Vous êtes sur la page 1sur 14

REVIEWS

Metforminmode of action and clinical


implications for diabetes and cancer
Ida Pernicova and Mrta Korbonits
Abstract | Metformin has been the mainstay of therapy for diabetes mellitus for many years; however, the
mechanistic aspects of metformin action remained ill-defined. Recent advances revealed that this drug, in
addition to its glucose-lowering action, might be promising for specifically targeting metabolic differences
between normal and abnormal metabolic signalling. The knowledge gained from dissecting the principal
mechanisms by which metformin works can help us to develop novel treatments. The centre of metformins
mechanism of action is the alteration of the energy metabolism of the cell. Metformin exerts its prevailing,
glucose-lowering effect by inhibiting hepatic gluconeogenesis and opposing the action of glucagon. The
inhibition of mitochondrial complex I results in defective cAMP and protein kinase A signalling in response to
glucagon. Stimulation of 5'-AMP-activated protein kinase, although dispensable for the glucose-lowering effect
of metformin, confers insulin sensitivity, mainly by modulating lipid metabolism. Metformin might influence
tumourigenesis, both indirectly, through the systemic reduction of insulin levels, and directly, via the induction
of energetic stress; however, these effects require further investigation. Here, we discuss theupdated
understanding of the antigluconeogenic action of metformin in the liver and the implications of the discoveries
of metformin targets for the treatment of diabetes mellitus and cancer.
Pernicova, I. & Korbonits, M. Nat. Rev. Endocrinol. 10, 143156 (2014); published online 7 January 2014; doi:10.1038/nrendo.2013.256

Introduction
The biguanide metformin has been used for its glucose- cardiovascular morbidity and mortality remains con-
lowering effect since 1957 in Europe and since 1995 troversial. Although the UK Prospective Diabetes Study
in the USA. Yet despite being the most frequently pre- and several systematic reviews found an overall reduced
scribed antidiabetic treatment worldwide, its mechanism cardiovascular mortality and morbidity risk with met-
of action remains largely elusive. Metformin and the formin treatment,10 two meta-analyses have failed to
related phenformin are derivatives of guanidine (Table1). determine any benefit of metformin therapy on all-cause
Discovered in the 1920s in extracts of the plant Galega or cardiovascular-related mortality or on diabetes-related
officinalis (French lilac), isoamylene guanidine, also called macrovascular complications, pointing to methodological
galegine, had been used for centuries for the treatment weaknesses of previous studies and insufficient data.11,12
of diabetes mellitus.1 Phenformin has been withdrawn Here, we discuss the updated understanding of the
for use in humans as a result of safety concerns in most molecular mechanisms through which metformin acts
parts of the world, whereas the clinical indications for on metabolism, mainly focussing on liver gluconeo
metformin therapy have expanded from type2 diabetes genesis, and on tumourigenesis. In addition, we will
mellitus (where it offers undisputable benefits) to gesta- review the potential implications of new discoveries
tional diabetes mellitus, polycystic ovary syndrome, the about metformin molecular targets for the development
metabolic syndrome and diabetesprevention.2 of antidiabetic and anticancer therapies.
Metformin lowers glucose levels and improves insulin
sensitivity.3 In addition, metformin has gained atten- Metformin and diabetes mellitus
Department of
Endocrinology, William
tion for its pleiotropic effects. It has been shown to Antidiabetic actions of metformin
Harvey Research decrease food intake (Box1)4 and body weight3 in some The antihyperglycaemic action of biguanides is mainly a
Institute, Barts and studies. Moreover, this drug can positively influence consequence of reduced glucose output owing to inhibi-
TheLondon School of
Medicine and Dentistry, multiple cardiovascular risk markers (Box2), includ- tion of liver gluconeogenesis13 and, possibly to a lesser
Queen Mary University ing lipid profile3,5 and fatty liver,6 modulate inflamma- extent, increased insulin-mediated glucose uptake in the
of London,
Charterhouse Square,
tory markers7,8 (Box2) and possibly reduce cancer risk.9 skeletal muscle.14 Metformin has little effect on glucose
London EC1A 6BQ, UK However, many promising results are not without dispute. absorption through the gastrointestinal tract but slightly
(I. Pernicova, For example, whether metformin treatment can improve delays the absorption process.15,16 The pharmacokinetics
M.Korbonits).
and response to metformin reveal a wide interindividual
Correspondence to: variability. The polarity of metformin makes it depend-
M. Korbonits Competing interests
m.korbonits@ The authors declare an association with the following company: ent on membrane transporters for cellular uptake and
qmul.ac.uk Merck. See the article online for full details of the relationships. secretion. The main metformin transporters are solute

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 10 | MARCH 2014 | 143


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Key points is mediated through an increase in the tyrosine kinase


