Metformin, an oral biguanide widely used to treat diabetes, has considerable potential and is in clinical trials as an experimental preventive or therapeutic agent for a range of cancers. Direct actions targeting cellular pathways, particularly via AMP-activated protein kinase and through inhibiting mitochondrial ATP synthesis, or systemic mechanisms involving insulin and insulin-like growth factors have been much studied in vitro and in preclinical models. Epidemiologic and retrospective studies also provide clinical evidence in support of metformin as an antitumor agent. Preoperative window-of-opportunity trials confirm the safety of metformin in women with primary breast cancer, and demonstrate reduction in tumor cell proliferation and complex pathways of gene suppression or overexpression attributable to metformin. Confirmation of insulin-mediated effects, independent of body mass index, also supports the potential benefit of adjuvant metformin therapy. Neoadjuvant, adjuvant, and advanced disease trials combining metformin with established anticancer agents are under way or proposed. Companion biomarker studies will utilize in vitro and preclinical understanding of the relevant molecular pathways to, in future, refine patient and tumor selection for metformin therapy. Clin Cancer Res; 20(10); 2508–15. ©2014 AACR.

Synthetic biguanides, based on the guanidine derivative galegine originally derived from Galega officinalis (French lilac), have been used clinically since the 1950s with metformin (N′,N′-dimethylbiguanide) prescribed to more than 120 million type II diabetics globally. Phenformin, previously used in the treatment of diabetes, was withdrawn from many countries in the 1970s due to drug-associated lactic acidosis (1). The potential for repurposing metformin to oncology is supported by evidence of reduced cancer incidence and increased time to develop cancer (2), reduced mortality from cancer (3), with lower HER2-positive breast cancer-specific mortality (4), or reduced risk of distant metastases in triple-negative breast cancer (5) in diabetic women. Additional supportive evidence has come from studies demonstrating that higher serum insulin levels are associated with increased risk of breast cancer (6) and shorter survival from breast cancer (7) which may be modified by metformin.

The relevance of the multiple proposed mechanisms of action of biguanides to cancer remains controversial. In vitro, in vivo animal model and human studies support several possible modes of action with extrapolation of studies in diabetes, the range of in vitro models, nonphysiologic metformin levels, the circumstantial evidence from epidemiologic studies, and retrospective reviews creating complexity. Focused initially on diabetes and metabolism more generally, both direct (cellular) pathways and indirect (systemic) actions have been proposed as key mechanisms for the action of biguanides in diabetes that may also be of relevance to cancer.

AMPK-mediated effects

The master sensor of cellular energy, the Ser/Thr protein kinase complex AMP-activated protein kinase (AMPK), remains central to studies of metformin. AMPK activation stimulates TSC2, hence indirectly as well as AMPK directly inhibits mTOR and consequently by downregulating S6kinase leads to reduced protein synthesis, reduced cellular growth, and lowers proliferation (Fig. 1). A complex containing Liver kinase B1 (LKB1), classed as a tumor suppressor gene, activates heterodimers of AMPK subunits through phosphorylation of AMPKa2Thr-172, established as a link between LKB1, AMPK, and cancer (8). Breast cancer cells lacking LKB1 are resistant to the growth inhibitory effects of metformin in vitro and in vivo (9, 10, 11), and as a distinct mechanism, epigenetic inactivation of LKB1 has been reported in sporadic papillary breast cancer (12). Furthermore, the intracellular pathways downstream of HER2 amplification, PIK3CA mutation, and PTEN mutation or loss of PTEN expression converge on elevated mTOR signaling and are considered potential mechanisms for breast cancer development and of resistance to therapy. This makes the potential for metformin to work via targeting the mTOR cascade an attractive mechanism of action (13). Other, AMPK and TSC2 independent, mechanisms of mTOR inhibition by metformin include suppression of RAG GTPases (14).

Figure 1.

Overview of proposed metformin actions in vivo. Metformin (M), absorbed from the gut, acts via systemic and intracellular molecular pathways. Systemic actions of metformin via metformin uptake through OCT in the liver, AMPK inhibition of gluconeogenesis, and consequent reduction in insulin reduces insulin receptor (IR) stimulation and downstream intracellular effects (including via PI3K, akt, and HIF-1α) and leads to increased glucose uptake in tissues such as skeletal muscle. Cell level molecular pathways include reductions in mitochondrial respiratory function and activation by metformin of AMPK with some of the consequent intracellular events converging with the systemically driven insulin receptor pathway. AMPK stimulates phosphorylation of p53, ACC1, and ACC2, inhibits aromatase enzymes, directly and indirectly inhibits mTOR itself stimulated by key drivers for cancer (HER2, PIK3CA, and PTEN), and hence curtails S6kinase activity and its consequences. Additional potentially important effects, for example, on the stroma, and the complexities of subcellular networks are not illustrated.

Figure 1.

