (HR 0.6, CI 0.53–0.67, P < 0.001), even among metastatic cases was also higher among metformin users (HR 0.77, CI 0.68–0.87, P < 0.001), and it was concluded that metformin significantly reduces colorectal adenoma and cancer incidence as well as enhanced colorectal cancer survival at all stages [78].

4.4 Endometrial cancer

In 19 studies reviewed in 2017, metformin used reversed atypical endometrial hyperplasia to normal, and decreased cell proliferation from 51.94% (CI = 36.23% to 67.46%) to 34.47% (CI = 18.55% to 52.43%) [79], while separately, a review of seven studies showed that metformin could significantly improve overall survival of in endometrial cancer (HR = 0.61, 95% CI 0.48–0.77, P < 0.05) and reduce their recurrence risk (OR = 0.50, 95% CI 0.28–0.92, P < 0.05) [80], whereas another review of six retrospective cohorts if 4723 endometrial cancer cases demonstrated that metformin use was associated with a significant reduction in overall mortality in comparison with not using metformin (adjusted HR 0.64, 95% CI 0.45–0.89, P = 0.009) irrespective of diabetic status [81], and these results corroborated the improved overall (HR, 0.58; 95% CI, 0.45–0.76; P = 0.207) as well as progression free survival (HR, 0.61; 95% CI, 0.49–0.76; P = 0.768) found in another review of 6242 patients from fourteen studies [82].

4.5 Lung cancer

An analysis of 13 observational studies found lung cancer incidence to be reduced in diabetic patients on metformin vs. no metformin (RR = 0.89; 95% CI, 0.83–0.96; P = 0.002) [83]. A separate meta-analysis found six studies comparing metformin usage and non-metformin usage significantly improved overall survival in diabetic patients with NSCLC [pooled HR =0.87 (0.77–0.99), P = 0.04] [84]. Especially noteworthy was an ambitious prospective clinical trial conducted by Marrone et al. which studied non-diabetics with advanced or metastatic NSCLC receiving platinum-based doublet chemotherapy and bevacizumab with or without metformin 1000 mg twice daily followed by maintenance therapy with bevacizumab and metformin combined or bevacizumab alone and showed a significant clinical benefit in PFS (9.6 vs. 6.7 months) with the addition of metformin [63].

4.6 Pancreas cancer

A review of seventeen studies involving 36791 participants study has evidenced a significant association of metformin adjuvant treatment in pancreas cancer with overall survival benefit (HR = 0.88, 95% CI = 0.80–0.97) especially in Asians, those with early stage disease and those undertaking surgery [85]. In terms of overall survival with metformin use in pancreas cancer, a study of 8 retrospective cohort studies and 2 randomized clinical trials representing 3,042 patients revealed overall survival to be improved with metformin (meta-HR = 0.79; 95% CI: 0.70, 0.92, P < 0.001) [86].

4.7 Prostate cancer

In a systematic review involving eleven studies with 877,058 patients, the odds ratio of metformin use for reducing prostate cancer was estimated at 0.89 (95%CI: 0.67–1.17) and it was concluded that metformin consumption reduced the risk of prostate cancer, although the result was not statistically significant [87]. Separately, a review of eight studies on diabetic patients with prostate cancer found no metformin use was associated with an increased risk of cancer recurrence (RR, 1.20; 95% CI, 1.00–1.44) [88], which concurs with another review of eight retrospective cohort studies and one nested-case–control study, metformin was found to be associated with a reduced risk of biochemical recurrence (pHR: 0.82, 95% CI 0.67, 1.01, P = 0.06) [89]. Finally, a large review of 30 cohort studies, including 1,660,795 prostate cancer patients revealed that metformin treatment compared with no treatment improved overall, prostate cancer specific, and recurrence free survival (HR = 0.72, 95% CI: 0.59–0.88, P = 0.001; HR = 0.78, 95% CI: 0.64–0.94, P = 0.009; and HR = 0.60, 95% CI: 0.42–0.87 P = 0.006, respectively) [90].

4.8 Ovarian cancer

One review of 13 studies involving ovarian cancer incidence and prognosis revealed metformin use to be associated with a lower incidence (pooled OR 0.76, 95% CI 0.62 to 0.93, P = 0.008) as well as improved prognosis (pooled OR 0.55, 95% CI 0.36 to 0.84, P = 0.006) [91].

