Open access peer-reviewed chapter

Preventive and (Neo)Adjuvant Therapeutic Effects of Metformin on Cancer

Written By

Yile Jiao, Xiaochen Wang and Zhijun Luo

Submitted: 26 November 2019 Reviewed: 21 January 2020 Published: 18 February 2020

DOI: 10.5772/intechopen.91291

From the Edited Volume

Metformin

Edited by Anca Mihaela Pantea Stoian and Manfredi Rizzo

Chapter metrics overview

918 Chapter Downloads

View Full Metrics

Abstract

Metformin, the first-line antidiabetic drug, has become an attractive candidate in cancer therapy since retrospective clinical investigations reported that patients with type 2 diabetes receiving metformin had lower incidence of cancer than those with other glucose lowering drugs. In line with this, preclinical studies have demonstrated that the antitumor activity of metformin could proceed through several mechanisms. Thus far, metformin has been used in cancer prevention with reduced risk as consequence and treatment of various cancers as an adjuvant or neoadjuvant drug. Thus, existing data support the beneficial effects of metformin on many types of cancers such as reducing metastasis and mortality and improving pathological responses and survival rates. However, some reports do not support this and even show adverse effects. The discrepancy may be attributed to expression levels of its transporters or genetic background. Hence, this chapter briefly reviews information on the mechanism of metformin action and summarizes both completed and ongoing clinical trials in an attempt to evaluate the value of metformin in prevention and treatment of various cancer types.

Keywords

  • metformin
  • AMPK
  • mTORC1
  • diabetes
  • lipogenesis
  • cancer prevention and therapy
  • clinical trials

1. Introduction

Metformin is derived from Galega officinalis, a natural herbal medicine. The herb was first used to relieve polyuria, a symptom of diabetes in ancient Egypt and medieval Europe [1]. Metformin is a widely used frontline drug for type 2 diabetes mellitus (T2DM). The major function of metformin is to decrease hepatic gluconeogenesis and enhance insulin sensitivity by increasing glucose uptake in muscle and adipose [2]. In addition to antidiabetes, metformin has proved to be beneficial to metabolic syndrome and nonalcoholic fatty liver disease [3, 4]. Cancer is characteristic of a metabolic disorder, inasmuch as metabolism is reprogrammed by switching oxidative phosphorylation into aerobic glycolysis, and thus, many of key molecules in these two routes are altered in their expression or posttranslational modification [5]. The incidence of cancer is higher in patients with T2DM than those without diabetes, indicating that diabetes is a risk factor of cancer [6]. Since Evan et al. reported in 2005 lower cancer incidence in patients with T2DM taking metformin than those with other antidiabetic drugs, great efforts have been made to elucidate the antitumor activity of metformin [7]. A considerable number of preclinical and clinical investigations support the beneficial effects of metformin on both prevention and treatment of various cancers. At the same time, some of mechanisms underlying metformin action on cancer cells have been unraveled, although much of them is still incomplete. Thus far, more than 300 clinical trials using metformin as a single or adjuvant agent in combination with other chemotherapies have been initiated in the treatment of various types of cancer in the world (www.clinicaltrials.gov).

Advertisement

2. Targets of metformin

Many functions of metformin are mediated by adenosine monophosphate-activated protein kinase (AMPK). Metformin at high doses leads to elevation of AMP, which binds to and allosterically activates AMPK, while at low doses, it engages lysosomes in the absence of AMP [8, 9]. The upstream kinases that phosphorylate AMPK α subunits at Thr172 include liver kinase B1 (LKB1), calmodulin-dependent kinase beta, and TGF-β-activated protein kinase [10, 11, 12].

AMPK plays important roles in regulating lipid and protein metabolisms by phosphorylating a series of target proteins. Thus, LKB1-AMPK pathway is critically important for metabolic adaption under stress condition, which aims to protect cells in the beginning [13]. However, persistent activation of AMPK by metformin can also cause cytostatic and even cytotoxic effects. Mounting evidence shows that metabolic syndrome and diabetes increase the risk of cancer, and correction of metabolic abnormalities alleviates cancer burdens and improves survival [14, 15, 16]. Drugs that target AMPK or downstream molecules are research focus nowadays for cancer prevention and treatment. Some of pathways downstream of AMPK essential for tumorigenesis and cancer progression are depicted in Figure 1.

Figure 1.

AMPK activation and its biological functions. AMPK is activated by increased AMP:ATP ratio induced by metabolic stress and metformin. In addition, metformin can activate AMPK through lysosomal pathway, where v-ATPase-regulator-AXIN/LKB1-AMPK complex is formed. After activation, AMPK acts on multiple molecules/pathways, including inhibition of mTORC1, lipogenesis and IGF-1 expression, and activation of p53 and FOXO3a [17, 22, 87, 88, 89]. As such, AMPK regulates cell proliferation, autophagy, and apoptosis of cancer cells.

PI3K-protein kinase B (AKT)-mammalian target of rapamycin (mTOR) pathway is well received as the target of AMPK. Mammalian target of rapamycin complex 1 (mTORC1) consists of mTOR, regulatory-associated protein of mTOR (Raptor), mammalian lethal with SEC13 protein 8, proline-rich AKT substrate 40 kDa, and DEP domain-containing mTOR-interacting protein [17]. Tuberous sclerosis complex 2 (TSC2) is a GTPase-activating protein that forms a complex with TSC1 to stimulate GTPase activity of Ras homolog enriched in brain (Rheb) and thus inhibits mammalian target of rapamycin complex 1 (mTORC1) activation. TSC2 is subjected to inhibition by AKT and activation by AMPK via phosphorylation at different sites. In addition, AMPK phosphorylates and inhibits Raptor, a scaffold of mTORC1. A plethora of cellular events, such as protein translation, lipogenesis, cell cycle progression, and autophagy, are regulated by the activated mTOR pathway, which are counteracted by AMPK [18]. Thus, control of mTORC1 activity is crucial for prevention and treatment of cancer.

Cancer cells always require large amount of building blocks for dividing progenitor cells. Thus, synthesis of fatty acid and cholesterol is very active [19]. Acutely, AMPK inhibits acetyl CoA carboxylase (ACC) and HMG-CoA reductase (HMGCR), which are rate-limiting enzymes for de novo synthesis of fatty acid and cholesterol, respectively [20]. In addition, AMPK activates malonyl-CoA decarboxylase (MAD) that converts malonyl-CoA to acetyl CoA. As cytosolic malonyl-CoA decreases, fatty acid synthesis is attenuated [17, 21]. AMPK also influences de novo synthesis of glycerolipid by inhibiting the rate-limiting enzyme glycerol phosphate acyltransferase (PAT) [17, 22]. Chronically, AMPK phosphorylates sterol regulatory element-binding protein-1c (SREBP-1c) and its related protein carbohydrate-response-element-binding protein (ChREBP), restricting the nuclear localization of these transcription factors, so as to inhibit transcription of target genes for lipogenesis, including those encoding ACC and fatty acid synthase (FASN) [23].

Advertisement

3. Clinical investigations

Decreases from 20 to 94% in cancer risk among patients with T2DM after the use of metformin have been reported since 2005 [24]. A large population study conducted by Taiwan National Health Insurance Data Survey evaluated 16,602 individuals treated with metformin or other antidiabetic drug between 2000 and 2007 and concluded a 88% reduction in the risk of various cancer types after metformin treatment [25, 26]. In line with this, numerous investigations provided supporting data that metformin reduced incidence of various cancers. For example, DeCenci et al. have found a 30% decrease in cancer incidence in patients with T2DM treated with metformin compared to those with other drugs [27, 28]. Currie et al. conducted a large cohort study with around 60,000 patients from the UK database and revealed that metformin alone decreased the incidence of colorectal and pancreatic cancer compared with insulin and sulfonylureas monotherapy after the adjustment of confound bias, but this was not seen in breast cancer (BC) and prostate cancer [29]. It is noteworthy that metformin plus insulin could alleviate the progression of cancer [hazard ratio (HR) = 0.54, 95% confidence interval (CI) 0.43–0.66] [29]. With respect to mortality, ZODIAC trial with a 10-year follow up has indicated a lower death rate of cancer among metformin users with T2DM [30]. According to Noto et al. meta-analysis, diabetic patients taking metformin showed significant reduction of incidence of multiple types of cancer [risk ratio (RR) = 0.67, 0.53–0.85], including colorectal cancer (CRC) (RR = 0.68, 0.53–0.88) and cancer mortality (RR = 0.66, 95% CI = 0.49–0.88) [31]. A study of Bowker et al. reported that metformin decreased cancer mortality in T2DM, as compared with insulin and/or sulfonylurea groups [32]. After 1-year observation, the cancer death rate of metformin, insulin, and sulfonylurea users is 3.5, 8.8, and 4.9 per 1000 patients, respectively.