Overview of proposed metformin actions in vivo. Metformin (M), absorbed from the gut, acts via systemic and intracellular molecular pathways. Systemic actions of metformin via metformin uptake through OCT in the liver, AMPK inhibition of gluconeogenesis, and consequent reduction in insulin reduces insulin receptor (IR) stimulation and downstream intracellular effects (including via PI3K, akt, and HIF-1α) and leads to increased glucose uptake in tissues such as skeletal muscle. Cell level molecular pathways include reductions in mitochondrial respiratory function and activation by metformin of AMPK with some of the consequent intracellular events converging with the systemically driven insulin receptor pathway. AMPK stimulates phosphorylation of p53, ACC1, and ACC2, inhibits aromatase enzymes, directly and indirectly inhibits mTOR itself stimulated by key drivers for cancer (HER2, PIK3CA, and PTEN), and hence curtails S6kinase activity and its consequences. Additional potentially important effects, for example, on the stroma, and the complexities of subcellular networks are not illustrated.

Close modal

Additional, potentially important, AMPK mechanisms for oncology include phosphorylating and hence inactivating ACC1 and ACC2, consequently blocking fatty acid synthesis but increasing mitochondrial fatty acid uptake and subsequent breakdown by β-oxidation (15). Inactivation of AMPK promotes a metabolic shift to aerobic glycolysis through stabilization of hypoxia-inducible factor-1α (HIF-1α), and increases transcription of genes involved in glycolytic pathways, resistance to apoptosis, angiogenesis, and metabolic adaptation (16, 17). AMPK may act as a negative regulator of the Warburg effect under nutrient-rich and growth factor replete conditions in the absence of evident energy imbalance (16). In contrast, AMPK activation may increase resistance to stress and viability of cancer cells under hypoxic conditions of metabolic stress (18), which may be of particular relevance as tumor cancer cells migrate and metastasize.

AMPK phosphorylates p53 and leads to cell-cycle checkpoint activation, at least in mouse embryonic fibroblasts, allowing cells to survive energy deprivation (19). Indeed, metformin inhibition of complex 1 of the mitochondrial electron transport chain (20, 21, 22) is particularly effective in p53-deficient colon cancer cells (23) although the p53 status did not affect metformin sensitivity in other cancer cell lines (24, 25). However, loss of functional AMPK or p53 (as is common through mutation or p53 network dysfunction in most cancers) may constitute a sufficient energy crisis to result in metformin having a cytotoxic synthetic lethality effect (26). Thus, there is evidence that metformin may be therapeutically effective acting via p53, particularly where normal p53 function is abrogated, as is the case in most epithelial cancers.

Mitochondrial effects

Biguanides accumulate in mitochondria, with uptake promoted (for the cationic metformin at least) by organic cation transporter 1 (OCT1) to inhibit complex 1 of the mitochondrial respiratory chain, inhibit mitochondrial ATP synthesis, increase ADP:ATP and AMP:ATP ratios, and hence activate AMPK indirectly. Phenformin is 50-fold more potent than metformin as an inhibitor of mitochondrial complex 1 and being lipophilic (21) is less dependent on OCT (27), retaining the dose-dependent antiproliferative effect in ovarian cancer cell lines even in the presence of OCT1 siRNA (28). Given that oxidative phosphorylation-dependent production of ATP seems to be secondary to mitochondrial synthesis of anabolic precursors in proliferating cells (29), it has been suggested that metformin exerts the chemopreventive effects by functioning as a weak mitochondrial poison inhibiting oxidative phosphorylation (30).

Additional, non-AMPK–mediated mechanisms for the antineoplastic effect of metformin have been proposed through NF-κB, a pleiotropic protein complex that controls DNA transcription. Metformin may inhibit IkB and IKKa/b subunits and hence NF-κB and may prevent the translocation of NF-κB from the cytoplasm to the nucleus (31). Metformin seems to downregulate genes involved in mitosis and may act synergistically in this setting with taxanes (32). Metformin may also act via the DNA damage response mechanisms through ataxia telangiectasia mutated (ATM) and Chk2 (33), also potentially accounting for the cancer prevention effect.

Systemic effects

Biguanides have long been known to suppress hepatic glucose production (34) with metformin uptake via OCT leading to AMKP inhibition of hepatic gluconeogenesis, increase peripheral insulin sensitivity (and hence glucose uptake, e.g., in skeletal muscle), and reduce circulating insulin and insulin-like growth factor (IGF-I) levels, although it is less clear how metformin regulates IGF-I.

Certainly, the insulin receptor lies upstream of the PI3K/AKT/mTOR signaling pathways and breast cancer cells may have greater sensitivity to the effects of insulin such that metformin reducing insulin levels, and hence binding of insulin to receptors on breast cancer cells present very attractive mechanisms to indirectly inhibit tumor growth (13). This provides molecular insights into and supports the clinical relevance of insulin/IGF-I reduction in relation to cancer progression (7, 35, 36).

Phenformin

In murine models, phenformin has been associated with a reduced incidence of 7,12-dimethylbenz(a)anthracene (DMBA)-induced rat mammary tumors (37), enhances the effects of chemotherapy (38), and elicits antiproliferative effects, alongside inducing apoptosis, in neuroblastoma (39). Phenformin, like metformin, is associated with enhanced apoptosis and ATP depletion in AMPK-deficient cancer cells and may be most effective in cancer cells without a functional LKB1–AMPK pathway, as demonstrated in non–small cell lung cancer (40).