activity of the insulin receptor 19 and through enhanced
The glucose-lowering, insulin-sensitizing agent metformin works mainly by
activity and translocation of glucose transporters, such
reducing gluconeogenesis and opposing glucagon-mediated signalling in the
liver and, to a lesser extent, by increasing glucose uptake in skeletal muscle
as GLUT4 (also known as SLC2A4), to the plasma
The primary site of metformin action is the mitochondrion membrane.28 Increased insulin receptor expression19,29
The antihyperglycaemic effect of metformin is probably owing to defective and an enhanced ability to restore enzymatic pathways
protein kinase A signalling involved in insulin signalling 30 have also been attributed
Metformin affects lipid metabolism primarily via 5'-AMP-activated protein kinase to metformin.
(AMPK) activation
Antitumourigenic effects of metformin, which require further study, might Altered endocrine function in the pancreas
be partially due to systemic metabolic alterations, including the reduced
The antidiabetic action of metformin is not the result of
availability of insulin
In cancer cells, metformin acts as an inducer of energetic stress; AMPK-driven increased insulin levels31,32 but rather has been associated
inhibition of mTOR seems to be required for much of its antimitotic activity with reduced insulin concentrations in the circulation.33,34
Consequently, metformin monotherapy is associated with
only a minimal risk of hypoglycaemia.16 Glucose clamp
carrier family 22 members (SLC22A) 1 and 4 (also studies in nondiabetic individuals treated with metformin
known as OCT1 and OCTN1, respectively),17 multi suggest that a reduction in glucose levels lowers insulin
drug and toxin extrusion protein (MATE) 1 and 2, and secretion and increases glucagon secretion.35 However,
the plasma membrane monoamine transporter hENT4 this mechanism might be less effective in patients with
(also known as PMAT). The function of these metformin type2 diabetes mellitus, which might account for rare
transporters is summarized in Table2. cases of hypoglycaemia among patients with diabetes
mellitus receiving metforminmonotherapy.25,35
Inhibition of gluconeogenesis in the liver Importantly, metformin seems to interact with the
The liver, which expresses high levels of SLC22A1, is incretin axis, as an enhancer and sensitizer for the actions
considered to be the main site of action of metformin of glucagon-like peptide 1 (GLP1).36 GLP1 increases
(Table2). In addition, metformin concentration is higher secretion of insulin and reduces secretion of glucagon in
in the portal circulation than elsewhere in the body, which response to glucose and has widespread tissue-specific
might contribute to metformin accumulation in the liver.18 metabolic effects.37 Metformin stimulates expression
In this organ, metformin is suggested to have an effect of GLP1 receptor in the pancreas and increases plasma
on the regulation of glucose uptake, gluconeogenesis, GLP1 levels.37,38 Moreover, circulating levels of dipep-
glycolysis and glycogen synthesis (Figure1). tidyl peptidase 4 (DDP4), which is known to degrade
Metformin increases the activity of the insulin receptor incretins, were reported to be lower in patients treated
and of insulin receptor substrate 2 (IRS2) and enhances with metformin than in untreated individuals.39 However,
glucose uptake via increased translocation of glucose metformin did not directly inhibit DDP4 activity
transporters, such as GLUT1 (also known as SLC2A1), invitro.38,40 Combining DDP4 inhibitors with metformin
to the plasma membrane. 19 As a result, metformin improved glycaemic control in patients with type2 dia-
enhances the insulin-mediated suppression of gluconeo- betes mellitus.41 The full clinical relevance of the effect of
genesis.20 Furthermore, and possibly of greater impor- metformin on GLP1 is yet to be established. Metformin
tance, metformin opposes the gluconeogenic action of did not preserve cell function in outcomestudies.42
the peptide hormone glucagon.2022 The net effect of the To summarize, the ability of metformin to reduce cir-
interactions is that metformin inhibits gluconeogenic culating glucose levels in patients with type2 diabetes
enzymes and stimulates glycolysis by altering the activ- mellitus can be explained by multiple mechanisms, such
ity of multiple enzymes in these pathways (Figure1).23 as increased glucose uptake in liver and muscle, reduced
Gluconeogenesis accounts for 2897% of overall hepatic gluconeogenesis, improved GLP1 and reduced gluca-
glucose output depending on the feeding status in non- gon functions. Nevertheless, the molecular principles of
diabetic individuals, the rate being higher in patients with metformin action remain debated.
advanced type2 diabetes mellitus.24 In this patient popu-
lation, metformin was reported to reduce hepatic glucose Molecular targets of antidiabetic effects
output by up to 75%.25 Mitochondriathe primary site of action
The uptake of gluconeogenic substrates, such as alanine The primary target of metformin within the cell is the
and lactate,26 is reduced in the presence of metformin, mitochondrion, where metformin transiently inhib-
possibly owing to depolarization of the hepatocyte mem- its complex I of the mitochondrial electron transport
brane through metformin-stimulated Cl efflux.26 The chain, which induces a drop in energy charge, 4346 a
effects of metformin on hepatic glycogen metabolism measure of the energetic state of the cell (defined as
are not well-established; however, invitro treatment of [ATP]+0.5[ADP])/([ATP]+[ADP]+[AMP]). 46 This
hepatocytes decreased glycogen synthesis.27 inhibition has been demonstrated more markedly invitro
than invivo.45 The resulting decrease in ATP production
Increased glucose uptake in skeletal muscle and increase in AMP levels probably drive two major
Metformin improves insulin sensitivity and insulin- pathways: inhibition of glucagon-induced cAMP syn
mediated glucose uptake in skeletal muscle. This effect thesis, as demonstrated in the liver,21 and the activation

144 | MARCH 2014 | VOLUME 10  www.nature.com/nrendo


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 1 | Comparison of phenformin and metformin