4.9 Other cancers

Metformin is also increasingly studied or planned in less common cancers, such as glioblastoma, thyroid cancer, and non-Hodgkin’s lymphoma. The recent study on newly diagnosed glioblastoma showed that temozolomide plus memantine, mefloquine, and metformin are feasible as an adjuvant therapy [92]. One planned phase 1b/2 clinical trial of metformin and chloroquine was recruiting patients with IDH1-mutated or IDH2-mutated solid tumors, including glioma [93]. In another recent retrospective study from Korea, cancer preventative effects of metformin on thyroid cancer were observed in individuals with T2DM on long duration or higher doses of the drug [94]. Separately, a trial in head and neck squamous cell cancer patients revealed metformin to inhibit cancer by enhancing apoptosis, and increasing cellular immune infiltration of the cancer [95]. In non-Hodgkin’s lymphoma, a retrospective analysis of looking at T2DM patients treated with standard therapy found improved progression-free survival and overall survival compared to control not taking metformin [96].

Advertisement

5. Discussion

Any discussion of a therapeutic agent is incomplete without covering its toxicity, side-effects and drug interactions. In this regard, metformin is probably one of the safest drugs in use, especially when compared with standard anti-cancer agents in its context as a potential cancer preventative or therapeutic. With its long history of widespread use, its pharmacokinetics and toxicity profile are well established. The most common side-effect is mild to moderate gastrointestinal discomfort or diarrhea which is usually self-limited and can be minimized if metformin is taken with food, while its most serious side-effect of lactic acidosis usually due to overdose is relatively rare, occurring once per 100,000 years of use or 3 case per 1,000,000 after long term treatment [97]. As in the case of all medications, it should be dispensed carefully in elderly patients and in those with impaired renal, cardiac, and hepatic function. For practical purposes, it needs to be emphasized that metformin as an antidiabetic and as monotherapy does not cause hypoglycemia or weight gain, unlike insulin or sulfonylureas. For cancer, because of its very common use in diabetics, it has practically seen combined use with most oncologic agents in the diabetic cancer patient and remarkably no serious interactions with standard cancer anti-cancer agents have been reported. The minimum toxic dose of metformin is not well defined, but rare case reports of severe toxicity has only been reported after ingestion of 25 to 35 grams of metformin by adults.

A treatment for any condition is ideal if it relatively non-toxic and scientifically well evidenced, as well as low in cost and convenient to administer. Metformin fits all the above criteria. It is apparent from our review that metformin has ample scientific evidence from bench to the bedside as a repurposed drug for cancer. In fact, it is safe to say that it is currently the most well evidenced repurposed drug for cancer. Also, its wide-spread and decades of experience of clinical use and low observed toxicities alone or in combination with other agents, as well as very low cost also marks it as an optimal therapeutic agent. Finally, the versatility it possesses against various cancers and its applicability from prevention to treatment further distinguishes it as an ideal or model repurposed drug for cancer.

Of course, there remains limitations and challenges to metformin’s use as an anticancer. The first obstacle we have in translating in vitro results of metformin to the clinical arena revolves around its dosage. The usual dosage of metformin in cancer trials is the same range as that prescribed normally for T2DM which is from 1000 mg – 2000 mg per day. Treatment is usually started at the lower dose with dose escalations weekly to the maximum dose which means starting at 500 mg of the immediate release version twice daily or 850 mg of the extended release versions once daily, with 500 mg increments weekly as tolerated, to a maximum of 2000–2550 mg per day for either immediate or extended release versions. It may not be apparent at first glance, but the concentration of metformin at 10–100 microM when used clinically at 1000-2000 mg per day is much less than the concentration of >2–5 mM demonstrated for its anti-cancer effects in vitro where metformin was usually experimented at concentrations between 5 to 20 mM, which is 2 000–10 000 times more concentrated than achieved with clinical dosing [98]. Fortunately, many clinical studies still yielded positive results at the much lower metformin in concentrations achieved with clinical dosing, but it may also explain why the clinical results of metformin in cancer may not be as dramatic as demonstrated in preclinical studies, and why it is never intended to be used as monotherapy for cancer treatment. Related to dosing is a possible dose-dependent effect of metformin on cancer risk [99], which raises the question of attempting higher doses of metformin in future clinical trials of metformin in cancer, this while taking into account that there are no cases of acute metformin overdose leading to death found in which patients with a peak serum metformin concentration is under 50 microg/mL [100]. Beyond dosing, another issue with the literature to date on metformin and cancer is that most of the clinical studies so far are retrospective that mainly involve observations in the T2DM patient population and thus subject to selection bias. However, many cohort studies in the non-diabetic is planned, and despite methodological limitations, it is apparent that the overwhelming evidence so far is in favor of potential benefits and a high benefit to risk and benefit to cost ratios for metformin’s application in cancer.