Regarding tumor types, dosage of metformin, study setting, and period of intervention associated with the treatment outcomes, examples are listed in Table 1.

Cancer typeInterventionOutcome
Breast cancer
Bodmer et al. [39]Metformin or other antidiabetic drugsDiabetic patients treated with metformin ≧ 5 years had a lower incidence of cancer, compared with nonusers or short-term (<5 years treatment) metformin users
Jiralerspong et al. [45]Metformin + chemotherapyThe pCR rate in 68 diabetic patients treated with metformin, 87 diabetic patients without metformin, and 2374 nondiabetic patients was 24, 8, and 16% (P = 0.02)
Niraula et al. [46]MetforminReduction of cancer cell proliferation (Ki67) by 3% (P = 0.016) and increases in apoptosis by 0.49% (P = 0.004) was compared between pre- and post-surgery, despite minor change of fasting insulin level
Hou et al. [51]Metformin + chemotherapy1013 BC patients with diabetes and 4621 BC patients without diabetes were analyzed. Nondiabetic group had higher 5-year survival rate than diabetic group (82 vs. 79%, P < 0.001). In diabetic subgroup, metformin-treated group had significant higher 5-year survival rates than nonmetformin-treated group (88 vs. 73%, P< 0.001)
El-Haggar et al. [42]Metformin + chemotherapy or +hormone therapy, tamoxifenNon-diabetic women with newly diagnosed BC (68/129) were prescribed with metformin (860 mg b.i.d.) along with chemotherapy or hormone therapy compared to nonmetformin-treated control arm over 6 or 12 months. A 3.27-fold decrease (P = 0.023, 95% CI 1.17–9.06) at the time of developing metastasis and an increase in average DFS by 2.137 (P = 0.044) in the metformin-treated group. Also, the levels of IGF-1, the ratio of IGF-1 to IGFBP-3, insulin, fasting blood glucose, HOMA-IR index notably decease, while IGFBP-3 levels significantly increase after using of metformin
He et al. [53]Metformin or other antidiabetic drugsA cohort study evaluated a total of 1983 women with stage ≧ 2 Her2 positive BC. Among 154/1983 diabetic patients who had already responded to previous chemotherapy. Metformin users had prolonged OS (HR = 0.52, 95% CI 0.28–0.97, P = 0.041) and reduced cancer-specific mortality of BC (HR = 0.47, 95% CI 0.24–0.90, P = 0.023), compared with nonusers
Colon cancer
Coyle et al. [33]MetforminSignificant benefit of RFS (n = 623 patients in two studies), OS (n = 1936 patients in five studies), and CSS (n = 535 patients in two studies) was observed in metformin-treated patients from 3094 patients with early stage CRC in nine studies, compared with that in nonmetformin using group
Rokkas and Portincasa [55]MetforminA significant decrease in the risk of developing colon neoplasia [RRs (95% CI) = 0.75 (0.65–0.87), Z = −3.95, P < 0.001], including the reduction of colon cancer [0.79 (0.69–0.91), Z = −3.34, P < 0.001] and colon polyps [0.58 (0.42–0.80), Z = −3.30, P < 0.001] among patients with T2DB after metformin treatment
Garrett et al. [58]Metformin or other antidiabetic drugsAfter adjustment of cofound variates, a 30% increase in OS was demonstrated among 424/4758 patients who were diagnosed of T2DM and CRC and administrated to metformin as compared with that in other antidiabetics users
Higurashi et al. [59]MetforminA total of 151 nondiabetic patients with CRC after polypectomy was randomized to metformin-treated arm (250 mg daily over 1 year) or placebo control arm with 1-year endoscopy reports. The incidence of total polyps and adenomas decreased in metformin-treated group by 18.5% [RR = 0.67, 95% CI (0.47–0.97), P = 0.034] and 21% [RR = 0.60, 95% CI (0.39–0.92), P = 0.16], compared with that in control group
Endometrial cancer
Sivalingam et al. [60]MetforminA total of 40 women with atypical endometrial hyperplasia (AEH) or EC was assigned to receive metformin 850 mg b.i.d. over average 20 day, or no treatment before hysterectomy. Ki67 was reduced by 17.2% (95% CI 27.4–7.0, P < 0.002) in metformin-treated group
Schuler et al. [61]Metformin20 nondiabetic women with EC and obesity (BMI ≥ 30) were administrated with metformin 850 mg daily for 1–4 weeks before surgery. The levels of Ki67 and p-S6 were reduced between pretreatment and postsurgery by 11.75% (P = 0.008) and 51.2% (P = 0.0002), respectively. Besides, the levels of p-AMPK (P = 0.00001), p-Akt (P = 0.0002), p-4EBP1 (P = 0.001), and ER (P = 0.0002) also decreased after surgery
Mitsuhashi et al. [63]Metformin + MPA17 AEH and 19 noninvasive EC patients received metformin (escalating from 750 to 2250 mg daily) after complete response treated by MPA and other drugs. Relapse rate among patients was 10%, and estimated 3-year RFS rate was 89%
Nevadunsky et al. [66]Adjuvant metforminMetformin significantly improved OS (HR = 0.54, 95% CI 0.30–0.97, P < 0.04) in diabetic patients with nonendometrioid EC when compared with that in nonusers with EC
Acute lymphoblastic leukemia
Ramos-Peñafiel et al. [67]Metformin + prednisoneA total of 102 nondiabetic patients with ALL was enrolled, 26 received metformin (850 mg t.i.d.) for 6 days during preinduction stage, and 76 were treated with traditional chemotherapy without metformin. The use of metformin prevented therapy failure and early relapse (P = 0.025) in patients bearing relative to high levels of ABCB1
Esophageal Cancer
Skinner et al. [68]Neoadjunvant metformin + CRTMetformin users along with CRT resulted in higher pCR (34.5%) than nonmetformin cohort (4.8%, P = 0.01) and nondiabetic patients (19.6%, P = 0.05). Higher pCR rate was found to be associated with higher metformin dose (≥1500 mg/d). Post-CRT maximum SUV decreased significantly in patients taking metformin (P = 0.05)
Lee et al. [25]Adjuvant metforminReduction of total CID and incidence of some gastroenterological cancers including CRC, HCC, and so on, among which the CID of esophageal cancer decreased in diabetic groups taking adjuvant metformin in comparison to non-DM groups. Metformin dosage giving rise to a significant decrease in cancer incidence was ≤500 mg/day
Leamm et al. [69]Metformin + neoadjuvant chemo(radio)therapyNo statistically significant difference between metformin users and nonmetformin users for median overall survival (43.6 vs. 42.8 months, P = 0.66) or for median DFS (31.1 vs.47.0 months, P = 0.68)
Prostate cancer
Wright et al. [70]MetforminA reduced risk of prostate cancer was showed among white men at age of 35–74 after the use of metformin, as reported by a case-control study
Rothermundt et al. [74]MetforminA total of 44 men with castration-resistant prostate cancer was assigned to receive metformin 500 mg b.i.d. until progression. After initial metformin treatment, changes in IGF and IGBP3 and improvement of insulin sensitivity from baseline were observed but without correlation with progression. At week 4, only four patients did not have progression (95% CI, 3–22). Average PFS was 2.8 months (95% CI, 2.8–3.2) and PSA double time declined in 23 patients but not significant
Joshua et al. [75]MetforminMetformin 500 mg t.i.d. was prescribed to 24 men with operable prostate cancer before prostatectomy. In a per patient and per tumor analyses, Ki67 was reduced by 29.5% (P = 0.0064) and 28.6% (P = 0.0042) in comparison with the initial biopsy and postprostatectomy sections
Rieken et al. [77]MetforminMetformin users with prostate cancer exhibited a minor improvement of RFS after prostatectomy
Spratt et al. [78]MetforminA retrospective study examined 2901 noninvasive prostate cancer patients through radiation therapy. In 157 patients treated with metformin, PSA-RFS and DMFS were improved and the castration-resistant prostate cancer progression was alleviated

Table 1.