Characteristic Phenformin Metformin
Chemical structure Biguanide Biguanide
Two guanidine groups linked together with the loss of an Two methyl groups on a guanide side-chain (Dimethylbiguanide)
ammonia group
Contains a phenyl-ethyl ring on a guanidine side-chain
(Phenethylbiguanide)
Physical properties Less polar and more lipid soluble than metformin127 Polar and unusually hydrophilic
and cellular Phenformin exhibits higher affinity and transport activity More reliant on active transcellular transport129*
transport with marked differences in uptake kinetics compared Lower affinity for mitochondrial membranes than phenformin
withmetformin128*
High affinity for mitochondrial membranes60
Effect on Powerful inhibitory effect on functioning of the Less powerful inhibitor of the mitochondrial respiratory chain (probably
mitochondrial mitochondrial respiratory chain the main reason for decreased risk of lactic acidosis)45
respiratory chain Inhibits lactate oxidation/increases lactate concentration Metformin increases lactate oxidation25
in plasma25 Not altering the release of lactate from muscle25
Increased release of lactate from muscle130
Drug metabolism Metabolized by CYP2D6 in the liver131 Not metabolized131
Phenformin metabolism can differ depending on
CYP2D6polymorphisms
Half-life Half-life of 715h132,133 Half-life of 1.56.5h20,132134
Elimination through the kidneys Food decreases and slightly delays absorption of metformin; metformin
is excreted by kidneys
Renal clearance is reduced by other factors, such as concomitant use
ofcimetidine
Clinical use Withdrawn from the markets in most parts of the world Widespread use and excellent safety profile
in the 1970s for safety reasons135 Many nephrologists propose that metformin is under-prescribed in
chronic kidney disease and guidelines should be revisited136
(metforminis deemed safe unless eGFR levels fall <30ml/min/1.73m2)
Adverse effects
Risk of lactic Increased risk of lactic acidosis, with a mortality rate Incidence of lactic acidosis is 0.03 cases per 1,000 patient-years
acidosis of3050% (1020times lower than with phenformin).131 The reported incidence of
0.40.64 cases per 1,000 patient-years131 lactic acidosis in diabetic patients on metformin is similar to the one in
diabetic patients not taking metformin137
Metformin has a wider therapeutic window and requires higher blood
levels than phenformin to cause lactic acidosis;132 metformin is generally
believed not to cause accumulation of lactate unless metabolism of
lactate is impaired for another reason
Cardiovascular risk Increased cardiovascular risk11 Reported cardiovascular benefits138
Gastrointestinal Gastrointestinal disturbance Gastrointestinal disturbance (especially at initiation of therapy), reported in
disturbances Reduced gastrointestinal glucose absorption15,16 ~30% of patients,139 may be one of the reasons for compliance issues
(reported in up to one third of patients140). The mechanisms of gastro-
intestinal side-effects are unknown but an increased mucosal serotonin
production139 and possibly rise in GLP1 may be partially responsible.
Little effect on intestinal glucose absorption15,16
B12 malabsorption and increased, homocystein levels with long-term use
Other clinical In cancer, invitro and invivo animal models show greater Following promising results of invitro and invivo animal models, the
effects antineoplastic activity with phenformin than metformin129,141 anti-tumour effects of metformin are being intensively investigated in
and phenformin was linked to an improved immunologic clinical trials. To date most available data is in colon and breast cancer,
status in breast cancer patients.142 However, the reporting surrogate markers143
anti-tumour effect of phenformin has not been formally
investigated in clinical trials
*Active transport is predominant in both phenformin and metformin.128 Abbreviations: CYP2D6, cytochrome P450 2D6; eGFR, estimated glomelural filtration rate; GLP1, glucagon-like peptide 1.

of 5'-AMP-activated protein kinase (AMPK).47 AMPK is for the metabolic effects of metformin.48 At normal pH,
an energy sensor and a master coordinator of an integra metformin is positively charged; therefore, its intra
ted signalling network that comprises metabolic and cellular transport depends on cationic transporters
growth pathways acting in synchrony to restore cellu- (Table2). The mitochondrial membrane potential might
lar energy balance. When activated, AMPK switches on drive accumulation of the positively charged metformin
catabolic pathways that generate ATP and switches off in the mitochondrial matrix.
anabolic, ATP-consumingpathways. The inhibition of complex I by metformin reduces
The specific molecular mechanisms involved in the NADH oxidation, lowering the proton gradient across
inhibition of mitochondrial complex I remain unclear. the inner mitochondrial membrane and the proton-
An interaction between biguanides and mitochondrial driven synthesis of ATP. As a result, the ATP:ADP:AMP
copper ions has been observed and reported to be crucial equilibrium is shifted towards increased AMP synthesis