Phenformin has been shown to prevent and retard growth of breast cancer cell lines (41) and to protect against tumor development in a spontaneous lymphoma murine model exhibiting PTEN heterozygous mutations and LKB1 hypomorphic mutations with greater potency than metformin (11).

Mechanistic insights from xenograft work suggest both systemic and direct tumor/stromal effects may be elicited (41) and have led to speculation that phenformin may have the capability to block a reverse Warburg effect of onconutrient production by stroma feeding breast cancer cells in vivo (42, 43).

Phenformin may initially seem most attractive for trials in the advanced breast cancer setting (providing renal and hepatic functions are adequate) with the potential to avoid lactic acidosis (1) and potentially enhance cancer cell kill by concomitant administration of 2 deoxyglucose with phenformin (if confirmed beyond colorectal cancer cell lines; ref. 44).

Metformin

In vitro models suggest that metformin may be active against breast cancer cell lines regardless of the estrogen receptor (ER), progesterone receptor (PR), HER2 status or p53 status, and subtype of the cells (24, 25).

For ER-positive breast cancer, dependent on aromatase conversion of precursors to estrogens in postmenopausal women, metformin may inhibit aromatase enzyme expression in breast adipose tissue via AMPK (45). Similar effects on other clinically relevant genes have yet to be explored. Given that aromatase inhibitors are now the mainstay of neoadjuvant, adjuvant, and extended treatment in postmenopausal women with breast cancer, the potential for synergism between an aromatase inhibitor and metformin is under exploration in a Korean neoadjuvant trial and is planned for an international extended adjuvant trial. For premenopausal women, where tamoxifen remains the principal adjuvant therapy for ER-positive breast cancer, the additive effects of metformin to tamoxifen, at least in terms of reducing cell proliferation (46), merits subgroup analysis within the ongoing MA32 adjuvant metformin trial (47).

For HER2, breast cancer lines expressing HER2 have increased sensitivity to metformin (48) mediated via inhibition of S6K; in trastuzumab-resistant models, metformin disrupted ERBB2/IGF1R complexes and significantly reduced cell proliferation (49). Metformin, used at comparable clinical doses, delayed the onset, reduced the growth, and increased the lifespan of MMTV-her2/neu mice (50) accompanied by reductions in circulating insulin levels. As HER2-targeted therapy extends to other tumor types (such as gastric and esophageal cancer), similar metformin therapy approaches may merit consideration.

For triple-negative breast cancer, metformin was effective in vitro (and in an animal model) inducing AMPK and suppressing EGF receptor, mitogen-activated protein kinase, and Src phosphorylation and lowering cyclin D1 and cyclin E (51). The effect of metformin on triple-negative cell lines may, in part, be attributable to STAT3 inhibition (52).

More generally, reduced immunohistochemical staining for pAMPK was described in 318 of 349 (91%) breast cancers, with all subtypes of breast cancer represented (53). Although AMPK seems to protect against initial tumor development, in established breast cancers, lack of functional AMPK-dependent pathways may render breast cancer susceptible to metformin therapy.

Additional support for metformin as a therapeutic agent comes from in vivo models where metformin seems to selectively inhibit CD44+/CD24 low cancer stem cell-like populations (54), repress genes associated with epithelial to mesenchymal transition such as ZEB1 TWIST, SNAi1 (55), and work synergistically with chemotherapy agents (54). In addition, inhibition of glutamine metabolism, at least in prostate models, may synergize with metformin (56). Injected into tumors, albeit at potentially nonphysiologic doses, metformin may be effective alongside paclitaxel and carboplatinum as part of a combination chemotherapy strategy for the treatment of breast cancer (57).

However, some have questioned the relevance of selected preclinical data given the concentrations, doses, and delivery of metformin used (58). Nevertheless, studies have demonstrated cell inhibition using physiologically possible concentrations of metformin in vitro (51) and in mouse models (41, 50, 59). Even so, the multiplicity of mechanisms deduced in vitro may differ significantly in vivo while animal models (usually in immune compromised mice) may be different again from the cancer prevention and cancer therapy settings in humans.

Metformin and breast cancer trials

Given the preclinical data and that metformin has known pharmacokinetics and manageable toxicities, with extensive use worldwide in diabetes, repurposing metformin within clinical trials to seek efficacy against breast cancer is very attractive. Evidence to date suggests that metformin could be clinically useful in the prevention, preoperative, adjuvant, extended adjuvant, and advanced disease settings, although the detail of dose, duration, and drug combinations will require elucidation. Fortunately, the research community is actively engaged to address many of these roles and through companion biomarker studies ascertain the tumor and patient characteristics most amenable to treatment and whether direct or systemic effects are both of clinical importance.

Trials are under way examining these issues in the setting of breast, brain, colorectal, endometrium, head and neck, lung, lymphoma, melanoma, ovary, pancreas, prostate, renal, and thyroid malignancy. Single-center, phase II biomarker studies and/or with clinical endpoints predominate; many trials use gradual increases in dose up to 1,500 to 2,250 mg metformin per day to mitigate the self-limiting initial gastrointestinal side effects of nausea, diarrhoea, and bloating. Caution over the potential but rare metformin-associated lactic acidosis (1/100,000 years of use) generally restricts the clinical trials populations to women under 75 years of age with good renal and hepatic function.