Examples of clinical investigations of metformin used as a neoadjuvant and adjuvant agent in cancer therapy.

3.1 The role of metformin in radiotherapy and chemotherapy

Metformin has been reported to be a useful adjuvant drug to radiotherapy or chemotherapy for different cancers, especially prostate and colon cancers [33]. The effects of metformin on overall survival (OS), relapse-free survival (RFS), and cancer-specific survival (CSS) after concurrent chemotherapy and/or radiotherapy vary on cancer types.

A previous study has shown that metformin increases radiosensitivity of luminal BC by influencing expression of thioredoxin and intracellular redox homeostasis [34]. A high level of AMPKα expression correlates with the increased radiosensitivity and better prognosis. A systemic review and meta-analysis conducted in 2018 summarized the impact of metformin on the efficacy of radiotherapy in 17 studies, including prostate cancer, head and neck cancer, rectal cancer, lung cancer, esophageal cancer, and liver cancer [35]. The study compared diabetic patients with metformin (D + M) and diabetic or nondiabetic cohort without metformin (D − M or N − M) after radiotherapy. An improved pathologic complete response (pCR), 2y-OS, and 5y-OS vary in different cancer types when analyzing D + M and D − M groups, supporting that metformin is beneficial to OS of diabetic patients while distant metastasis-free survival (DMFS) and 5-year OS were not significantly different between D + M and N − M groups. With respect to the possible mechanisms by which metformin enhances radiosensitivity, studies have indicated that p53 and AMPKα are involved [36, 37]. Despite the increased sensitivity to radiotherapy and chemotherapy, cumulative side effects and toxicity concur with the use of metformin. For example, a study has shown that combination of metformin with radiochemotherapy can lead to less tolerance to cisplatin and radiotherapy and exacerbate gastrointestinal adverse effects such as grade ≥ 3 nausea/vomiting [38].

3.2 Breast cancer

Several lines of clinical investigations have been conducted to assess the beneficial effects of metformin on BC [39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52]. Two retrospective studies revealed that long-term use of metformin (>5 years) reduced the risk of BC in T2DM women as compared with other antidiabetic drugs [39, 40]. However, Currie et al. reported that metformin use did not affect risk of breast and prostate cancer, but the reduced risk was found in colon and pancreas cancer [29].

He et al. have shown improvement of disease-free survival (DFS), DMFS, and OS in diabetic women who well-responded to previous hormone therapy and then received metformin treatment. The results demonstrated that metformin synergizes with hormone therapy [53, 54].

Metformin was used as neoadjuvant chemotherapy of BC to improve pathological conditions prior to surgery [45, 46, 47, 48]. The increased pCR in 2529 women with BC has been demonstrated in metformin-treated diabetic patients, compared to nonmetformin-treated patients with or without diabetes [45]. Another study by Niraula et al. evaluated the effect of metformin on serum biomarkers in nondiabetic BC patients before surgery [46]. The patients were treated with metformin for 2 weeks, and serum biomarkers were assessed. A notably reduction of Ki67 and elevation of apoptosis were observed in invasive tumor after the use of metformin. The significant decrease of homeostatic model assessment of insulin resistance (HOMA-IR) was also observed, while insulin and leptin displayed a modest change. However, a study showed that metformin increased phospho-AMPK (p-AMPK) and decreased p-Akt and Ki67 without induction of apoptosis, suggesting a cytostatic effect [47].

The long-term use of metformin has been shown to reduce risk of distant metastasis and mortality of BC patients with type 2 diabetes [49, 50, 51]. Furthermore, metformin use as adjuvant therapy can also improve outcomes of BC in nondiabetic patients [41, 42, 52]. For example, a single-arm phase II trial enrolled nondiabetic women with M0 stage BC. After receiving metformin of 500 mg t.i.d. for 6 months, the result showed that fasting insulin level and HOMA-IR were significantly reduced. Total cholesterol, low density lipoprotein, and leptin also similarly declined [52]. Another study focused on the optimal dose of metformin that achieves favorable effects on BC by comparing dose between 1500 and 1000 mg daily [41]. For postmenopausal women with basal testosterone levels≧0.28 ng/mL, it seemed that metformin of 1500 mg/d was better than 1000 mg/d in reduction of insulin and testosterone levels, which were associated with cancer incidence and prognosis. Combination of metformin with other chemotherapy usually generates better outcomes in nondiabetic BC patients with the higher HOMA-IR (>2.8), and HOMA-IR can be improved by metformin [42, 43, 44, 48].

In summary, studies showing beneficial effects of metformin are more than those without effects. Metformin as an adjuvant agent can suppress BC at various doses ranging from 500 to 1500 mg. The outcomes mainly include reduced risk of BC, decreases in cancer-promoting markers and metastatic events, increases in apoptotic markers, and improvement of progression-free survival (PFS) and OS.

3.3 Colon cancer

The role of metformin in preventing colon cancer has been documented in the following studies conducted in both diabetic and nondiabetic patients. A meta-analysis was carried out in 709,980 individuals with T2DM from 17 studies showing a significant decrease in the risk of colon neoplasia among metformin-treated patients compared to those without metformin, with respective reduction for either cancer or polys [55]. A randomized study enrolled a total of 26 nondiabetic individuals with aberrant crypt foci (ACF) (biomarker of CRC development) and assigned them to either receive metformin 250 mg daily for 1 month or control group [56]. Significant decreases in the average number of ACF by a 3.67-fold (P = 0.007) and in proliferating cell nuclear antigen index were discovered in metformin arm. This indicates that metformin prevents CRC by attenuating cell proliferation and ACF development.

Metformin has been used as an adjuvant agent in the treatment of CRC. First, a single-arm study has demonstrated a median PFS of 1.8 months and an OS of 7.9 months in metastatic CRC with combination of metformin (850 mg b.i.d.) and 5-fluorouracil treatment. Surprisingly, the improvement in median survival was more obvious in obese patients [57]. Second, Coyle et al. have evaluated 3092 patients with early stage of CRC [33]. It was found that the use of metformin significantly improved RFS (HR = 0.63, 95% CI 0.47–0.85), OS (0.69, 95% CI 0.58–0.83), and CSS (0.58, 95% CI 0.39–0.86) in patients with T2DM, compared with other antidiabetic drugs. Likewise, progression of CRC is also inhibited by metformin. A similar study showed prolonged OS in patients with T2DM with CRC receiving metformin, as compared with nonmetformin users (79.6 vs. 56.9 months, P = 0.048) [58]. The last randomized trial used metformin (250 mg daily) for a year in nondiabetic patients with high-risk adenoma recurrence and no colorectal polyps after polypectomy [59]. The results showed that polyps and adenomas are noticeably fewer in the metformin arm than in the control arm. The study also showed that average HOMA-IR status was significantly reduced in nonrecurrent patients by metformin, while the value remained stable in recurrent patients, indicating that insulin resistance is associated with chemoprevention outcome.