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 10 | MARCH 2014 | 145


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Box 1 | The role of metformin on feeding behaviour and glucagon signalling, and the antidiabetic effect
ofmetformin.21
In some studies, metformin suppresses food intake,4 probably by increasing
Interestingly, although mitochondria seem to be the
the levels of glucagon-like peptide 1 (GLP-1)37 and by interacting with signalling
upstream or downstream of other hormones or cytokines (such as ghrelin, primary targets of metformin action, metformin can also
leptin and insulin) influence erythrocytes, which lack mitochondria, possi-
Metformin has been detected in the cerebrospinal fluid after oral administration158 bly by affecting membrane fluidity.50 Further exploration
Metformin has been shown to reduce appetite by counteracting ghrelin-induced of this mechanism is awaited.
5'-AMP-activated kinase (AMPK) signalling and inhibition of mammalian target
of rapamycin (mTOR) in the hypothalamus;159 metformin has also been reported Glucagon signalling and metformin
to inhibit dexamethasone-induced hypothalamic AMPK activity invitro157
Glucagon concentrations are abnormally elevated in
AMPK has been proposed to mediate feeding behaviour in response to different
the circulation of individuals with diabetes mellitus,
signalling pathways (including ghrelin, leptin and cannabinoid signalling) 160163
In the hypothalamus, metformin inhibits hypoglycaemia-induced AMPK activity as hyperglycaemia blunts suppression of the glucagon-
and the expression of the orexigenic neuropeptide Y,164 although the inhibition secreting pancreatic cells. Glucagon promotes gluco-
of neuropeptide Y and agouti-related peptide in other scenarios was mediated neogenesis, glycogenolysis and ketogenesis, and reduces
by signal transducer and activator of transcription 3 (STAT3) signalling rather glycogenesis and glycolysis (Figure1). These actions
than AMPK activity158,165 lead to increased hepatic glucose production during
fasting and an attenuated decline in hepatic glucose
synthesispostprandially.51
Box 2 | Metabolic* and anti-inflammatory benefits associated with metformin Glucagon performs its main effect via activation
Moderate reduction of BMI or weight-neutral effects (as opposed to the effects
of adenylate cyclase. The adenylate-cyclase-derived
of many other antidiabetic treatments)3 cAMP activates protein kinase A (PKA), which then
Weight loss preferentially involving adipose tissue25 phosphorylates downstream targets, such as the
Reduction in blood pressure that is independent of weight change;33 however, bifunctional enzyme 6phosphofructo2-kinase/
no change in blood pressure could be detected in other studies166,167 fructose2,6-bisphosphatase (one of the isoforms being
Beneficial effect on lipid levels in some studies,3,5 but not others166,167 PFK/FBPase1, encoded by PFKFB1). PFK/FBPase1
Reduced resistin levels168 and increased adiponectin levels169 phosphorylation inhibits PFK activity and stimulates
Reduced fatty liver in rodents6
FBPase1 activity, thereby lowering the intracellular
Reduced fibrinogen170 and upregulated fibrinolytic system171
Reduced levels of PAI1172 levels of fructose2,6-bisphosphate (Figure2). This com-
Inhibition of platelet aggregation171 pound is an allosteric inhibitor of glycolysis and activa-
Improved endothelium-dependent vasodilatation173 tor of gluconeogenesis, as it alters the activity of several
Reduced progression of carotid intima thickening was identified in patients enzymes in these pathways (Figures1 and 2). Activation
with type2 diabetes mellitus174 treated with metformin; however, no difference of PKA also leads to changes in gene expression, but this
in carotid intima progression was noted in nondiabetic patients with proven effect is somewhat slower than the one on enzymatic
coronary artery disease175
activity. For example, phosphorylation of the cAMP-
Reduced levels of Creactive protein167
responsive element-binding protein (CREB1) induces
Reduced levels of proinflammatory cytokines;7,8 for example, metformin
inhibited production of TNF by human monocytes invitro176 and lowered TNF binding of this protein to cAMP response element sites
levels in patients with diabetes mellitus7 within the promoter of gluconeogenic genes such as
Reduced expression of the adhesion molecules ICAM1 and VCAM1;177 for PEPCK and G6PC (Figure3).52,53 During fasting, PKA
example, metformin exerted an antiatherogenic effect in vascular endothelial also phosphorylates inositol 1,4,5-triphosphate receptors
cells through inhibition of TNFNF-B-driven expression of proinflammatory (I3PRs), thereby inducing an increase in intracellular
and cell adhesion molecules, in an AMPK-dependent manner178 and through Ca2+ levels.54 This increase, in turn, activates CREB-
blockade of the PI3KAKT pathway179
regulated transcription coactivator 2 (CRTC2), enab
Metformin has been reported to enhance CD8+ Tcell memory by altering fatty
acid metabolism,180 thereby linking metabolism and immunomodulation
ling interaction between CRTC2 and CREB1 to activate
Metformin has been shown to reduce endotoxin-induced hepatic injury in mice181 gluconeogenic gene expression (Figure3).55 This process
Metformin inhibited osteoclasts and bone resorption through immune is reduced during feeding owing to an increase in insulin
mechanisms182 signalling induced by the inactivation of I3PR via AKT
Metformin favourably modulated immune responses in animal models of (also known as PKB).55
multiple sclerosis,8 uveitis,183 autoimmune arthritis,184 and chronic asthma185 Following on from earlier reports,22 Miller etal.
*The metabolic benefits of metformin administration listed here are not consistent in all showed in a series of invitro and invivo experiments
studies. Abbreviations: AMPK, 5'-AMP-activated kinase; NFB, nuclear factor B; PI3K,
phosphoinositide 3kinase; PAI1, plasminogen activator inhibitor 1; TNF, tumor necrosis factor. that biguanides exert their glucose-lowering effect mainly
by opposing glucagon signalling (Figure3).21 Biguanides
were shown to rapidly inhibit the glucagon-stimulatory
by adenylate kinase,45 thereby reducing the energy charge effect on cAMP levels in hepatocytes, leading to dis-
of the cell. Direct inhibition of AMP deaminase, an rupted PKA activity and repressed phosphorylation of
enzyme that degrades AMP, by metformin might also PFK/FBPase 1, I3PR and CREB1.21 So, it was proposed
increase AMP levels.49 Raised AMP levels inhibit ade- that, at therapeutic concentrations, metformin leads to
nylate cyclase (see below), a membrane-bound enzyme inhibition of glucagon signalling, possibly by increas-
that catalyses the conversion of ATP to cAMP. Hence, ing AMP levels through inhibition of mitochondrial
metformin action ultimately reduces cAMP levels. A complexI (Figure3). AMP then binds to the regulatory
link has been made between reduced cAMP synthesis intracellular inhibitory Psite (a purine moietysite)

146 | MARCH 2014 | VOLUME 10  www.nature.com/nrendo


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 2 | Membrane transporters involved in metformin pharmacokinetics


Metformin transporter Encoded by Function
SLC22A1 (also known SLC22A1 Main transporter accountable for metformin uptake18
as OCT1) Expressed in liver and kidney
Some SNPs shown to associate with reduced metformin uptake, increased metformin
elimination as a result of reduced renal tubular reabsorption and lower therapeutic
response owing to diminished action of metformin in the liver144
Wide disparity in frequency distribution of SNPs among ethnic groups18
SLC22A2 (also known SLC22A2 Mediates metformin secretion (kidney)
as OCT2) Accountable for 80% of the total metformin clearance85
SLC22A3 (also known SLC22A3 Expressed in multiple tissues (including liver, kidney, heart, skeletal muscle, brain, placenta)
as OCT3) May be important in the uptake of metformin in muscle145
SLC22A4 (also known SLC22A4 Involved in the gastrointestinal absorption of metformin146
as OCTN1) Role in the mitochondrial uptake of phenformin17
MATE1 SLC47A1 Mediates metformin secretion (kidney; liverexcretion into bile)
Rs2289669 polymorphism was associated with an amplified glucose-lowering effect of
metformin in diabetic patients147
MATE2 SLC47A2 Mediates metformin secretion (kidney)
hENT4 (also known SLC29A4 Mediates renal and intestinal metformin uptake148
as PMAT)
Abbreviations: hENT4, equilibrative nucleotide transporter 4; MATE1, multidrug and toxin extrusion transporter 1; PMAT, plasma membrane monoamine transporter;
SLC22A1, solute carrier family 22 member 1; SNP, single nucleotide polymorphism.