A proposed study of metformin versus placebo to prevent the progression of atypical ductal hyperplasia, ductal, or lobular in situ neoplasias seems attractive given the evidence that even low-dose metformin (250 mg/day) is sufficient to reduce proliferation and aberrant crypt foci in rectal epithelium (60). The progression from ductal carcinoma in situ (DCIS) to invasive breast cancer may be mechanistically linked to glycolytic cancer-associated fibroblast activity (exporting lactate to the tumor cells as evidenced by a fall in caveolin and rise in MCT4 expression) with the potential for blockade by metformin (61).

The first randomized clinical trial using metformin (1,000 mg bd) in breast cancer was reported in 2011 and used the preoperative window-of-opportunity setting (62). This novel trial randomized patients to receive metformin for 2 weeks or watchful waiting for 2 weeks between diagnostic biopsy and definitive surgery. Core needle tumor biopsies were taken at the time of diagnosis and on the operating table before resection of the operable breast cancer to allow direct comparison of tumor proliferation (Ki-67) and tumor messenger RNA expression alongside serum insulin. The trial demonstrated a significant reduction in Ki-67 with metformin but not in the control arm, reduced gene expression [including in the phosphoinositide 3-kinase (PI3K) pathway], and increased gene expression (e.g., TNFR1), with a flattening of the serum insulin levels in response to surgery for the patients on metformin. Thus, both in vivo antitumor effects and systemic effects were demonstrated for the first time in women with primary breast cancer receiving metformin. Although women were not fasting at the time of diagnostic biopsy, the insulin effect and the lack of association between metformin effects and body mass index (BMI) confirm the potentially beneficial systemic effect of metformin as an insulin suppressor in women with breast cancer (7, 63).

A second, single-center, preoperative window-of-opportunity placebo-controlled trial of similar size confirmed the reduction in Ki-67 with metformin use over 2 weeks, again not associated with BMI, and provided enhanced evidence for the systemic insulin-mediated effects (64). The need for meticulous trial design was flagged by a subsequent trial (65) where failure to continue metformin up to the time of surgery and using core samples at diagnosis versus resection specimens (66) make data interpretation difficult.

Overall, these prospective, preoperative window studies in breast cancer (62, 64, 65), despite reservations about trial design and methodology, demonstrate that metformin is safe for women with primary breast cancer and confirm the attraction of metformin as a therapeutic agent.

Given the proposed mechanisms of action of metformin and the preclinical data, synergism of the biguanide with chemotherapy has been explored. A retrospective case note review of patients with breast cancer receiving neoadjuvant chemotherapy (67) demonstrated a remarkable 24% complete pathologic response (cPR) in diabetic patients incidentally taking metformin and receiving neoadjuvant chemotherapy compared with 16% cPR in nondiabetics and 8% cPR in diabetics not receiving metformin. A similar, also historical, series of epithelial ovarian cancer has demonstrated clinical benefit with increased progression-free survival and overall survival in diabetics taking metformin and receiving taxane/carboplatin chemotherapy compared with nondiabetics or diabetics not on metformin (68). However, incidental metformin use has not demonstrated survival benefit in two case series of diabetics with breast cancer (5, 67). This concept of combining metformin with neoadjuvant chemotherapy is in prospective trials of metformin alongside paclitaxel and FAC; docetaxel, epirubicin, and cyclophosphamide ± metformin; and in ISPY2 (ganitumab + metformin). An innovative trial being conducted in Oxford combines the window-of-opportunity and neoadjuvant concepts. The Oxford trial sequences a 2-week run in with metformin and then neoadjuvant chemotherapy (± metformin) is administered up to the time of surgery. Noninvasive positron emission tomography scanning and lipid metabolism studies are being used alongside tissue biopsies to examine the effects of metformin on breast cancer metabolism and lipid synthesis.

In addition to the potentially synergistic effects of metformin with chemotherapy against breast cancer, murine models suggest a cardio-protective effect of metformin which seems to abrogate the effects of doxorubicin on lactate dehydrogenase, CKMB, and glutathione (69), although evidence for such effects in humans is, as yet, lacking.

In the adjuvant setting, the NCIC CTG MA .32 trial (47) is examining the efficacy of adjuvant metformin for 5 years after complete resection of invasive breast cancer and following completion of chemotherapy and/or radiotherapy but concomitant with endocrine therapy or anti-HER2 therapy. Early data suggest beneficial effects of metformin compared with placebo on patient weight and metabolic factors which, if sustained, could significantly improve survival from breast cancer (7). Further clinical efficacy, companion biomarker, and mammographic density studies are eagerly awaited as the trial matures. There are also plans under way to trial metformin in the extended adjuvant setting, beyond 5 years, for ER-positive patients at risk of recurrence. Further survivorship trials using metformin along with exercise in breast and colon cancer are proposed.