3.4 Endometrial cancer

Clinical investigations support that metformin could serve as a potential drug for protection against endometrial cancer (EC) [60, 61, 62, 63, 64, 65]. Several studies have evaluated the effects of short-term use of metformin as a neoadjuvant therapy between initial recruitment and hysterectomy surgery in nondiabetic women with EC [60, 61, 62]. The first nonrandomized trial has examined the change of Ki67 and shown a remarkable reduction after metformin use at 850 mg b.i.d. for average 20 days [60]. A significant reduction in phospho-4E-binding protein 1 (p-4EFBP1) downstream of mTOR was also observed by immunohistochemistry, while indirect serum markers of insulin resistance (fasting glucose, insulin, and HOMA-IR) and leptin only showed a decrease trend but not significant after adjusting difference. Another preoperative clinical trial was done in nondiabetic women with body mass index (BMI) ≧ 30 [61]. After taking metformin 850 mg daily for 1–4 weeks prior to surgery, Ki67, p-AMPK, p-Akt, phospho-S6 Ribosomal Protein (p-S6), and p-4EBP were significantly lower in resected specimens than in pretreatment. The reduction of p-AMPK is inconsistent with purported positive effect of metformin. This study also showed a decrease in estrogen receptor (ER) but not progesterone receptor.

According to a study evaluating the effect of metformin on EC of diabetic patients (n = 114) as compared with diabetic (n = 136) and nondiabetic (n = 735) patients without metformin from 1999 to 2009, metformin-treated group exhibits prolonged OS than nonusers before and after the adjustment of confound bias [66]. A phase II study has examined the effects of long-term metformin (2250 mg daily until recurrence) on RFS after a complete response to medroxyprogesterone acetate (MPA) in 17 individuals with atypical endometrial hyperplasia and 19 with EC [63]. The 3-year estimated RFS was 89%, and the 3-year recurrence rate showed a 4.7-fold decrease in this study compared with a previous study [64]. In contrast to short-term treatment, the other randomized factorial study does not have a significant change in PFS/OS after metformin treatment (1700 mg/d for 16 weeks and 1-year follow up) [65].

3.5 Acute lymphoid leukemia

A single study randomized to assign 102 patients with nondiabetic acute lymphoid leukemia (ALL) into a group of 26 with metformin at 850 mg t.i.d. for 10 days and the rest to the group without metformin before remission therapy [67]. Metformin displayed a beneficial effect on OS in the patients with high levels of ABCB1 expression, the gene encoding multidrug resistant protein-1. The failure rate of therapy was significantly reduced and early relapse after remission prevented by metformin, as compared with nonusers.

3.6 Oesophagal cancer

Oesophagal cancer is deadly cancer with poor prognosis, and patients usually do survive or die no longer than 30 months after chemoradiation and surgery [68]. A prospective cohort study by Taiwan National Health Insurance revealed a positive effect of metformin as an adjunct to standard chemotherapies on the cancer incidence density (CID) of gastroenterological cancers [25]. In this study, a decrease in total CID including esophageal cancer was found in diabetic groups taking adjuvant metformin in comparison to nondiabetic groups. Another study reported that metformin enhanced the efficacy of radiochemotherapy in patients with T2DM resulting in superior pCR and low postconcurrent chemoradiation (CRT) maximum SUV compared to patients with T2DM without metformin and non-DM patients [68]. Additionally, higher pCR rate was correlated with higher metformin dose (≥1500 mg/d). However, a report in 2015 demonstrated inconsistent results, in which no difference in pCR was found between metformin users and nonmetformin users [69]. Furthermore, it was shown that together with neoadjunvant chemoradiation, metformin did not improve the median OS or median DFS in diabetic patients with esophageal cancer.

3.7 Prostate cancer

The effect of metformin on prostate cancer is ambiguous. Studies of Wright and Stanford have provided a 44% decrease in the risk of prostate cancer among Caucasian men with diabetes [70]. However, investigations by others could not obtain the same conclusion on the incidence of prostate cancer in diabetic patients treated with metformin, but the mortality might be reduced [71, 72, 73]. A single-arm clinical trial has revealed a decrease in insulin-like growth factor-1 (IGF-1) and an increase in insulin-like growth factor-binding protein-3 (IGFBP-3), alongside lowering prostate-specific antigen (PSA), after giving metformin 500 mg b.i.d. over 12 weeks to patients with castration-resistant prostate cancer [74]. In a single-arm study on men with biopsy-proven localized prostate cancer, 22 patients were selected to receive metformin at 500 mg/d or b.i.d., followed by t.i.d. for 28–84 days preceding their prostatectomy. The results revealed that Ki67 index was reduced by comparing the initial biopsy with postprostatectomy sections [75]. However, the changes were not recapitulated by another study, although metformin in the prostate tissue was detected after a median of 34 days prior to prostatectomy [76]. In a retrospective study, metformin-treated diabetic individuals gained the improvement of RFS among 6863 patients after radical prostatectomy [77]. Study of Spratt et al. also demonstrated the significantly elevated PSA-RFS, DFS, and lower cancer mortality in localized prostate cancer with metformin treatment compared with that of nonusers [78].

Advertisement

4. Ongoing clinical trials

Previous studies of metformin use as neoadjuvant or adjuvant therapy for various types of cancer provide strong rationale of clinical trials in more vigorous settings. Thus far, more than 300 clinical trials have initiated in the world despite some are somehow either terminated or withdrawn. Table 2 lists some of them. For example, NCT02065687 is a randomized, metformin-placebo, phase II/III study that enrolls a total of 540 participants and examines the effect of adjuvant metformin together with paclitaxel and carboplatin in treatment of stages III–IV or recurrent EC. Patients receive metformin twice a day in a 5-year follow up until disease progression or undesirable adverse effects appear. According to this trial, prolonged PFS and OS will be observed after the use of metformin together with other chemotherapeutic drugs. One of the ongoing phase II trials carrying out in 151 premenopausal BC patients with BMI ≧ 25 kg/m2 evaluates treatment effect with 850 mg metformin b.i.d. vs. placebo for a year, by examining the primary outcome changes of breast density at time points of 6 and 12 months. This study spanning from March 7, 2014 to June 30, 2020 also identifies biomarkers associated with metabolic effects of metformin and attempts to find prediction factors of BC risk (NCT02028221). Also, a trial (NCT02614339) is undergoing to follow-up 3-year DFS and 5-year OS in nondiabetic patients with stage II high-risk/III CRC treated with metformin (1000 mg/day) for 48 months. This study has enrolled 593 participants and is still recruiting and expected to complete in July 2021.

NCT numberStatusParticipantsPeriodInterventionCancer type
NCT02581137
https://ClinicalTrials.gov/show/NCT02581137
Active(a) 26, (b)18 years and older (adult, older adult), (c) all sexJune 10, 2016 to not indicatedDrug: metformin hydrochlorideOral cancer
NCT02028221
https://ClinicalTrials.gov/show/NCT02028221
Active(a) 151, (b) 21 years to 54 years (adult), (c) femaleMarch 7, 2014 to June 30, 2020Drug: metformin & placeboBC
NCT02431676
https://ClinicalTrials.gov/show/NCT02431676
Active(a) 100, (b) 50 years to 65 years (adult, older adult), (c) femaleMay 1, 2013 to September 1, 2022Drug: metformin & placeboEC
NCT01697566
https://ClinicalTrials.gov/show/NCT01697566
Active(a) 100, (b) 50 years to 65 years (adult, older adult), (c) femaleMay 1, 2013 to September 1, 2022Drug: metformin & placeboEC
NCT01797523
https://ClinicalTrials.gov/show/NCT01797523
Active(a) 62, (b) 18 years and older (adult, older adult), (c) all sexMay 1, 2013 to October 1, 2020Drug: metformin, letrozole, & everolimusEC
NCT02065687
https://ClinicalTrials.gov/show/NCT02065687
Active(a) 540, (b) 18 years to older (adult, older adult), (c) femaleMatch 17, 2014 toDrug: carboplatin, metformin hydrochloride, paclitaxel, & placeboEC
NCT03047837
https://ClinicalTrials.gov/show/NCT03047837
Recruiting(a) 160, (b) 18 years to 80 years (adult, older adult), (c) all sexMarch 15, 2017 to March 15, 2020Drug: aspirin (ASA) + metformin (MET)|Drug: ASA|Drug: MET|Drug: placebosTertiary prevention in colon cancer
NCT01905046
https://ClinicalTrials.gov/show/NCT01905046
Recruiting(a) 128, (b) 25 years to 55 years (adult), (c) femaleAugust 2013 toDrug: metformin hydrochloride & placeboBC
NCT02614339
https://ClinicalTrials.gov/show/NCT02614339
Recruiting(a) 593, (b) 20 years to 80 years (adult, older adult), (c) all sexDecember 2015 to July 2021Drug: metformin & placeboCRC
NCT03378297
https://ClinicalTrials.gov/show/NCT03378297
Recruiting(a) 143, (b) 18 years and older (adult, older adult), (c) femaleMay 4, 2018 to June 1, 2020Drug: metformin & acetylsalicylic acid & drug: olaparib & drug: letrozoleOvarian cancer
NCT03685409
https://ClinicalTrials.gov/show/NCT03685409
Recruiting(a) 62, (b)20 years to 70 years (adult, older adult), (c) all sexOctober 1, 2018 to September 30, 2020Drug: metformin hydrochloride & placeboOral cancer
NCT01864096
https://ClinicalTrials.gov/show/NCT01864096
Recruiting(a) 408, (b) 18 years to 79 years (adult, older adult), (c) maleOctober 1, 2013 to August 1, 2024Drug: metformin & placeboProstate cancer