of adenylate cyclase, 56 thereby inhibiting glucagon- gluconeogenesis was shown to be inhibited by the AMPK
mediated activation of adenylate cyclase and reducing activator AICAR,62 and was also reduced in a mutant
endogenous synthesis of cAMP and PKA activity. This rodent model expressing a constitutively active form of
action culminates in a glucose-lowering effect (Figure3). AMPK.63 In the liver, deletion of liver kinase B1 (LKB1;
It was also demonstrated that AMPKalthough acti- an upstream kinase that phosphorylates the catalytic
vated by metforminis dispensable for the above action domain of AMPK) prevented activation of AMPK and
and that AKT phosphorylation is unaltered shortly after negated the antidiabetic effect of metformin in mice fed
metformin administration,21 which suggests that met- a high-fat diet.13 Multiple studies reported involvement
formin does not affect insulin responsiveness acutely. of AMPK in the deactivation of CRTC2,13,6466 one of the
However, how metformin works at higher concentrations key regulators of gluconeogenic gene expression.
remains unclear, asin addition to inhibiting glucagon However, reservations regarding the hypothesis of
signallingit reduces glucose output in response to a AMPK being the main driving force behind reduced
membrane-p ermeable analogue of cAMP, bypassing hepatic gluconeogenesis have been accumulating
adenyl ate cyclase. 21 Furthermore, whereas glucagon over years, primarily owing to a lack of correlation
receptor knockout mice show hypoglycaemia,57 this between gluconeogenic gene expression and hepatic
outcome is unusual in patients receiving metformin glucose output. 67,68 The AMPK model was seriously
monotherapy, which suggests that either metformin inhi challenged when metformin lowered glucose produc-
bits glucagon signalling incompletely in humans or that tion in the liver of transgenic mice that lacked AMPK
other compensatory mechanisms are present. or its upstream activator LKB1.69 The investigators of
this breakthrough study69 hypothesized that the dis
The role of AMPK in hepatic gluconeogenesis cordance between their findings and the previous
Biguanides are recognized as indirect activators of model, which suggested that the antidiabetic action of
AMPK.47 For about a decade, AMPK was the assumed metformin requires LKB1,13 reflected possible benefits
prime mediator of metformin action. As mentioned of the LKB1AMPK axis for lipotoxicity in animals fed
above, metformin can activate AMPK by promot- a high-fat diet rather than a direct effect on gluconeo
ing AMP accumulation. Moreover, the drug has been genesis.69 Unlike PKA, AMPK does not control PFK
shown to activate AMPK without inducing any detect- activity in the liver.70 Foretz etal. demonstrated that
able changes in AMP, ADP and ATP,58,59 whereas phen- the ability of metformin to lower glucose output was
formin has been more consistent in modifying adenine preserved under a forced overe xpression of pe roxi
nucleotide ratio.60 some p roliferator-activated receptor- coactivator-1
The link between metformin and AMPK activa- (PGC-1) and an increase in protein levels of gluconeo-
tion was supported by experiments showing that the genic enzymes, such as phosphoenolpyruvate carboxy
effects of metformin on reduced glucose output and kinase (PEPCK) and glucose-6-phosphatase (G6Pase),
lipogenesis were mitigated by treatment with the concluding that metformin disrupts the activity of the
AMPK inhibitor compound C47 (although it was later gluconeogenic enzymes rather than the expression of the
deemed nonselective), or under adenoviral-mediated genes encoding them (PEPCK and G6PC, respectively).69
expression of dominant negative AMPK.61 Moreover, The proposed role of metformin in glucagon signalling

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 10 | MARCH 2014 | 147


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

oxidation71 (by regulating the activity of acetyl coen-


Glucose
zyme A carboxylase47 and the expression of multiple
GLUT-2 Plasma membrane enzymes involved in lipogenesis47,63 and oxidation23).
This mechanism is consistent with the observations of
altered fatty acid metabolism and improved hepatic stea-
+ Gluconeogenesis Glycolysis +
Glucose Glycogen tosis in mice receiving metformin.6 Free fatty acids can
G6Pase + GCK GYS PYGL
reduce glucose transport by hampering insulin signal-
+ +
ling 72 and have been implicated as the main culprit of
Glucose-6-phosphate Glycogen-1-phosphate
impaired insulin secretion by pancreatic cells.73 In addi-
Glucogenesis Glucogenolysis +
tion to free fatty acids, acetyl coenzymeA and citrate
Fructose-2,6-bisphosphate byproducts of oxidationinhibit key enzymes of the
Fructose-6-phosphate AMP glycolytic pathway (Figure1).70 Therefore, metformin
+ +
might improve insulin secretion and sensitivity by low-
FBP1 PFKL Fructose-2,6-bisphosphate ering the levels of glucose and free fatty acids, and by
+
preventing lipid deposition in insulin-sensitivetissues.
AMP Fructose-1,6-bisphosphate ATP, citrate, fatty acids