Advertisement

6. Conclusion

As an old repurposed drug, metformin is inexpensive and generic and its research is thus carried out usually without industry support. Despite such challenges, it is heartening that overall preclinical and clinical results is overwhelmingly suggestive of a protective effect from metformin against various stages of a wide spectrum of cancers. Moreover, there are over three hundred registered clinical trials on metformin and cancer internationally as of mid-2020, of which approximately one third are actively recruiting. The trials involve metformin for pre-cancers, early stage as well as metastatic solid tumors, alone or in combination with other interventions including chemotherapy, radiotherapy, hormone therapy, immunotherapy (ICIs), targeted agents, statins, aspirin, doxycycline, nelfinavir, melatonin, disulfiram, vitamin C, diet in diabetics and non-diabetics. We thus look forward for the further establishment of metformin as an ideal repurposed agent for cancer prevention and treatment.

Acronyms and abbreviations

Akt

AKT8 virus oncogene cellular homolog

AMPK

AMP-activated protein kinase

CI

confidence interval

CR

complete remission

CSC

cancer stem cell

CTL

cytotoxic T lymphocytes

EGFR

epidermal growth factor receptor

GPDH

glycerol-3-phosphate dehydrogenase

HER2

human epidermal growth factor receptor 2

HIF

hypoxia inducible factor; HR: hazard ratio

hs-CRP

C-reactive protein

ICI

immune checkpoint inhibitor

JNK

Jun N-terminal kinase

mTOR

mammalian target of rapamycin

MRP-1

multidrug resistance-associated protein 1

Myc

myelocytomatosis oncogene cellular homolog

NAD+/NADH

nicotinamide adenine dinucleotide (oxidized)/nicotinamide adenine dinucleotide + hydrogen