Table 2.

Summary of ongoing clinical trials approved by FDA.

The trial of double-blinded 2 × 2 factorial (aspirin × metformin) design registers 160 patients with stages I–III colon cancer who undertake a completed polypectomy within recent 24 months (NCT03047837). After randomized allocation, patients will receive metformin at 850 mg b.i.d. or aspirin at 100 mg daily or two drugs together vs. placebo over 1 year. Immunohistochemistry for NF-κB, glucose metabolism, pS6K, and other biomarker will be compared pre- and postintervention (ClinicalTrials.gov Identifier: NCT03047837).

Advertisement

5. Cautions to be considered

5.1 Cancer type-specific effects

Whether a cancer type is sensitive to metformin depends on expression level of OCT1 in the cell membrane. Thus far, majority of previous studies have demonstrated that metformin exerts beneficial effects on different types of cancer, while some do not respond. On contrary, in some cases, for example, in glioma and leukemia cancer cells, metformin reduces cisplatin-induced apoptosis, suggesting that metformin exerts a protective effect on cytotoxic agents in some cells [79]. Hence, before going to clinical trials, preclinical tests should be undertaken to ascertain if metformin enhances the inhibitory effect of other drugs. This is feasible when PDX animal models or organoid culture techniques are available.

5.2 Genetic background of cancer

Responses of cancer cells with and without LKB1 to metformin are different. Metformin exerts cytostatic effect on cancer cells with wild-type LKB1, while it causes cytotoxicity in cells lacking LKB1. If metformin is used together with most of chemotherapeutic drugs that are cytotoxic in cancer containing wild-type LKB1, the cooperative effects might not be achieved. The reason is that more rapidly dividing cells are more sensitive to cytotoxic drugs, while cytostatic drugs slow down speed of cell growth, which might compromise the efficacy of cytotoxic chemotherapy. In this scenario, it might be a good idea to take metformin and cytotoxic drug alternately. For example, patients take a couple of cycles of cytotoxic chemotherapy and then have rest for period of time during which metformin is alternately used. The purpose is to restrain cancer in dormancy and allow the patients to restore healthy condition. In addition, Birsoy et al. have delineated that the most metformin-sensitive cells contain mutations of genes responsible for upregulation of mitochondrial oxidative phosphorylation, for example, complex I components, or glucose utilization [80]. Thus, these genes may serve as biomarkers for metformin use. Altogether, these studies point to importance of personalized medicine to determine the efficacy of metformin in cancer therapy.

5.3 Sensitivity of cancer stem cells

Cancer stem cells (CSCs) are refractory to chemotherapy, leading to the relapse of cancer. These cells metastasize to distant organs after flowing in circulation, resulting in poor prognosis. Thus, CSCs have become an important target for anticancer therapies. Hirsch et al. have reported that the CSCs derived from BC are preferentially sensitive to metformin that is used from 10 to 100 times less dosage than nonstem cancer cells [81]. This finding suggests that metformin could effectively prevent metastasis. It is especially meaningful in the case of surgically resected cancer when local metastasis in lymph nodes is cytologically tested negative, but a few CSCs may escape to circulation. At this time, metformin can be used as preventive measure.

Previous studies have demonstrated that metformin selectively targets CSCs via regulation of different pathways in various cancer types including breast, pancreatic, prostate, and colon cancer [82, 83]. For example, Zhu et al. have shown that metformin inhibits CD61high/CD49fhigh subpopulation, markers of tumor initiating cells, by inactivating epidermal growth factor receptor/ErbB2 signaling. Similarly, CD133+, aldehyde dehydrogenases 1+, and other molecules are inhibited in pancreatic and colon cancer through inhibition of the Akt/mTOR pathway [84, 85]. However, a recent study using head and neck squamous cell carcinoma has shown that metformin protects CSCs against the cisplatin-induced cell death when combining these two, which discord with previous studies [86]. Thus, it should be cautious to ascertain if metformin exerts inhibitory or protective effects on specifically originated CSCs.

Advertisement

6. Conclusion

Metformin is a cheap and nontoxic first-line antidiabetic medicine. It is an attractive drug that is being repurposed for multiple usages in treatment of other diseases in addition to diabetes. Metformin implements its function through AMPK-dependent and independent mechanisms. Preclinical and retrospective clinical investigations have inspired clinical trials of metformin use in various cancer therapies. It is a promising drug in neoadjuvant and adjuvant therapies. We hope these trials will come to end with positive or negative results in the next few years. In considering genetic heterogeneity of cancer, responses of different cancer types and subpopulations in the same cancer might be different. Therefore, we still have long way to go and loads of questions to be addressed.