Direct modulation through energy charge
Phosphoenolpyruvate
The data presented by Miller etal. suggest a good corre
PEPCK + lation between metformin-induced elevation of AMP and
Pyruvate kinase + ATP the drop in cAMP and glucose levels in mice.21 In addition
Oxaloacetate Pyruvate Lactate to fructose2,6-bisphosphate, which regulates phospho
fructokinase1 (also called 6phosphofructokinase, liver
type; PFKL) and fructose1,6-bisphosphatase1 (FBP1)
Malate Acetyl-CoA with the greatest affinity, various other factors modulate
Oxaloacetate
gluconeogenic and glycolytic enzymes independently
Citric of glucagon and AMPK signalling, including metabo-
acid cycle Ketogenesis +
lites (such as citrate), the AMP:ATP ratio46,70,74 and the
Malate Citrate NADH:NAD+ status46 (Figure1). These findings suggest
-oxidation +
that metformin-induced changes in energy charge have
Mitochondrion contributory effects on gluconeogenesis, independent of
Glucagon effect cAMP and AMPK signalling.
Biguanide effect The molecular targets of metformin include the mito-
Figure 1 | The effects of glucagon and biguanides on gluconeogenic and glycolytic chondrial complex I and other enzymes modulated by
fluxes. The role of glucagon signalling on the expression and activity of various the altered energy charge, notably adenylate cyclase
enzymes and its opposition by biguanides is illustrated in a simplified scheme of (inhibited by increased AMP levels), affecting gluca-
hepatic glucose metabolism. Glycolysis is a pathway that converts glucose into gon signalling, and AMPK (stimulated by the increased
pyruvate whilst generating ATP; gluconeogenesis is an energy-consuming process AMP:ATPratio).
of glucose synthesis from non-carbohydrate precursors such as lactate or pyruvate.
Many of the metabolic steps of gluconeogenesis are the reverse of the glycolytic
Metformin and cancer
pathway. Both glucagon and biguanides can regulate these pathways. A rise in
fructose2,6-bisphosphate, induced by metformin, inhibits FBP1 and activates
In 2005, a report associated metformin use with a
PFKL. Biguanides abrogate glucagons effect on the gluconeogenic flux and reduced incidence of cancer,75 putting the drug into the
influence fatty acid metabolism. AMP and ATP have modulatory effects on several cancer research spotlight. As of October 2013, 173 clini-
metabolic steps. The rate of glycolysis and gluconeogenesis is also determined by cal trials on metformin and cancer have been registered.76
the concentration of glucose and lactate (and other precursors of glucose). The Diabetes mellitus has been associated with a 1.22.0-fold
stimulatory (+) and inhibitory () effects are highlighted in green for glucagon and in increase in cancer incidence.9 A report in 2010 suggested
red for biguanide signalling. Abbreviations: FBP1, fructose1,6-bisphosphatase 1; that metformin reduces this risk by approximately 40%
G6Pase, glucose6-phosphatase; GCK, glucokinase; GLUT2, glucose transporter 2;
compared with any other antidiabetic treatment.9 Several
GYS, glycogen synthase; PEPCK, phosphoenolpyruvate carboxykinase; PFKL,
6phosphofructokinase, liver type; PYGL, glycogen phosphorylase. studies have claimed a notable reduction in the risk of
all-cause and cancer-specific mortality, and with attenu-
ated cancer progression. 77,78 However, a population-
(increased AMP reduces cAMP levels)21 has supported based analysis has failed to show an association between
the AMPKindependent antihyperglycaemic action improved survival and metformin use in diabetic patients
ofmetformin. with breast cancer aged >65 years,79 and these findings
Nevertheless, a number of metformin effects are were in line with results of several other studies.80,81
still attributed to AMPK. AMPK phosphorylates and Little is known about the cancer-related effect of met-
increases the activity of insulin receptor and IRS2 formin in nondiabetic patients. A human study showed
and enhances translocation of glucose transporters that short-term treatment with metformin suppressed
(Figure3).19 AMPK is also responsible for the effect of formation of colorectal aberrant crypt foci;82 a trial of
metformin on fatty acid metabolism (Figure3).47 Acti longer duration is currently underway. Metformin treat-
vated AMPK inhibits fatty acid synthesis and enhances ment also reduced the cellular marker of proliferation,

148 | MARCH 2014 | VOLUME 10  www.nature.com/nrendo


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

a Glucagon b Biguanides

cAMP cAMP

PKA PKA
Phosphorylation Lack of phosphorylation
promotes FBPase 1 prevents FBPase 1
P PFK/FBPase 1 activity and activity and promotes PFK/FBPase 1
inactivates PFK PFK activity

FBPase 1 FBPase 1
F2,6BP F6P + P F2,6BP F6P + P
PFK PFK
Allosteric Allosteric Allosteric Allosteric
inhibition activation inhibition activation
by F2,6BP by F2,6BP by F2,6BP by F2,6BP
FBP1 PFKL FBP1 PFKL

Gluconeogenesis Glycolysis Gluconeogenesis Glycolysis

Figure 2 | The effect of glucagon and biguanide signalling on fructose2,6-bisphosphate. a | Glucagon binds to glucagon
receptor coupled to Gs-protein (not shown). Gs activates adenylate cyclase which generates cAMP from ATP (not shown).
cAMP stimulates protein kinase A (PKA) that phosphorylates 6phosphofructo2-kinase/fructose2,6-bisphosphatase 1
(PFK/FBPase 1). PFK/FBPase 1 is a bifunctional enzyme that regulates the levels of fructose2,6-biphosphate (F2,6BP) by
catalysing both synthesis and hydrolysis of F2,6BP to and from fructose6-phosphate (F6P), respectively. F2,6BP is an
allosteric activator of phosphofructokinase1 (PFKL), therefore it controls the rate of conversion of fructose6-phosphate to
fructose1,6-bisphosphate in the glycolytic pathway. At the same time, F2,6BP is also an allosteric inhibitor of fructose1,6-
bisphosphatase 1 (FBP1), thereby regulating the rate of gluconeogenesis. PFK/FBPase 1 phosphorylation leads to
conformational changes that favour FBPase 1 activity, therefore lowering F2,6BP levels. A drop in F2,6BP levels promotes
gluconeogenesis and inhibits glycolysis. b | Biguanides abrogate glucagon signalling. cAMP production is inhibited resulting
in decreased PKA activity, increased PFK activity leading to increased F2,6BP levels. A rise in F2,6BP levels suppresses
gluconeogenesis and stimulates glycolysis. Abbreviation: F2,6BP, fructose2,6-bisphosphate.

Ki67, in biopsy samples obtained from nondiabetic principle remains limited. Although exogenous insulin
women with breast cancer treated with metformin.83 used in the treatment of diabetes mellitus does not pro
Mechanisms by which metformin attenuates tumouri- mote cancer,84 there is some controversy regarding the
genesis and has chemoprotective properties are not well- risk for patients treated with the long-acting insulin
defined. An important limitation of many experimental analogueglargine.87
studies is that metformin inhibits cell proliferation invitro Moreover, it is possible that other hormones, cytokines
at supraphysiological concentrations,84 which are gener- and metabolic intermediates that influence tumourigenesis
ally thought to be unachievable in patients. Moreover, are also affected by metformin. A systemic effect related
many factors influence availability and response to met- to reduced glucagon signalling in glucagon-responsive
formin in tissue.85 For example, the tissue expression cells21 warrants exploration. More studies are needed to
of transporters that mediate metformin uptake varies clarify this principle in patients withnormoglycaemia.
between normal and tumour cells, and can be influenced
by various drugs such as antibiotics and proton pump Direct effects of metformin in cancer
inhibitors.86 Poor uptake into target cells might limit The antitumourigenic effects of metformin can be inde-
the therapeutic potential of metformin in cancer; on the pendent of insulinaemia.88 Several findings support the
other hand, this fact could play a crucial part in the drugs notion of a direct action of metformin in cancer cells. In
excellent safety profile. Overall, the interplay between the this context, AMPK-dependent and AMPK-independent
patients metabolic make-up and the molecular character- mechanisms have been described, which are likely to
istics of the tumour adds to the complexity of the effects coexist and interact.89,90
of metformin on tumourigenesis, which are probably
both systemic (indirect) and local (direct) (Figure4). AMPK-dependent metfomin effects in cancer
LKB1-dependent and AMPK-dependent suppression of
Systemic metformin effects on tumourigenesis the mammalian target of rapamycin (mTOR) pathway
Hyperinsulinaemia has been associated with adverse is possibly the most potent antineoplastic effect of met-
prognosis in several cancers (including breast, colon and formin (Figure4). mTOR inhibition disturbs protein
prostate cancer).84 In states of insulin resistance, met- synthesis and, thereby, tumour cell proliferation. Loss
formin action in the liver lowers systemic glucose levels of LKB1 is frequent in cancer, and a germline mutation
and improves secondary hyperinsulinaemia, preventing in LKB1 is responsible for PeutzJeghers syndrome, a
the latters effects on tumour growth and progression. cancer-predisposing condition.91,92
This action means that metformin can affect insulin- mTOR is a catalytic subunit of two multiprotein com-
sensitive neoplastic tissues indirectly, without the need plexes, mTORC1 and mTORC2. These complexes are
to accumulate in cancer cells. Our understanding of this central in the regulation of cellular growth and integrate