NF-kB

nuclear factor k B

NSCLC

non-small cell lung cancer

OS

overall survival

PD-1

programmed cell death protein 1

PD-L1

programmed death-ligand 1

p-gp

P glycoprotein

PI3K

phosphoinositide 3 kinase

PFS

progression free survival

T2DM

type 2 diabetes mellitus

Treg

regulatory T cell

References

  1. 1. Mountjoy KG, Finlay GJ, Holdaway IM. Effects of metformin and glibenclamide on insulin receptors in fibroblasts and tumor cells in vitro. J Endocrinol Invest. 1987 Dec 27;10(6):553-557.
  2. 2. Czyżyk A, Szczepanik Z. Diabetes mellitus and cancer. Eur J Intern Med. 2000 Oct;11(5):245-252.
  3. 3. Tankova T. Current indications for metformin therapy. Rom J Intern Med. 2003;41(3):215-225.
  4. 4. Evans JMM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metformin and reduced risk of cancer in diabetic patients. BMJ. 2005 Jun 4;330(7503):1304-1305.
  5. 5. Del Barco, S. et al. Metformin: multifaceted protection against cancer. Oncotarget 2011, 2(12):896-917.
  6. 6. Schulte, H. Pleiotropic effects of metformin on cancer, Int. J. Mol. Sci. 2018, 19(10), 2850; https://doi.org/10.3390/ijms19102850
  7. 7. Ferreira LMR. Cancer metabolism: The Warburg effect today. Exp Mol Pathol. 2010 Dec;89(3):372-380.
  8. 8. Soukas AA, Hao H, Wu L. Metformin as Anti-Aging Therapy: Is It for Everyone? Trends Endocrinol Metab. 2019 Oct;30(10):745-755.
  9. 9. Yu X, Mao W, Zhai Y, Tong C, Liu M, Ma L, et al. Anti-tumor activity of metformin: from metabolic and epigenetic perspectives. Oncotarget. 2017 Jan 17;8(3):5619-5628.
  10. 10. Kim KJ et al. A new aspect of an old friend: the beneficial effect of metformin on anti-tumor immunity. BMB Rep. 2020 Jul 31:5114
  11. 11. Verdura S, Cuyàs E, Martin-Castillo B, Menendez JA. Metformin as an archetype immuno-metabolic adjuvant for cancer immunotherapy. Oncoimmunology. 2019;
  12. 12. Pearce EL, Walsh MC, Cejas PJ, Harms GM, Shen H, Wang L-S, et al. Enhancing CD8 T-cell memory by modulating fatty acid metabolism. Nature. 2009 Jul 3;460(7251):103-107.
  13. 13. Gopalakrishnan V, Helmink BA, Spencer CN, Reuben A, Wargo JA. The Influence of the Gut Microbiome on Cancer, Immunity, and Cancer Immunotherapy. Cancer Cell. 2018 Apr;33(4):570-580.
  14. 14. Forslund K, Hildebrand F, Nielsen T, Falony G, Le Chatelier E, Sunagawa S, et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature. 2015 Dec 2;528(7581):262-266.
  15. 15. Kyriachenko Y, et al. Crosstalk between gut microbiota and anti-diabetic drug action. World J Diabetes. 2019, 10(3):154-168. doi: 10.4239/wjd.v10.i3.154
  16. 16. Grivennikov SI, Greten FR, Karin M. Immunity, Inflammation, and Cancer. Cell. 2010 Mar;140(6):883-899.
  17. 17. Demaria S, Pikarsky E, Karin M, Coussens LM, Chen Y-C, El-Omar EM, et al. Cancer and Inflammation: Promise for Biologic Therapy. J Immunother. 2010 May;33(4):335-351.
  18. 18. Greten, FR, Grivennokov SI. Inflammation and cancer: triggers, mechanisms and consequences. Immunity. 2019, 51(1):27-21. doi:10.1016/j.immuni.2019.06.025
  19. 19. Mathews ST, Kothari V, Galdo J. Hypoglycemic agents and potential anti-inflammatory activity. J Inflamm Res. 2016 Apr;27.
  20. 20. Martin-Montalvo A, Mercken EM, Mitchell SJ, Palacios HH, Mote PL, Scheibye-Knudsen M, et al. Metformin improves healthspan and lifespan in mice. Nat Commun. 2013 Oct 30;4(1):2192.
  21. 21. CHEN QW, ZHU XY, LI YY, MENG ZQ. Epigenetic regulation and cancer (Review). Oncol Rep. 2014 Feb;31(2):523-532.
  22. 22. Liu J jie, Lin M, Yu J ying, Liu B, Bao J ku. Targeting apoptotic and autophagic pathways for cancer therapeutics. Cancer Letters. 2011.
  23. 23. Su M, Mei Y, Sinha S. Role of the Crosstalk between Autophagy and Apoptosis in Cancer. J Oncol. 2013;2013:1-14.
  24. 24. Queiroz EAIF, Puukila S, Eichler R, Sampaio SC, Forsyth HL, Lees SJ, et al. Metformin Induces Apoptosis and Cell Cycle Arrest Mediated by Oxidative Stress, AMPK and FOXO3a in MCF-7 Breast Cancer Cells. Katoh M, editor. PLoS One. 2014 May 23;9(5):e98207.
  25. 25. Wang Y, Xu W, Yan Z, Zhao W, Mi J, Li J, et al. Metformin induces autophagy and G0/G1 phase cell cycle arrest in myeloma by targeting the AMPK/mTORC1 and mTORC2 pathways. J Exp Clin Cancer Res. 2018 Dec 20;37(1):63.