Advertisement

Acknowledgments

ZL is supported by the National Natural Science Foundation of China (81572753, 31660332) and the Innovation and Entrepreneurship grant from Jiangxi Provincial Bureau of Foreign Experts, China.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Rena G, Hardie DG, Pearson ER. The mechanisms of action of metformin. Diabetologia. 2017;60(9):1577-1585. Available from: http://www.ncbi.nlm.nih.gov/pubmed/28776086
  2. 2. Zhou M, Xia L, Wang J. Metformin transport by a newly cloned proton-stimulated organic cation transporter (plasma membrane monoamine transporter) expressed in human intestine. Drug Metabolism & Disposition. 2007;35(10):1956-1962. Available from: http://www.ncbi.nlm.nih.gov/pubmed/27011019
  3. 3. Mazza A, Fruci B, Garinis GA, Giuliano S, Malaguarnera R, Belfiore A. The role of metformin in the management of NAFLD. Experimental Diabetes Research. 2012;2012:1-13. Available from: http://www.hindawi.com/journals/jdr/2012/716404/
  4. 4. Ghatak SB, Dhamecha PS, Bhadada SV, Panchal SJ. Investigation of the potential effects of metformin on atherothrombotic risk factors in hyperlipidemic rats. European Journal of Pharmacology. 2011;659(2–3):213-223. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0014299911003414
  5. 5. Seyfried TN, Shelton LM. Cancer as a metabolic disease. Nutrition & Metabolism. 2010;7(1):7. Available from: http://nutritionandmetabolism.biomedcentral.com/articles/10.1186/1743-7075-7-7
  6. 6. Kasznicki J, Sliwinska A, Drzewoski J. Metformin in cancer prevention and therapy. Annals of Translational Medicine. 2014;2(6):1-11
  7. 7. Ikhlas S, Ahmad M. Metformin: Insights into its anticancer potential with special reference to AMPK dependent and independent pathways. Life Sciences. 2017;185:53-62. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0024320517303685
  8. 8. Zhang C-S, Hawley SA, Zong Y, Li M, Wang Z, Gray A, et al. Fructose-1,6-bisphosphate and aldolase mediate glucose sensing by AMPK. Nature. 2017;548(7665):112-116. Available from: http://www.nature.com/articles/nature23275
  9. 9. Zhang C-S, Li M, Ma T, Zong Y, Cui J, Feng J-W, et al. Metformin activates AMPK through the lysosomal pathway. Cell Metabolism. 2016;24(4):521-522. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1550413116304818
  10. 10. Foretz M, Guigas B, Bertrand L, Pollak M, Viollet B. Metformin: From mechanisms of action to therapies. Cell Metabolism. 2014;20(6):953-966. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1550413114004410
  11. 11. Muaddi H, Chowdhury S, Vellanki R, Zamiara P, Koritzinsky M. Contributions of AMPK and p53 dependent signaling to radiation response in the presence of metformin. Radiotherapy and Oncology. 2013;108(3):446-450. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0167814013002922
  12. 12. Pollak MN. Investigating metformin for cancer prevention and treatment: The end of the beginning. Cancer Discovery. 2012;2(9):778-790. Available from: http://cancerdiscovery.aacrjournals.org/lookup/doi/10.1158/2159-8290.CD-12-0263
  13. 13. Jeon S-M, Chandel NS, Hay N. AMPK regulates NADPH homeostasis to promote tumour cell survival during energy stress. Nature. 2012;485(7400):661-665. Available from: http://www.nature.com/articles/nature11066
  14. 14. LeRoith D, Novosyadlyy R, Gallagher E, Lann D, Vijayakumar A, Yakar S. Obesity and type 2 diabetes are associated with an increased risk of developing cancer and a worse prognosis; epidemiological and mechanistic evidence. Experimental and Clinical Endocrinology and Diabetes. 2008;116(S 01):S4-S6. Available from: http://www.thieme-connect.de/DOI/DOI?10.1055/s-2008-1081488
  15. 15. Nawroth P. Diabetes, obesity, insulin resistance: Different pathways to cancer? Experimental and Clinical Endocrinology & Diabetes. 2009;117(10):561-562. Available from: http://www.thieme-connect.de/DOI/DOI?10.1055/s-0029-1241797
  16. 16. Percik R, Stumvoll M. Obesity and cancer. Experimental and Clinical Endocrinology & Diabetes. 2009;117(10):563-566. Available from: http://www.thieme-connect.de/DOI/DOI?10.1055/s-0029-1241870
  17. 17. Beauchamp EM, Platanias LC. The evolution of the TOR pathway and its role in cancer. Oncogene. 2013;32(34):3923-3932. Available from: http://www.nature.com/articles/onc2012567
  18. 18. Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell. 2017;168(6):960-976. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0092867417301824
  19. 19. Roux PP, Blenis J. ERK and p38 MAPK-activated protein kinases: A family of protein kinases with diverse biological functions. Microbiology and Molecular Biology Reviews. 2004;68(2):320-344. Available from: http://mmbr.asm.org/cgi/doi/10.1128/MMBR.68.2.320-344.2004
  20. 20. Luo Z, Saha AK, Xiang X, Ruderman NB. AMPK, the metabolic syndrome and cancer. Trends in Pharmacology Sciences. 2005;26(2):69-76. Available from: https://www.sciencedirect.com/science/article/abs/pii/S0165614704003402
  21. 21. Li X, Wu C, Chen N, Gu H, Yen A, Cao L, et al. PI3K/Akt/mTOR signaling pathway and targeted therapy for glioblastoma. Oncotarget. 2016;7(22):33440-33450. Available from: http://www.oncotarget.com/fulltext/7961
  22. 22. Hay N. Upstream and downstream of mTOR. Genes & Development. 2004;18(16):1926-1945. Available from: http://www.genesdev.org/cgi/doi/10.1101/gad.1212704
  23. 23. Dowling RJO, Topisirovic I, Fonseca BD, Sonenberg N. Dissecting the role of mTOR: Lessons from mTOR inhibitors. Biochimica et Biophysica Acta, Proteins and Proteomics. 2010;1804(3):433-439. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1570963909003598
  24. 24. Suissa S, Azoulay L. Metformin and cancer: Mounting evidence against an association. Diabetes Care. 2014;37(7):1786-1788. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24963109
  25. 25. Lee M-S, Hsu C-C, Wahlqvist ML, Tsai H-N, Chang Y-H, Huang Y-C. Type 2 diabetes increases and metformin reduces total, colorectal, liver and pancreatic cancer incidences in Taiwanese: A representative population prospective cohort study of 800,000 individuals. BMC Cancer. 2011;11(1):20. Available from: http://bmccancer.biomedcentral.com/articles/10.1186/1471-2407-11-20
  26. 26. Suissa S, Azoulay L. Metformin and cancer: Mounting evidence against an association: Figure 1. Diabetes Care. 2014;37(7):1786-1788. Available from: http://care.diabetesjournals.org/lookup/doi/10.2337/dc14-0500
  27. 27. Rêgo DF, Pavan LMC, Elias ST, De Luca Canto G, Guerra ENS. Effects of metformin on head and neck cancer: A systematic review. Oral Oncology. 2015;51(5):416-422
  28. 28. 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 Prevention Research. 2010;3(11):1451-1461
  29. 29. Currie CJ, Poole CD, Gale EAM. The influence of glucose-lowering therapies on cancer risk in type 2 diabetes. Diabetologia. 2009;52(9):1766-1777. Available from: http://link.springer.com/10.1007/s00125-009-1440-6
  30. 30. Landman GWD, Kleefstra N, van Hateren KJJ, Groenier KH, Gans ROB, Bilo HJG. Metformin associated with lower cancer mortality in type 2 diabetes: ZODIAC-16. Diabetes Care. 2010;33(2):322-326. Available from: http://care.diabetesjournals.org/cgi/doi/10.2337/dc09-1380
  31. 31. Noto H, Goto A, Tsujimoto T, Noda M. Cancer risk in diabetic patients treated with metformin: A systematic review and meta-analysis. PLoS One. 2012;7(3):e33411. Available from: http://dx.plos.org/10.1371/journal.pone.0033411
  32. 32. Bowker SL, Yasui Y, Veugelers P, Johnson JA. Glucose-lowering agents and cancer mortality rates in type 2 diabetes: Assessing effects of time-varying exposure. Diabetologia. 2010;53(8):1631-1637. Available from: http://link.springer.com/10.1007/s00125-010-1750-8
  33. 33. Coyle C, Cafferty FH, Vale C, Langley RE. Metformin as an adjuvant treatment for cancer: A systematic review and meta-analysis. Annals of Oncology. 2016;27(12):2184-2195. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0923753419365354