In contrast with most clinical drug development, where molecular pathways are usually explored in advanced disease, this setting has only recently been reported for metformin. For ER+ advanced disease, there is no disadvantage to taking metformin concomitant with exemestane on exemestane levels (70) providing reassurance for the neoadjuvant and adjuvant trails using concomitant metformin and endocrine therapy under way. Interactions of metformin with a range of agents are proposed on the basis of an appreciation of the intracellular pathways outlined above, including trials of metformin with everolimus and exemestane in ER+ disease, metformin with erlotinib in triple-negative breast cancer, and combination chemotherapy ± metformin in HER2-negative advanced disease.

Biomarker studies built in to all these trials will be essential to identify the breast cancers with characteristics (such as the LKB1–AMPK or p53-defective pathways) most likely to be susceptible to the effects of metformin. Cancer heterogeneity of OCT1, and hence for cancer cell uptake of metformin, as observed in ovarian cancer (28), may merit measurement before considering metformin therapy in the clinical setting given, for example, the low expression of OCT1 in normal breast tissues. The potential effects of polymorphisms of LKB1, OCT 1,2,3, and ATM on resistance to metformin (70) and interactions between proton pump inhibitors and metformin via OCT 1,2,3 (70) will clearly need to be considered when metformin translates into routine clinical practice.

Biguanides, and particularly metformin, present an attractive proposition for repurposing as low toxicity antitumor agents, thus demonstrating how understanding the molecular pathways of cancer combined with clinical data will lead to further advances in oncology.

No potential conflicts of interest were disclosed.

This work was supported by Breast Cancer Campaign, Breast Cancer Research Scotland, Cancer Research UK, and Breakthrough Breast Cancer.