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 10 | MARCH 2014 | 149


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

to a complex tumour syndrome, which suggests that


Metformin Glucagon
Adenylate cyclase
induction of DICER1 expression by metformin could
Plasma
membrane SLC22A1 activity be an additional antineoplastic mechanism (Figure4).98
Metformin also inhibits the proto-oncogene cMYC
Mitochondrion
P and hypoxia-inducible factor 1 (HIF1) via AMPK
Complex I Binding to (Figure4).98 Importantly, AMPK and HIF1 have a key
P-site
Pyruvate kinase ATP cAMP levels role in metabolic transformation in cancer.
PFKL activity ATP production Rapidly dividing cells, such as tumour or inflamma-
tory cells, show altered metabolism that favours glyco
PFKL activity AMP PKA activity
FBP1 activity lysis to oxidative phosphorylation as energy source, even
AMPK activation
under aerobic conditions, a process called the Warburg
effect.99,100 This metabolic transformation promotes fatty
I3PR CREB-1/PGC-1 PFK/FBPase 1
acid synthesis and ensures availability of intermediates as
Improved Improved Reduced
insulin glucose fatty building blocks for proliferating cells. Probably one of the
receptor transport acid
function synthesis key mediators of this reprogramming is the mTORC1-
Ca+ activated HIF1,101 a transcription factor promoting
PEPCK
and G6Pase Glycolytic Gluconeogenic expression of glycolytic enzymes, GLUT1 and mono-
Improved insulin sensitivity CRTC2 expression pathways pathways carboxylate transporter 4 (MCT4), involved in lactic
Figure 3 | Model of metformin action in the hepatocyte. Metformin enters the cell
acidtransport.
through transporters such as SLC22A1 and inhibits mitochondrial complex I. A defect in the LKB1AMPK pathway potentiates the
Complex I inhibition results in reduced ATP levels and an accumulation of AMP. risk of metabolic transformation of pre-neoplastic cells;
AMP binds to the so called P-site at the adenylate cyclase enzyme and inhibits its hence, many tumours have inactive LKB1.102 AMPK has
activity, leading to reduced generation of cAMP upon stimulation of the glucagon been shown to negatively regulate the Warburg effect
receptor. As a result, PKA activation and its downstream pathways are inhibited. and tumour growth invivo.103 Metformin decreased
Gluconeogenesis is suppressed as a result of reduced activity of enzymes involved HIF1 mRNA and protein levels in a breast cancer
in the gluconeogenic flux (for example, owing to lack of phosphorylation of PFK/
model,98 exhibiting an antiproliferative, anti-Warburg
FBPase 1) and reduced gene expression (owing to decreased phosphorylation of the
transcription factor CREB1). Metformin-induced change in energy charge also potential, probably via AMPK. Metformin also acted
activates AMPK, which suppresses fat metabolism and possibly also contributes to antimitotically by inhibiting expression of fatty acid
the reduced gluconeogenic gene expression. In addition, AMP and ATP have an synthase104 and favouring fatty acid oxidation in cancer
independent modulatory role on these pathways. Abbreviations: AMPK, 5'-AMP- cells of the colon.105
activated protein kinase; CREB1, cAMP response element-binding protein; CRTC2, The effect of metformin on cell metabolism and
CREB regulated transcription coactivator 2; G6Pase, glucose6-phosphatase; I3PR, tumour growth is controversial. In pre-neoplastic cells
inositol 1,4,5-triphosphate receptors; PEPCK, phosphoenolpyruvate carboxykinase;
with an intact AMPK axis, metformin can counter-
PFK/FBPase 1, 6phosphofructo2-kinase/fructose2,6-bisphosphatase 1; PGC1,
peroxisome proliferator-activated receptor coactivator1; PKA, protein kinase A;
act the Warburg effect (that is, inhibit oxidative glyco
SLC22A1, solute carrier family 22 member 1. lysis).106 However, in established tumours, as opposed to
pre-neoplastic cells, the presence of LKB1 and AMPK
might confer a survival advantage to tumour cells by
input from various hormonal signalling and energy- protecting them against energetic stress.107 It can even
sensing pathways, including the insulin, insulin-like growth cause increased glycolytic flux under certain micro
factor 1 (IGF1), IGF2 and AMPK pathways (Figure4).93 environments, such as acidic pH;108 therefore, AMPK
AMPK-dependent downregulation of mTORC1 results activators could potentially be harmful. Conversely,
from activation of the tumour suppressor genes tuber- absence of LKB1 or AMPK in established tumours makes
ous sclerosis complex 1 (TSC1) and TSC2, which form the cancer cells selectively more vulnerable to depleted
an mTOR-inhibiting complex. Moreover, AMPK directly ATP incurred by metformin, as their ability to restore
inhibits RAPTOR, a positive regulator of mTOR.94 energy balance is impaired.109
Metformin can inhibit IGF1insulin signalling
through AMPK-dependent phosphorylation of IRS1, AMPK-independent metformin effects in cancer
which transmits signals from the insulin receptor and Whilst growth factors regulate mTORC1 through the
IGF1 receptor to the PI3KAKT pathway (Figure4). PI3KAKTTSC1TSC2 axis, amino acids can activate
This activity also has the potential to downregulate mTORC1 signalling through the RAG family of GTPases
mTOR signalling.95 However, multiple regulatory feed- (also called the Ras-related GTPases), independently of
back loops possibly counteract the antitumour effect of AMPK.110,111 Following its activation by the Ragulatory
this cascade during chronic metformin exposure.96 complex, the RAG GTPases recruit mTORC1 to the lyso-
The tumour suppressor protein p53 activates various somal surface, where it is activated by RHEB. Metformin
genes that can inhibit the AKT and mTORC1 pathways.97 can inhibit mTORC1 signalling by inactiv ating the
p53 is one of the targets of AMPK (Figure4), but the role Ragulatory complex, thereby mimicking the effect of
of metformin in p53 activation remains controversial.97 amino-acid withdrawal.89 This mechanism of action,
Furthermore, metformin induces expression of which is responsive to perturbations in energy status, may
DICER1, an enzyme that is involved in microRNA prove important in certain types of cancer 110 and, parallel
synthesis. Loss of function mutations in DICER1 lead to dietary energy restriction, in cancerprevention.112