  34. 34. Zhang Y, Storr SJ, Johnson K, Green AR, Rakha EA, Ellis IO, et al. Involvement of metformin and AMPK in the radioresponse and prognosis of luminal versus basal-like breast cancer treated with radiotherapy. Oncotarget. 2014;5(24):12936-12949. Available from: http://www.oncotarget.com/fulltext/2683
  35. 35. Rao M, Gao C, Guo M, Law BYK, Xu Y. Effects of metformin treatment on radiotherapy efficacy in patients with cancer and diabetes: A systematic review and meta-analysis. Cancer Management and Research. 2018;10:4881-4890. Available from: https://www.dovepress.com/effects-of-metformin-treatment-on-radiotherapy-efficacy-in-patients-wi-peer-reviewed-article-CMAR
  36. 36. Skinner HD, Sandulache VC, Ow TJ, Meyn RE, Yordy JS, Beadle BM, et al. TP53 disruptive mutations lead to head and neck cancer treatment failure through inhibition of radiation-induced senescence. Clinical Cancer Research. 2012;18(1):290-300
  37. 37. Fasih A, Elbaz HA, Hüttemann M, Konski AA, Zielske SP. Radiosensitization of pancreatic cancer cells by metformin through the AMPK pathway. Radiation Research. 2014;182(1):50-59. Available from: http://www.bioone.org/doi/10.1667/RR13568.1
  38. 38. Chang PH, Yeh KY, Wang CH, Chen EYC, Yang SW, Chou WC, et al. Impact of metformin on patients with advanced head and neck cancer undergoing concurrent chemoradiotherapy. Head & Neck. 2017;39(8):1573-1577
  39. 39. 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(6):1304-1308. Available from: http://care.diabetesjournals.org/cgi/doi/10.2337/dc09-1791
  40. 40. Bosco JLF, Antonsen S, Sorensen HT, Pedersen L, Lash TL. Metformin and incident breast cancer among diabetic women: A population-based case-control study in Denmark. Cancer Epidemiology, Biomarkers & Prevention. 2011;20(1):101-111. Available from: http://cebp.aacrjournals.org/cgi/doi/10.1158/1055-9965.EPI-10-0817
  41. 41. Campagnoli C, Pasanisi P, Abbà C, Ambroggio S, Biglia N, Brucato T, et al. Effect of different doses of metformin on serum testosterone and insulin in non-diabetic women with breast cancer: A randomized study. Clinical Breast Cancer. 2012;12(3):175-182. DOI: 10.1016/j.clbc.2012.03.004
  42. 42. El-Haggar SM, El-Shitany NA, Mostafa MF, El-Bassiouny NA. Metformin may protect nondiabetic breast cancer women from metastasis. Clinical & Experimental Metastasis. 2016;33(4):339-357
  43. 43. Goodwin PJ, Ligibel JA, Stambolic V. Metformin in breast cancer: Time for action. Journal of Clinical Oncology. 2009;27(20):3271-3273. DOI: 10.1200/JCO.2009.22.1630
  44. 44. Nanni O, Amadori D, De Censi A, Rocca A, Freschi A, Bologna A, et al. Metformin plus chemotherapy versus chemotherapy alone in the first-line treatment of HER2-negative metastatic breast cancer. The MYME randomized, phase 2 clinical trial. Breast Cancer Research and Treatment. 2019;174(2):433-442. DOI: 10.1007/s10549-018-05070-2
  45. 45. 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. Journal of Clinical Oncology. 2009;27(20):3297-3302. Available from: http://ascopubs.org/doi/10.1200/JCO.2009.19.6410
  46. 46. 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 Research and Treatment. 2012;135(3):821-830
  47. 47. Hadad SM, Coates P, Jordan LB, Dowling RJO, Chang MC, Done SJ, et al. Evidence for biological effects of metformin in operable breast cancer: Biomarker analysis in a pre-operative window of opportunity randomized trial. Breast Cancer Research and Treatment. 2015;150(1):149-155
  48. 48. Bonanni B, Puntoni M, Cazzaniga M, Pruneri G, Serrano D, Guerrieri-Gonzaga A, et al. Dual effect of metformin on breast cancer proliferation in a randomized presurgical trial. Journal of Clinical Oncology. 2012;30(21):2593-2600. Available from: http://ascopubs.org/doi/10.1200/JCO.2011.39.3769
  49. 49. Bayraktar S, Hernadez-Aya LF, Lei X, Meric-Bernstam F, Litton JK, Hsu L, et al. Effect of metformin on survival outcomes in diabetic patients with triple receptor-negative breast cancer. Cancer. 2012;118(5):1202-1211. Available from: http://doi.wiley.com/10.1002/cncr.26439
  50. 50. El-Benhawy SA, El-Sheredy HG. Metformin and survival in diabetic patients with breast cancer. The Journal of the Egyptian Public Health Association. 2014;89(3):148-153. Available from: http://content.wkhealth.com/linkback/openurl?sid=WKPTLP:landingpage&an=00004765-201412000-00007
  51. 51. Hou G, Zhang S, Zhang X, Wang P, Hao X, Zhang J. Clinical pathological characteristics and prognostic analysis of 1,013 breast cancer patients with diabetes. Breast Cancer Research and Treatment. 2013;137(3):807-816. Available from: http://link.springer.com/10.1007/s10549-012-2404-y
  52. 52. Goodwin P, Pritchard K, Ennis M, Clemons M, Graham M, Fantus IG. Insulin-lowering effects of metformin in women with early breast cancer. Clinical Breast Cancer. 2008;8(6):501-505. DOI: 10.3816/CBC.2008.n.060
  53. 53. He X, Esteva FJ, Ensor J, Hortobagyi GN, Lee MH, Yeung SCJ. Metformin and thiazolidinediones are associated with improved breast cancer-specific survival of diabetic women with HER2+ breast cancer. Annals of Oncology. 2012;23(7):1771-1780. Available from: https://academic.oup.com/annonc/article-lookup/doi/10.1093/annonc/mdr534
  54. 54. Ferguson RD, Gallagher EJ, Cohen D, Tobin-Hess A, Alikhani N, Novosyadlyy R, et al. Hyperinsulinemia promotes metastasis to the lung in a mouse model of Her2-mediated breast cancer. Endocrine-Related Cancer. 2013;20(3):391-401. Available from: https://erc.bioscientifica.com/view/journals/erc/20/3/391.xml
  55. 55. Rokkas T, Portincasa P. Colon neoplasia in patients with type 2 diabetes on metformin: A meta-analysis. European Journal of Internal Medicine. 2016;33:60-66. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0953620516301431
  56. 56. 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 Prevention Research. 2010;3(9):1077-1083
  57. 57. Miranda VC, Braghiroli MI, Faria LD, Bariani G, Alex A, Bezerra Neto JE, et al. Phase 2 trial of metformin combined with 5-fluorouracil in patients with refractory metastatic colorectal cancer. Clinical Colorectal Cancer. 2016;15(4):321-328.e1. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1533002816300597
  58. 58. Garrett CR, Hassabo HM, Bhadkamkar NA, Wen S, Baladandayuthapani V, Kee BK, et al. Survival advantage observed with the use of metformin in patients with type II diabetes and colorectal cancer. British Journal of Cancer. 2012;106(8):1374-1378. Available from: http://www.nature.com/articles/bjc201271
  59. 59. Higurashi T, Hosono K, Takahashi H, Komiya Y, Umezawa S, Sakai E, et al. Metformin for chemoprevention of metachronous colorectal adenoma or polyps in post-polypectomy patients without diabetes: A multicentre double-blind, placebo-controlled, randomised phase 3 trial. The Lancet Oncology. 2016;17(4):475-483. DOI: 10.1016/S1470-2045(15)00565-3
  60. 60. 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. British Journal of Cancer. 2016;114(3):281-289. DOI: 10.1038/bjc.2015.453
  61. 61. Schuler KM, Rambally BS, Difurio MJ, Sampey BP, Gehrig PA, Makowski L, et al. Antiproliferative and metabolic effects of metformin in a preoperative window clinical trial for endometrial cancer. Cancer Medicine. 2015;4(2):161-173
  62. 62. Laskov I, Drudi L, Beauchamp M-C, Yasmeen A, Ferenczy A, Pollak M, et al. Anti-diabetic doses of metformin decrease proliferation markers in tumors of patients with endometrial cancer. Gynecologic Oncology. 2014;134(3):607-614. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0090825814010543
  63. 63. Mitsuhashi A, Sato Y, Kiyokawa T, Koshizaka M, Hanaoka H, Shozu M. Phase II study of medroxyprogesterone acetate plus metformin as a fertility-sparing treatment for atypical endometrial hyperplasia and endometrial cancer. Annals of Oncology. 2016;27(2):262-266. Available from: https://academic.oup.com/annonc/article-lookup/doi/10.1093/annonc/mdv539