1.
Nattrass
M
,
Alberti
KG
. 
Biguanides
.
Diabetologia
1978
;
14
:
71
4
.
2.
Evans
JM
,
Donnelly
LA
,
Emslie-Smith
AM
,
Alessi
DR
,
Morris
AD
. 
Metformin and reduced risk of cancer in diabetic patients
.
BMJ
2005
;
330
:
1304
5
.
3.
Landman
GW
,
Kleefstra
N
,
van Hateren
KJ
,
Groenier
KH
,
Gans
RO
,
Bilo
HJ
. 
Metformin associated with lower cancer mortality in type 2 diabetes: ZODIAC-16
.
Diabetes Care
2010
;
33
:
322
6
.
4.
He
X
,
Esteva
FJ
,
Ensor
J
,
Hortobagyi
GN
,
Lee
MH
,
Yeung
SC
. 
Metformin and thiazolidinediones are associated with improved breast cancer-specific survival of diabetic women with HER2+ breast cancer
.
Ann Oncol
2012
;
23
:
1771
80
.
5.
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
:
1202
11
.
6.
Pisani
P
. 
Hyper-insulinaemia and cancer, meta-analyses of epidemiological studies
.
Arch Physiol Biochem
2008
;
114
:
63
70
.
7.
Goodwin
PJ
,
Ennis
M
,
Pritchard
KI
,
Trudeau
ME
,
Koo
J
,
Madarnas
Y
, et al
Fasting insulin and outcome in early-stage breast cancer: results of a prospective cohort study
.
J Clin Oncol
2002
;
20
:
42
51
.
8.
Hawley
SA
,
Gadalla
AE
,
Olsen
GS
,
Hardie
DG
. 
The antidiabetic drug metformin activates the AMP-activated protein kinase cascade via an adenine nucleotide-independent mechanism
.
Diabetes
2002
;
51
:
2420
5
.
9.
Zakikhani
M
,
Dowling
R
,
Fantus
IG
,
Sonenberg
N
,
Pollak
M
. 
Metformin is an AMP kinase-dependent growth inhibitor for breast cancer cells
.
Cancer Res
2006
;
66
:
10269
73
.
10.
Dowling
RJ
,
Zakikhani
M
,
Fantus
IG
,
Pollak
M
,
Sonenberg
N
. 
Metformin inhibits mammalian target of rapamycin-dependent translation initiation in breast cancer cells
.
Cancer Res
2007
;
67
:
10804
12
.
11.
Huang
X
,
Wullschleger
S
,
Shpiro
N
,
McGuire
VA
,
Sakamoto
K
,
Woods
YL
, et al
Important role of the LKB1–AMPK pathway in suppressing tumorigenesis in PTEN-deficient mice
.
Biochem J
2008
;
412
:
211
21
.
12.
Klubo-Gwiezdzinska
J
,
Costello
J
,
Patel
A
,
Bauer
A
,
Jensen
K
,
Mete
M
, et al
Treatment with metformin is associated with higher remission rate in diabetic patients with thyroid cancer
.
J Clin Endocrinol Metab
2013
;
98
:
3269
79
.
13.
Dowling
RJO
,
Niraula
S
,
Stambolic
V
,
Goodwin
PJ
. 
Metformin in cancer: translational challenges
.
J Mol Endocrinol
2012
;
48
:
R31
R43
.
14.
Kalender
A
,
Selvaraj
A
,
Kim
SY
,
Gulati
P
,
Brule
S
,
Viollet
B
, et al
Metformin, independent of AMPK, inhibits mTORC1 in a rag GTPase-dependent manner
.
Cell Metab
2010
;
11
:
390
401
.
15.
Steinberg
GR
,
Kemp
BE
. 
AMPK in health and disease
.
Physiol Rev
2009
;
89
:
1025
78
.
16.
Faubert
B
,
Boily
G
,
Izreig
S
,
Griss
T
,
Samborska
B
,
Dong
Z
, et al
AMPK is a negative regulator of the Warburg effect and suppresses tumor growth in vivo
.
Cell Metab
2013
;
17
:
113
24
.
17.
Semenza
GL
. 
Targeting HIF-1 for cancer therapy
.
Nat Rev Cancer
2003
;
3
:
721
32
.
18.
Jeon
SM
,
Chandel
NS
,
Hay
N
. 
AMPK regulates NADPH homeostasis to promote tumour cell survival during energy stress
.
Nature
2012
;
485
:
661
5
.
19.
Jones
RG
,
Plas
DR
,
Kubek
S
,
Buzzai
M
,
Mu
J
,
Xu
Y
, et al
AMP-activated protein kinase induces a p53-dependent metabolic checkpoint
.
Mol Cell
2005
:
18
;
283
93
.
20.
El-Mir
MY
,
Nogueira
V
,
Fontaine
E
,
Averet
N
,
Rigoulet
M
,
Leverve
X
. 
Dimethylbiguanide inhibits cell respiration via an indirect effect targeted on the respiratory chain complex I
.
J Biol Chem
2000
;
275
:
223
8
.
21.
Owen
MR
,
Doran
E
,
Halestrap
AP
. 
Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain
.
Biochem J
2000
;
348
:
607
14
.
22.
Brunmair
B
,
Staniek
K
,
Gras
F
,
Scharf
N
,
Althaym
A
,
Clara
R
, et al
Thiazolidinediones, like metformin, inhibit respiratory complex I: a common mechanism contributing to their antidiabetic actions?
Diabetes
2004
;
53
:
1052
9
.
23.
Buzzai
M
,
Jones
RG
,
Amaravadi
RK
,
Lum
JJ
,
DeBerardinis
RJ
,
Zhao
F
, et al
Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth
.
Cancer Res
2007
;
67
:
6745
52
.
24.
Zhuang
Y
,
Miskimins
WK
. 
Cell cycle arrest in metformin treated breast cancer cells involves activation of AMPK, downregulation of cyclin D1, and requires p27Kip1 or p21Cip1
.
J Mol Signal
2008
;
3
:
18
25.
Alimova
IN
,
Liu
B
,
Fan
Z
,
Edgerton
SM
,
Dillon
T
,
Lind
SE
, et al
Metformin inhibits breast cancer cell growth, colony formation and induces cell cycle arrest in vitro
.
Cell Cycle
2009
;
8
:
909
15
.
26.
Pollak
MN
. 
Investigating metformin for cancer prevention and treatment: the end of the beginning
.
Cancer Discov
2012
;
2
:
778
90
.
27.
Shu
Y
,
Sheardown