  26. 26. Bao B, Ahmad A, Azmi AS, Ali S, Sarkar FH. Overview of Cancer Stem Cells (CSCs) and Mechanisms of Their Regulation: Implications for Cancer Therapy. Curr Protoc Pharmacol. 2013 Jun;61(1):14.25.1-14.25.14.
  27. 27. Buckanovich RJ, Brown J, Shank J, Griffith KA, Reynolds RK, Johnston C, et al. A phase II clinical trial of metformin as a cancer stem cell targeting agent in stage IIc/III/IV ovarian, fallopian tube, and primary peritoneal cancer. J Clin Oncol. 2017 May 20;35(15_suppl):5556-5556.
  28. 28. FOLKMAN J. Antiangiogenesis in cancer therapy—endostatin and its mechanisms of action. Exp Cell Res. 2006 Mar 10;312(5):594-607.
  29. 29. Kurelac I, Umesh Ganesh N, Iorio M, Porcelli AM, Gasparre G. The multifaceted effects of metformin on tumor microenvironment. Semin Cell Dev Biol. 2020 Feb;98:90-97.
  30. 30. Dallaglio K, Bruno A, Cantelmo AR, Esposito AI, Ruggiero L, Orecchioni S, et al. Paradoxic effects of metformin on endothelial cells and angiogenesis. Carcinogenesis. 2014 May;35(5):1055-1066.
  31. 31. Heckman-Stoddard BM, DeCensi A, Sahasrabuddhe V V., Ford LG. Repurposing metformin for the prevention of cancer and cancer recurrence. Diabetologia. 2017 Sep 3;60(9):1639-1647.
  32. 32. Coyle C, Cafferty FH, Vale C, Langley RE. Metformin as an adjuvant treatment for cancer: a systematic review and meta-analysis. Ann Oncol Off J Eur Soc Med Oncol. 2016;
  33. 33. Zhang T, Zhang L, Zhang T, Fan J, Wu K, Guan Z, et al. Metformin Sensitizes Prostate Cancer Cells to Radiation Through EGFR/p-DNA-PKCS In Vitro and In Vivo. Radiat Res. 2014 May 20;181(6):641.
  34. 34. Smith TAD, Phyu SM, Akabuogu EU. Effects of Administered Cardioprotective Drugs on Treatment Response of Breast Cancer Cells. Anticancer Res. 2016 Jan;36(1):87-93.
  35. 35. Zhang H-H, Guo X-L. Combinational strategies of metformin and chemotherapy in cancers. Cancer Chemother Pharmacol. 2016 Jul 27;78(1):13-26.
  36. 36. Deng J, Peng M, Wang Z, Zhou S, Xiao D, Deng J, et al. Novel application of metformin combined with targeted drugs on anticancer treatment. Cancer Sci. 2019 Jan;110(1):23-30.
  37. 37. Samsuri NAB, Leech M, Marignol L. Metformin and improved treatment outcomes in radiation therapy – A review. Cancer Treat Rev. 2017 Apr;55:150-162.
  38. 38. Zhang Y, Feng X, Li T, Yi E, Li Y. Metformin synergistic pemetrexed suppresses non-small-cell lung cancer cell proliferation and invasion in vitro. Cancer Med. 2017;
  39. 39. Lee JE, Lim JH, Hong YK, Yang SH. High-Dose Metformin Plus Temozolomide Shows Increased Anti-tumor Effects in Glioblastoma In Vitro and In Vivo Compared with Monotherapy. Cancer Res Treat. 2018 Oct 15;50(4):1331-1342.
  40. 40. Lee JO, Kang MJ, Byun WS, Kim SA, Seo IH, Han JA, et al. Metformin overcomes resistance to cisplatin in triple-negative breast cancer (TNBC) cells by targeting RAD51. Breast Cancer Res. 2019 Dec 22;21(1):115.
  41. 41. Chai X, Chu H, Yang X, Meng Y, Shi P, Gou S. Metformin Increases Sensitivity of Pancreatic Cancer Cells to Gemcitabine by Reducing CD133+ Cell Populations and Suppressing ERK/P70S6K Signaling. Sci Rep. 2015 Nov 22;5(1):14404.
  42. 42. dos Santos Guimarães I, Ladislau-Magescky T, Tessarollo NG, dos Santos DZ, Gimba ERP, Sternberg C, et al. Chemosensitizing effects of metformin on cisplatin- and paclitaxel-resistant ovarian cancer cell lines. Pharmacol Reports. 2018 Jun;70(3):409-417.
  43. 43. Harada K, Ferdous T, Harada T, Ueyama Y. Metformin in combination with 5-fluorouracil suppresses tumor growth by inhibiting the Warburg effect in human oral squamous cell carcinoma. Int J Oncol. 2016 Jul;49(1):276-284.
  44. 44. Yi Y, Gao L, Wu M, Ao J, Zhang C, Wang X, et al. Metformin Sensitizes Leukemia Cells to Vincristine via Activation of AMP-activated Protein Kinase. J Cancer. 2017;8(13):2636-2642.
  45. 45. Wang X, Chen K, Yu Y, Xiang Y, Kim JH, Gong W, et al. Metformin sensitizes lung cancer cells to treatment by the tyrosine kinase inhibitor erlotinib. Oncotarget. 2017 Dec 12;8(65):109068-109078.
  46. 46. Lee J, Park D, Lee Y. Metformin Synergistically Potentiates the Antitumor Effects of Imatinib in Colorectal Cancer Cells. Dev Reprod. 2017 Jun;21(2):139-150.