  64. 64. Ushijima K, Yahata H, Yoshikawa H, Konishi I, Yasugi T, Saito T, et al. Multicenter phase II study of fertility-sparing treatment with medroxyprogesterone acetate for endometrial carcinoma and atypical hyperplasia in young women. Journal of Clinical Oncology. 2007;25(19):2798-2803. Available from: http://ascopubs.org/doi/10.1200/JCO.2006.08.8344
  65. 65. Yates MS, Coletta AM, Zhang Q, Schmandt RE, Medepalli M, Nebgen D, et al. Prospective randomized biomarker study of metformin and lifestyle intervention for prevention in obese women at increased risk for endometrial cancer. Cancer Prevention Research. 2018;11(8):477-490. Available from: http://cancerpreventionresearch.aacrjournals.org/lookup/doi/10.1158/1940-6207.CAPR-17-0398
  66. 66. Nevadunsky NS, Van Arsdale A, Strickler HD, Moadel A, Kaur G, Frimer M, et al. Metformin use and endometrial cancer survival. Gynecologic Oncology. 2014;132(1):236-240. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0090825813012754
  67. 67. Ramos-Peñafiel C, Olarte-Carrillo I, Cerón-Maldonado R, Rozen-Fuller E, Kassack-Ipiña JJ, Meléndez-Mier G, et al. Effect of metformin on the survival of patients with ALL who express high levels of the ABCB1 drug resistance gene. Journal of Translational Medicine. 2018;16(1):245. DOI: 10.1186/s12967-018-1620-6
  68. 68. 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 Oncologica. 2013;52(5):1002-1009
  69. 69. Leamm S, Lagarde SM, van Oijen MGH, Gisbertz SS, Wilmink JW, Hulshof MCCM, et al. Metformin use during treatment of potentially curable esophageal cancer patients is not associated with better outcomes. Annals of Surgical Oncology. 2015;22(S3):766-771. Available from: http://link.springer.com/10.1245/s10434-015-4850-3
  70. 70. 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(9):1617-1622. Available from: http://link.springer.com/10.1007/s10552-009-9407-y
  71. 71. Azoulay L, Dell’Aniello S, Gagnon B, Pollak M, Suissa S. Metformin and the incidence of prostate cancer in patients with type 2 diabetes. Cancer Epidemiology, Biomarkers & Prevention. 2011;20(2):337-344. Available from: http://cebp.aacrjournals.org/cgi/doi/10.1158/1055-9965.EPI-10-0940
  72. 72. He XX, Tu SM, Lee MH, Yeung S-CJ. Thiazolidinediones and metformin associated with improved survival of diabetic prostate cancer patients. Annals of Oncology. 2011;22(12):2640-2645. Available from: https://academic.oup.com/annonc/article-lookup/doi/10.1093/annonc/mdr020
  73. 73. Feng T, Sun X, Howard LE, Vidal AC, Gaines AR, Moreira DM, et al. Metformin use and risk of prostate cancer: Results from the REDUCE study. Cancer Prevention Research. 2015;8(11):1055-1060. Available from: http://cancerpreventionresearch.aacrjournals.org/cgi/doi/10.1158/1940-6207.CAPR-15-0141
  74. 74. Rothermundt C, Hayoz S, Templeton AJ, Winterhalder R, Strebel RT, Bärtschi D, et al. Metformin in chemotherapy-naive castration-resistant prostate cancer: A multicenter phase 2 trial (SAKK 08/09). European Urology. 2014;66(3):468-474. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0302283813014826
  75. 75. Joshua AM, Zannella VE, Downes MR, Bowes B, Hersey K, Koritzinsky M, et al. A pilot ‘window of opportunity’ neoadjuvant study of metformin in localised prostate cancer. Prostate Cancer and Prostatic Diseases. 2014;17(3):252-258. DOI: 10.1038/pcan.2014.20
  76. 76. Nguyen MM, Martinez JA, Hsu CH, Sokoloff M, Krouse RS, Gibson BA, et al. Bioactivity and prostate tissue distribution of metformin in a preprostatectomy prostate cancer cohort. European Journal of Cancer Prevention. 2018;27(6):557-562
  77. 77. Rieken M, Kluth LA, Xylinas E, Fajkovic H, Becker A, Karakiewicz PI, et al. Association of diabetes mellitus and metformin use with biochemical recurrence in patients treated with radical prostatectomy for prostate cancer. World Journal of Urology. 2014;32(4):999-1005. Available from: http://link.springer.com/10.1007/s00345-013-1171-7
  78. 78. Spratt DE, Zhang C, Zumsteg ZS, Pei X, Zhang Z, Zelefsky MJ. Metformin and prostate cancer: Reduced development of castration-resistant disease and prostate cancer mortality. European Urology. 2013;63(4):709-716. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0302283812014728
  79. 79. Janjetovic K, Vucicevic L, Misirkic M, Vilimanovich U, Tovilovic G, Zogovic N, et al. Metformin reduces cisplatin-mediated apoptotic death of cancer cells through AMPK-independent activation of Akt. European Journal of Pharmacology. 2011;651(1–3):41-50. Available from: https://linkinghub.elsevier.com/retrieve/pii/S001429991001126X
  80. 80. Solano ME, Sander V, Wald MR, Motta AB. Dehydroepiandrosterone and metformin regulate proliferation of murine T lymphocytes. Clinical and Experimental Immunology. 2008;153(2):289-296. Available from: http://doi.wiley.com/10.1111/j.1365-2249.2008.03696.x
  81. 81. 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 Research. 2009;69(19):7507-7511. Available from: http://cancerres.aacrjournals.org/cgi/doi/10.1158/0008-5472.CAN-09-2994
  82. 82. Zhu P, Davis M, Blackwelder AJ, Bachman N, Liu B, Edgerton S, et al. Metformin selectively targets tumor-initiating cells in ErbB2-overexpressing breast cancer models. Cancer Prevention Research. 2014;7(2):199-210. Available from: http://cancerpreventionresearch.aacrjournals.org/cgi/doi/10.1158/1940-6207.CAPR-13-0181
  83. 83. Zhang Y, Guan M, Zheng Z, Zhang Q, Gao F, Xue Y. Effects of metformin on CD133+ colorectal cancer cells in diabetic patients. PLoS One. 2013;8(11):e81264. Available from: https://dx.plos.org/10.1371/journal.pone.0081264
  84. 84. Mohammed A, Janakiram NB, Brewer M, Ritchie RL, Marya A, Lightfoot S, et al. Antidiabetic drug metformin prevents progression of pancreatic cancer by targeting in part cancer stem cells and mTOR signaling. Translational Oncology. 2013;6(6):649-IN7. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1936523313800046
  85. 85. Montales MTE, Simmen RCM, Ferreira ES, Neves VA, Simmen FA. Metformin and soybean-derived bioactive molecules attenuate the expansion of stem cell-like epithelial subpopulation and confer apoptotic sensitivity in human colon cancer cells. Genes and Nutrition. 2015;10(6):49. Available from: http://link.springer.com/10.1007/s12263-015-0499-6
  86. 86. Kuo SZ, Honda CO, Li WT, Honda TK, Kim E, Altuna X, et al. Metformin results in diametrically opposed effects by targeting non-stem cancer cells but protecting cancer stem cells in head and neck squamous cell carcinoma. International Journal of Molecular Sciences. 2019;20(1):193. Available from: https://www.mdpi.com/1422-0067/20/1/193
  87. 87. Imamura K, Ogura T, Kishimoto A, Kaminishi M, Esumi H. Cell cycle regulation via p53 phosphorylation by a 5′-AMP activated protein kinase activator, 5-aminoimidazole-4-carboxamide-1-β--ribofuranoside, in a human hepatocellular carcinoma cell line. Biochemical and Biophysical Research Communications. 2001;287(2):562-567. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0006291X0195627X
  88. 88. Jones RG, Plas DR, Kubek S, Buzzai M, Mu J, Xu Y, et al. AMP-activated protein kinase induces a p53-dependent metabolic checkpoint. Molecular Cell. 2005;18(3):283-293. Available from: https://linkinghub.elsevier.com/retrieve/pii/S1097276505012207
  89. 89. Zhou J, Huang W, Tao R, Ibaragi S, Lan F, Ido Y, et al. Inactivation of AMPK alters gene expression and promotes growth of prostate cancer cells. Oncogene. 2009;28(18):1993-2002. Available from: http://www.nature.com/articles/onc200963

Written By

Yile Jiao, Xiaochen Wang and Zhijun Luo

Submitted: 26 November 2019 Reviewed: 21 January 2020 Published: 18 February 2020