SA
,
Brown
C
,
Owen
RP
,
Zhang
S
,
Castro
RA
, et al
Effect of genetic variation in the organic cation transporter 1 (OCT1) on metformin action
.
J Clin Invest
. 
2007
;
117
:
1422
31
.
28.
Segal
ED
,
Yasmeen
A
,
Beauchamp
MC
,
Rosenblatt
J
,
Pollak
M
,
Gotlieb
WH
. 
Relevance of the OCT1 transporter to the antineoplastic effect of biguanides
.
Biochem Biophys Res Commun
2011
;
414
:
694
9
.
29.
Ward
PS
,
Thompson
CB
. 
Metabolic reprogramming: a cancer hallmark even Warburg did not anticipate
.
Cancer Cell
2012
;
21
:
297
308
.
30.
Sanchez-Alvarez
R
,
Martinez-Outschoorn
UE
,
Lamb
R
,
Hulit
J
,
Howell
A
,
Gandara
R
, et al
Mitochondrial dysfunction in breast cancer cells prevents tumor growth: understanding chemoprevention with metformin
.
Cell Cycle
2013
;
12
:
172
82
.
31.
Moiseeva
O
,
Deschênes-Simard
X
,
St-Germain
E
,
Igelmann
S
,
Huot
G
,
Cadar
AE
, et al
Metformin inhibits the senescence-associated secretory phenotype by interfering with IKK/NF-κB activation
.
Aging Cell
2013
;
12
:
489
98
.
32.
Rocha
GZ
,
Dias
MM
,
Ropelle
ER
,
Osório-Costa
F
,
Rossato
FA
,
Vercesi
AE
, et al
Metformin amplifies chemotherapy-induced AMPK activation and antitumoral growth
.
Clin Cancer Res
2011
;
17
:
3993
4005
.
33.
Vazquez-Martin
A
,
Oliveras-Ferraros
C
,
Cufí
S
,
Martin-Castillo
B
,
Menendez
JA
. 
Metformin activates an ataxia telangiectasia mutated (ATM)/Chk2-regulated DNA damage-like response
.
Cell Cycle
2011
;
10
:
1499
501
.
34.
Bailey
CJ
,
Turner
RC
. 
Metformin
.
N Engl J Med
1996
;
334
:
574
9
.
35.
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
.
Clin Breast Cancer
2012
;
12
:
175
82
.
36.
Goodwin
PJ
,
Ennis
M
,
Pritchard
KI
,
Trudeau
ME
,
Koo
J
,
Hartwick
W
, et al
Insulin-like growth factor binding proteins 1 and 3 and breast cancer outcomes
.
Breast Cancer Res Treat
2002
;
74
:
65
76
.
37.
Dilman
VM
,
Berstein
LM
,
Zabezhinski
MA
,
Alexandrov
VA
,
Bobrov
JF
,
Pliss
GB
. 
Inhibition of DMBA induced carcinogenesis by phenformin in the mammary gland of rats
.
Arch Geschwulstforsch
1978
;
48
:
1
8
.
38.
Dilman
VM
,
Anisimov
VN
. 
Potentiation of antitumor effect of cyclophosphamide and hydrazine sulfate by treatment with the antidiabetic agent, 1-phenylethylbiguanide (phenformin)
.
Cancer Lett
1979
;
7
:
357
61
.
39.
Caraci
F
,
Chisari
M
,
Frasca
G
,
Chiechio
S
,
Salomone
S
,
Pinto
A
, et al
Effects of phenformin on the proliferation of human tumor cell lines
.
Life Sci
2003
;
74
:
643
50
.
40.
Shackelford
DB
,
Abt
E
,
Gerken
L
,
Vasquez
DS
,
Seki
A
,
Leblanc
M
, et al
LKB1 inactivation dictates therapeutic response of non-small cell lung cancer to the metabolism drug phenformin
.
Cancer Cell
2013
;
23
:
143
58
.
41.
Appleyard
MV
,
Murray
KE
,
Coates
PJ
,
Wullschleger
S
,
Bray
SE
,
Kernohan
NM
, et al
Phenformin as prophylaxis and therapy in breast cancer xenografts
.
Br J Cancer
2012
;
106
:
1117
22
.
42.
Witkiewicz
AK
,
Whitaker-Menezes
D
,
Dasgupta
A
,
Philp
NJ
,
Lin
Z
,
Gandara
R
, et al
Using the “reverse Warburg effect” to identify high-risk breast cancer patients: stromal MCT4 predicts poor clinical outcome in triple-negative breast cancers
.
Cell Cycle
2012
;
11
:
1108
17
.
43.
Thompson
AM
,
Newman
TJ
. 
Milking the stroma in triple-negative breast cancer
.
Cell Cycle
2012
;
11
:
3
.
44.
Lea
MA
,
Chacko
J
,
Bolikal
S
,
Hong
JY
,
Chung
R
,
Ortega
A
, et al
Addition of 2-deoxyglucose enhances growth inhibition but reverses acidification in colon cancer cells treated with phenformin
.
Anticancer Res
2011
;
31
:
421
6
.
45.
Brown
KA
,
Hunger
NI
,
Docanto
M
,
Simpson
ER
. 
Metformin inhibits aromatase expression in human breast adipose stromal cells via stimulation of AMP-activated protein kinase
.
Breast Cancer Res Treat
2010
;
123
:
591
6
.
46.
Berstein
LM
,
Yue
W
,
Wang
JP
,
Santen
RJ
. 
Isolated and combined action of tamoxifen and metformin in wild-type, tamoxifen-resistant, and estrogen-deprived MCF-7 cells
.
Breast Cancer Res Treat
2010
;
128
:
109
17
.
47.
Good win
PJ
,
Stambolic
V
,
Lemieux
J
,
Chen
BE
,
Parulekar
WR
,
Gelmon
KA
, et al
Evaluation of metformin in early breast cancer: a modification of the traditional paradigm for clinical testing of anti-cancer agents
.
Breast Cancer Res Treat
2011
;
126
:
215
20
.
48.
Vazquez-Martin
A
,
Oliveras-Ferraros
C
,
Menendez
JA
. 
The antidiabetic drug metformin suppresses HER2 (erbB-2) oncoprotein overexpression via inhibition of the mTOR effector p70S6K1 in human breast carcinoma cells
.
Cell Cycle
2009
;
8
:
88
96
.
49.
Liu
B
,
Fan
Z
,
Edgerton
SM
,
Yang
X
,
Lind
SE
,