  47. 47. Peng M, Huang Y, Tao T, Peng C-Y, Su Q, Xu W, et al. Metformin and gefitinib cooperate to inhibit bladder cancer growth via both AMPK and EGFR pathways joining at Akt and Erk. Sci Rep. 2016 Jun 23;6(1):28611.
  48. 48. Liu B, Fan Z, Edgerton SM, Yang X, Lind SE, Thor AD. Potent anti-proliferative effects of metformin on trastuzumab-resistant breast cancer cells via inhibition of erbB2/IGF-1 receptor interactions. Cell Cycle. 2011 Sep 28;10(17):2959-2966.
  49. 49. Cao N, Lu Y, Liu J, Cai F, Xu H, Chen J, et al. Metformin Synergistically Enhanced the Antitumor Activity of Celecoxib in Human Non-Small Cell Lung Cancer Cells. Front Pharmacol. 2020;
  50. 50. Yang Q, Guo X, Yang L. Metformin Enhances the Effect of Regorafenib and Inhibits Recurrence and Metastasis of Hepatic Carcinoma After Liver Resection via Regulating Expression of Hypoxia Inducible Factors 2α (HIF-2α) and 30 kDa HIV Tat-Interacting Protein (TIP30). Med Sci Monit. 2018 Apr 14;24:2225-2234.
  51. 51. Hue-Fontaine L, Lemelin A, Forestier J, Raverot G, Milot L, Robinson P, et al. Metformin and everolimus in neuroendocrine tumours: A synergic effect? Clin Res Hepatol Gastroenterol. 2020 Mar;
  52. 52. Xia C, Chen R, Chen J, Qi Q, Pan Y, Du L, et al. Combining metformin and nelfinavir exhibits synergistic effects against the growth of human cervical cancer cells and xenograft in nude mice. Sci Rep. 2017 Mar 2;7(1):43373.
  53. 53. Rico M, Baglioni M, Bondarenko M, Laluce NC, Rozados V, André N, et al. Metformin and propranolol combination prevents cancer progression and metastasis in different breast cancer models. Oncotarget. 2017 Jan 10;8(2):2874-2889.
  54. 54. Zhu J, Zheng Y, Zhang H, Sun H. Targeting cancer cell metabolism: The combination of metformin and 2-Deoxyglucose regulates apoptosis in ovarian cancer cells via p38 MAPK/JNK signaling pathway. Am J Transl Res. 2016;8(11):4812-4821.
  55. 55. Ling S, Xie H, Yang F, Shan Q, Dai H, Zhuo J, et al. Metformin potentiates the effect of arsenic trioxide suppressing intrahepatic cholangiocarcinoma: roles of p38 MAPK, ERK3, and mTORC1. J Hematol Oncol. 2017 Dec 28;10(1):59.
  56. 56. Yu C, Jiao Y, Xue J, Zhang Q, Yang H, Xing L, et al. Metformin Sensitizes Non-small Cell Lung Cancer Cells to an Epigallocatechin-3-Gallate (EGCG) Treatment by Suppressing the Nrf2/HO-1 Signaling Pathway. Int J Biol Sci. 2017;13(12):1560-1569.
  57. 57. Zhang HH, Zhang Y, Cheng YN, Gong FL, Cao ZQ, Yu LG, et al. Metformin incombination with curcumin inhibits the growth, metastasis, and angiogenesis of hepatocellular carcinoma in vitro and in vivo. Mol Carcinog. 2018;
  58. 58. Akula SM, Candido S, Libra M, Abrams SL, Steelman LS, Lertpiriyapong K, et al. Abilities of berberine and chemically modified berberines to interact with metformin and inhibit proliferation of pancreatic cancer cells. Adv Biol Regul. 2019 Aug;73:100633.
  59. 59. LEE Y-S, DOONAN BB, WU JM, HSIEH T-C. Combined metformin and resveratrol confers protection against UVC-induced DNA damage in A549 lung cancer cells via modulation of cell cycle checkpoints and DNA repair. Oncol Rep. 2016 Jun;35(6):3735-3741.
  60. 60. Peng M, Darko KO, Tao T, Huang Y, Su Q, He C, et al. Combination of metformin with chemotherapeutic drugs via different molecular mechanisms. Cancer Treat Rev. 2017 Mar;54:24-33.
  61. 61. Li K, Li L, Zhang P, Kang J, Wang Y, Chen H, et al. A Multicenter Double-blind Phase II Study of Metformin With Gefitinib as First-line Therapy of Locally Advanced Non–Small-cell Lung Cancer. Clin Lung Cancer. 2017 May;18(3):340-343.
  62. 62. Pusceddu S, Vernieri C, Di Maio M, Marconcini R, Spada F, Massironi S, et al. Metformin Use Is Associated With Longer Progression-Free Survival of Patients With Diabetes and Pancreatic Neuroendocrine Tumors Receiving Everolimus and/or Somatostatin Analogues. Gastroenterology. 2018 Aug;155(2):479-489.e7.
  63. 63. Marrone KA, Zhou X, Forde PM, Purtell M, Brahmer JR, Hann CL, et al. A Randomized Phase II Study of Metformin plus Paclitaxel/Carboplatin/Bevacizumab in Patients with Chemotherapy-Naïve Advanced or Metastatic Nonsquamous Non-Small Cell Lung Cancer. Oncologist. 2018 Jul 27;23(7):859-865.
  64. 64. Afzal MZ, Dragnev K, Sarwar T, Shirai K. Clinical outcomes in non-small-cell lung cancer patients receiving concurrent metformin and immune checkpoint inhibitors. Lung Cancer Manag. 2019 Oct;8(2):LMT11.
  65. 65. Libby G, Donnelly LA, Donnan PT, Alessi DR, Morris AD, Evans JMM. New Users of Metformin Are at Low Risk of Incident Cancer: A cohort study among people with type 2 diabetes. Diabetes Care. 2009 Sep 1;32(9):1620-1625.
  66. 66. DeCensi A, Puntoni M, Goodwin P, Cazzaniga M, Gennari A, Bonanni B, et al. Metformin and cancer risk in diabetic patients: A systematic review and meta-analysis. Cancer Prev Res. 2010;
  67. 67. Skinner HD, McCurdy MR, Echeverria AE, Lin SH, Welsh JW, O’Reilly MS, et al. Metformin use and improved response to therapy in esophageal adenocarcinoma. Acta Oncol (Madr). 2013 Jun 5;52(5):1002-1009.
  68. 68. Kim JM, Park JW, Lee JH, Park YH, Park SJ, Cheon JH, et al. Survival Benefit for Metformin Through Better Tumor Response by Neoadjuvant Concurrent Chemoradiotherapy in Rectal Cancer. Dis Colon rectum. 2020 Jun;63(6):758-768.
  69. 69. Sivalingam VN, Kitson S, McVey R, Roberts C, Pemberton P, Gilmour K, et al. Measuring the biological effect of presurgical metformin treatment in endometrial cancer. Br J Cancer. 2016 Feb 21;114(3):281-289.
  70. 70. Niraula S, Dowling RJO, Ennis M, Chang MC, Done SJ, Hood N, et al. Metformin in early breast cancer: a prospective window of opportunity neoadjuvant study. Breast Cancer Res Treat. 2012 Oct 30;135(3):821-830.
  71. 71. Jiralerspong S, Palla SL, Giordano SH, Meric-Bernstam F, Liedtke C, Barnett CM, et al. Metformin and pathologic complete responses to neoadjuvant chemotherapy in diabetic patients with breast cancer. J Clin Oncol. 2009;
  72. 72. Hu J, Chen JB, Cui Y, Zhu YW, Ren WB, Zhou X, et al. Association of metformin intake with bladder cancer risk and oncologic outcomes in type 2 diabetes mellitus patients: A systematic review and meta-analysis. Medicine (United States). 2018.
  73. 73. Zhang Z-J, Yuan J, Bi Y, Wang C, Liu Y. The effect of metformin on biomarkers and survivals for breast cancer- a systematic review and meta-analysis of randomized clinical trials. Pharmacol Res. 2019 Mar;141:551-555.
  74. 74. Niraula S, Dowling RJ, Enis M, Chang M, Done S, Hood N, et al. Metformin in early breast cancer (BC): A prospective, open-label, neoadjuvant “window of opportunity” study. J Clin Oncol. 2012 May 20;30(15_suppl):1019-1019.
  75. 75. Tang GH, Satkunam M, Pond GR, Steinberg GR, Blandino G, Schünemann HJ, et al. Association of Metformin with Breast Cancer Incidence and Mortality in Patients with Type II Diabetes: A GRADE-Assessed Systematic Review and Meta-analysis. Cancer Epidemiol Biomarkers Prev. 2018 Jun;27(6):627-635.
  76. 76. Col NF, Ochs L, Springmann V, Aragaki AK, Chlebowski RT. Metformin and breast cancer risk: a meta-analysis and critical literature review. Breast Cancer Res Treat. 2012 Oct 31;135(3):639-646.
  77. 77. Sonnenblick A, Agbor-Tarh D, Bradbury I, Di Cosimo S, Azim HA, Fumagalli D, et al. Impact of Diabetes, Insulin, and Metformin Use on the Outcome of Patients With Human Epidermal Growth Factor Receptor 2–Positive Primary Breast Cancer: Analysis From the ALTTO Phase III Randomized Trial. J Clin Oncol. 2017 May 1;35(13):1421-1429.
  78. 78. Ng C-AW, Jiang AA, Toh EMS, Ng CH, Ong ZH, Peng S, et al. Metformin and colorectal cancer: a systematic review, meta-analysis and meta-regression. Int J Colorectal Dis. 2020 Aug;35(8):1501-1512.
  79. 79. Meireles CG, Pereira SA, Valadares LP, Rêgo DF, Simeoni LA, Guerra ENS, et al. Effects of metformin on endometrial cancer: Systematic review and meta-analysis. Gynecol Oncol. 2017 Oct;147(1):167-180.
  80. 80. Chu D, Wu J, Wang K, Zhao M, Wang C, Li L, et al. Effect of metformin use on the risk and prognosis of endometrial cancer: a systematic review and meta-analysis. BMC Cancer. 2018 Dec 18;18(1):438.
  81. 81. Perez-Lopez FR, Pasupuleti V, Gianuzzi X, Palma-Ardiles G, Hernandez-Fernandez W, Hernandez A V. Systematic review and meta-analysis of the effect of metformin treatment on overall mortality rates in women with endometrial cancer and type 2 diabetes mellitus. Maturitas. 2017 Jul;101:6-11.