Thor
AD
. 
Potent antiproliferative effects of metformin on trastuzumab-resistant breast cancer cells via inhibition of erbB2/IGF-1 receptor interactions
.
Cell Cycle
2011
;
10
:
2959
66
.
50.
Anisimov
VN
,
Berstein
LM
,
Egormin
PA
,
Piskunova
TS
,
Popovich
IG
,
Zabezhinski
MA
, et al
Effect of metformin on life span and on the development of spontaneous mammary tumors in HER-2/neu transgenic mice
.
Exp Gerontol
2005
;
40
:
685
93
.
51.
Liu
B
,
Fan
Z
,
Edgerton
SM
,
Deng
XS
,
Alimova
IN
,
Lind
SE
, et al
Metformin induces unique biological and molecular responses in triple negative breast cancer cells
.
Cell Cycle
2009
;
8
:
2031
40
.
52.
Deng
XS
,
Wang
S
,
Deng
A
,
Liu
B
,
Edgerton
SM
,
Lind
SE
, et al
Metformin targets Stat3 to inhibit cell growth and induce apoptosis in triple-negative breast cancers
.
Cell Cycle
2012
;
11
:
367
76
.
53.
Hadad
SM
,
Baker
L
,
Quinlan
PR
,
Robertson
KE
,
Bray
SE
,
Thomson
G
, et al
Histological evaluation of AMPK signalling in primary breast cancer
.
BMC Cancer
2009
;
9
:
307
.
54.
Hirsch
HA
,
Iliopoulos
D
,
Tsichlis
PN
,
Struhl
K
. 
Metformin selectively targets cancer stem cells, and acts together with chemotherapy to block tumor growth and prolong remission
.
Cancer Res
2009
;
69
:
7507
11
.
55.
Vazquez-Martin
A
,
Oliveras-Ferraros
C
,
Cufí
S
,
Del Barco
S
,
Martin-Castillo
B
,
Menendez
JA
. 
Metformin regulates breast cancer stem cell ontogeny by transcriptional regulation of the epithelial-mesenchymal transition (EMT) status
.
Cell Cycle
2010
;
9
:
3807
14
.
56.
Fendt
SM
,
Bell
EL
,
Keibler
MA
,
Davidson
SM
,
Wirth
GJ
,
Fiske
B
, et al
Metformin decreases glucose oxidation and increases the dependency of prostate cancer cells on reductive glutamine metabolism
.
Cancer Res
2013
;
73
:
4429
38
.
57.
Iliopoulos
D
,
Hirsch
HA
,
Struhl
K
. 
Metformin decreases the dose of chemotherapy for prolonging tumor remission in mouse xenografts involving multiple cancer cell types
.
Cancer Res
2011
;
71
:
3196
201
.
58.
Stambolic
V
,
Woodgett
JR
,
Fantus
IG
,
Pritchard
KI
,
Goodwin
PJ
. 
Utility of metformin in breast cancer treatment, is neoangiogenesis a risk factor?
Breast Cancer Res Treat
2009
;
114
:
387
9
.
59.
Ben Sahra
I
,
Laurent
K
,
Loubat
A
,
Giorgetti-Peraldi
S
,
Colosetti
P
,
Auberger
P
, et al
The antidiabetic drug metformin exerts an antitumoral effect in vitro and in vivo through a decrease of cyclin D1 level
.
Oncogene
2008
;
27
:
3576
86
.
60.
Hosono
K
,
Endo
H
,
Takahashi
H
,
Sugiyama
M
,
Sakai
E
,
Uchiyama
T
, et al
Metformin suppresses colorectal aberrant crypt foci in a short-term clinical trial
.
Cancer Prev Res
2010
;
3
:
1077
83
.
61.
Lisanti
MP
,
Sotgia
F
,
Pestell
RG
,
Howell
A
,
Martinez-Outschoorn
UE
. 
Stromal glycolysis and MCT4 are hallmarks of DCIS progression to invasive breast cancer
.
Cell Cycle
2013
:
12
;
17
.
62.
Hadad
S
,
Iwamoto
T
,
Jordan
L
,
Purdie
C
,
Bray
S
,
Baker
L
, et al
Evidence for biological effects of metformin in operable breast cancer: a pre-operative, window-of-opportunity, randomized trial
.
Breast Cancer Res Treat
2011
;
128
:
783
94
.
63.
Goodwin
PJ
,
Pritchard
KI
,
Ennis
M
,
Clemons
M
,
Graham
M
,
Fantus
IG
. 
Insulin-lowering effects of metformin in women with early breast cancer
.
Clin Breast Cancer
2008
;
8
:
501
5
.
64.
Niraula
S
,
Dowling
RJ
,
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
;
135
:
821
30
.
65.
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
.
J Clin Oncol
2012
;
30
:
2593
600
.
66.
Mann
GB
,
Fahey
VD
,
Feleppa
F
,
Buchanan
MR
. 
Reliance on hormone receptor assays of surgical specimens may compromise outcome in patients with breast cancer
.
J Clin Oncol
2005
;
23
:
5148
54
.
67.
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
;
27
:
3297
302
.
68.
Romero
IL
,
McCormick
A
,
McEwen
KA
,
Park
S
,
Karrison
T
,
Yamada
SD
, et al
Relationship of type II diabetes and metformin use to ovarian cancer progression, survival, and chemosensitivity
.
Obstet Gynecol
2012
;
119
:
61
7
.
69.
Ashour
AE
,
Sayed-Ahmed
MM
,
Abd-Allah
AR
,
Korashy
HM
,
Maayah
ZH
,
Alkhalidi
H
, et al
Metformin rescues the myocardium from doxorubicin-induced energy starvation and mitochondrial damage in rats
.
Oxid Med Cell Longev
2012
;
2012
:
434195
.
70.
Esteva
FJ
,
Moulder
SL
,
Gonzalez-Angulo
AM
,
Ensor
J
,
Murray
JL
,
Green
MC
, et al
Phase I trial of exemestane in combination with metformin and rosiglitazone in nondiabetic obese postmenopausal women with hormone receptor-positive metastatic breast cancer
.
Cancer Chemother Pharmacol
2013
;
71
:
63
72
.