  82. 82. Guo J, Xu K, An M, Zhao Y. Metformin and endometrial cancer survival: a quantitative synthesis of observational studies. Oncotarget. 2017 Sep 12;8(39):66169-66177.
  83. 83. Yao L, Liu M, Huang Y, Wu K, Huang X, Zhao Y, et al. Metformin Use and Lung Cancer Risk in Diabetic Patients: A Systematic Review and Meta-Analysis. Dis Markers. 2019 Feb 10;2019:1-9.
  84. 84. Zhang J, Wu J, He Q, Liang W, He J. The prognostic value of metformin for advanced non-small cell lung cancer: a systematic review and meta-analysis. Transl Lung Cancer Res. 2018 Jun;7(3):389-396.
  85. 85. Wan G, Sun X, Li F, Wang X, Li C, Li H, et al. Survival Benefit of Metformin Adjuvant Treatment For Pancreatic Cancer Patients: a Systematic Review and Meta-Analysis. Cell Physiol Biochem. 2018;49(3):837-847.
  86. 86. E J-Y, Graber JM, Lu S-E, Lin Y, Lu-Yao G, Tan X-L. Effect of Metformin and Statin Use on Survival in Pancreatic Cancer Patients: a Systematic Literature Review and Meta-analysis. Curr Med Chem. 2018 Jul 4;25(22):2595-2607.
  87. 87. Ghiasi B, Sarokhani D, Najafi F, Motedayen M, Dehkordi AH. The Relationship Between Prostate Cancer and Metformin Consumption: A Systematic Review and Meta-analysis Study. Curr Pharm Des. 2019 Jul 9;25(9):1021-1029.
  88. 88. Hwang IC, Park SM, Shin D, Ahn HY, Rieken M, Shariat SF. Metformin Association with Lower Prostate Cancer Recurrence in Type 2 Diabetes: a Systematic Review and Meta-analysis. Asian Pacific J Cancer Prev. 2015 Feb 25;16(2):595-600.
  89. 89. Raval AD, Thakker D, Vyas A, Salkini M, Madhavan S, Sambamoorthi U. Impact of metformin on clinical outcomes among men with prostate cancer: a systematic review and meta-analysis. Prostate Cancer Prostatic Dis. 2015 Jun 10;18(2):110-121.
  90. 90. He K, Hu H, Ye S, Wang H, Cui R, Yi L. The effect of metformin therapy on incidence and prognosis in prostate cancer: A systematic review and meta-analysis. Sci Rep. 2019;
  91. 91. Shi J, Liu B, Wang H, Zhang T, Yang L. Association of metformin use with ovarian cancer incidence and prognosis: a systematic review and meta-analysis. Int J Gynecol Cancer. 2019 Jan 7;29(1):140-146.
  92. 92. Maraka S, Groves MD, Mammoser AG, Melguizo-Gavilanes I, Conrad CA, Tremont-Lukats IW, et al. Phase 1 lead-in to a phase 2 factorial study of temozolomide plus memantine, mefloquine, and metformin as postradiation adjuvant therapy for newly diagnosed glioblastoma. Cancer. 2019 Feb 25;125(3):424-433.
  93. 93. Molenaar RJ, Coelen RJS, Khurshed M, Roos E, Caan MWA, van Linde ME, et al. Study protocol of a phase IB/II clinical trial of metformin and chloroquine in patients with IDH1 -mutated or IDH2 -mutated solid tumours. BMJ Open. 2017 Jun 10;7(6):e014961.
  94. 94. Cho YY, Kang MJ, Kim SK, Jung JH, Hahm JR, Kim TH, et al. Protective Effect of Metformin Against Thyroid Cancer Development: A Population-Based Study in Korea. Thyroid. 2018 Jul;28(7):864-870.
  95. 95. Curry J, Johnson J, Tassone P, Vidal MD, Menezes DW, Sprandio J, et al. Metformin effects on head and neck squamous carcinoma microenvironment: Window of opportunity trial. Laryngoscope. 2017 Aug;127(8):1808-1815.
  96. 96. Singh AR, Gu JJ, Zhang Q, Torka P, Sundaram S, Mavis C, et al. Metformin sensitizes therapeutic agents and improves outcome in pre-clinical and clinical diffuse large B-cell lymphoma. Cancer Metab. 2020 Dec 6;8(1):10.
  97. 97. Wang GS, Hoyte C. Review of Biguanide (Metformin) Toxicity. J Intensive Care Med. 2019 Nov 21;34(11-12):863-876.
  98. 98. Bailey CJ, Turner RC. Metformin. Wood AJJ, editor. N Engl J Med. 1996 Feb 29;334(9):574-579.
  99. 99. Lin H-C, Kachingwe BH, Lin H-L, Cheng HW, Uang Y-S, Wang L-H. Effects of Metformin Dose on Cancer Risk Reduction in Patients with Type 2 Diabetes Mellitus: A 6-Year Follow-up Study. Pharmacother J Hum Pharmacol Drug Ther. 2014 Jan;34(1):36-45.
  100. 100. Dell’Aglio DM, Perino LJ, Kazzi Z, Abramson J, Schwartz MD, Morgan BW. Acute Metformin Overdose: Examining Serum pH, Lactate Level, and Metformin Concentrations in Survivors Versus Nonsurvivors: A Systematic Review of the Literature. Ann Emerg Med. 2009 Dec;54(6):818-823.

Written By

Raymond Chang

Submitted: 21 September 2020 Reviewed: 19 January 2021 Published: 05 February 2021