150 | MARCH 2014 | VOLUME 10  www.nature.com/nrendo


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Indirect effects Direct effects

Metformin Metformin Insulin Metformin p53

IGF-1 and insulin receptors

IRS-1
P ROS ATM
Body PI3K
weight Ragulator

Other upstream AKT


Inflammation kinases RAG GTPases
P
P
Insulin resistance LKB1 TSC2TSC1 RHEB
Glucose levels P P
Insulin levels P
CaMKK2 AMPK mTORC1
P P

P
P P P HIF-1 Protein synthesis
Fatty acid Cell growth
synthesis cMYC DICER p53 NF-B IL-6 Cell viability

Figure 4 | Proposed actions of metformin in cancer. The antitumour effect of metformin is probably a combination of indirect
(systemic) and direct effects. Systemic influence is secondary to the effects of metformin on metabolism in insulin-sensitive
target tissues. Metformin lowers systemic glucose and insulin levels, which decreases insulin-mediated tumour growth and
progression. Possible anti-inflammatory effects may also reduce cancer risk. Metformin might inhibit mTORC1 through
regulation at multiple levels. Low energy charge in metformin-treated cancer cells activates AMPK, which can restrain cell
growth and proliferation. AMPK activates the tumour suppressor gene TSC2, resulting in inhibition of the mTORC1 activator
RHEB. AMPK can directly phosphorylate and inhibit RAPTOR, a member of the mTORC1 complex. Metformin might also
decrease signalling downstream of IGF1insulin receptors by lowering insulin levels and through AMPK-dependent
phosphorylation of IRS1. This action inhibits AKT and mTORC1 signalling. Moreover, AMPK has been suggested to hinder
cancer cell growth through numerous other mechanisms, including activation of HIF1, p53, cMYC and DICER1, and
suppression of fatty acid synthesis. AMPK may also be a mediator of an attenuated inflammatory feedback loop (NF-B/IL-6
pathway), restraining malignant transformation. Metformin can inhibit mTORC1 also in an AMPK-independent manner via direct
inactivation of the Ragulator complex, which results in inhibition of RAG GTPases and dissociation of mTORC1 from its activator
RHEB. Other AMPK-independent effects include inhibition of the serine-protein kinase ATM and reduction of ROS levels, which
are produced by mitochondrial complex I and confer mutagenesis risk. Abbreviations: AMPK, 5'-AMP-activated protein kinase;
CaMKK2, Calcium/calmodulin-dependent protein kinase kinase 2; IGF1, insulin-like growth factor 1; IRS1, insulin receptor
substrate 1; IL-6, interleukin 6; LKB1,liver kinase B1; mTORC1, mammalian target of rapamycin complex 1; NF-B, nuclear
factor B, PI3K, phosphatidylinositide 3kinase; ROS, reactive oxidative species; TSC2, tuberous sclerosis complex 2.

Through inhibition of mitochondrial complex I, met- cytotoxic therapies.116 Metformin has been reported to
formin reduces production of reactive oxygen species, improve cancer responses to radiation therapy, probably
oxidative stress and DNA damage,113 therefore reduc via downregulation of the hyperactive PI3KAKTmTOR
ing the risk of mutagenesis. Variation in the glycaemic pathway. 117 Moreover, metformin enhanced apopto-
response to metformin in patients with type2 dia sis induced by paclitaxel and cisplatin in endometrial
betes mellitus has been associated with the presence of cancer by repressing glyoxalase I expression through an
common genetic variants near the ataxia telangiectasia undefined mechanism.118 However, metformin antago-
mutated (ATM) gene locus.114 This observation suggests nized cisplatin-induced cytotoxicity in glioma, neuro
that metformin-dependent signalling involves ATM blastoma, fibrosarcoma and leukaemia cell lines through
activation. ATM encodes a tumour-suppressor protein, an AMPK-independent activation of AKT.119 Thus, use
a crucial component of the DNA-damage response of metformin in combination with chemotherapy drugs
network system that is required for DNA repair and in cancer warrants a degree ofcaution.119
cell-cycle control. ATM is mutated in ataxia telangi Tyrosine kinase inhibitors reduce insulinIGF1
ectasia, a neurodegenerative condition associated with receptor signalling, therefore metformin is frequently
a predisposition to cancer, insulin resistance and type2 used in combination with these agents to counter
diabetes mellitus. Both AMPK-dependent and AMPK- iatrogenic hyperglycaemia. The contribution of met-
independent mechanisms are probably involved in the formin to the antineoplastic success of this combination
ATM-mediated reparatory effect. Metformin, as a cellu remainsunclear.84
lar stressor, activates reparatory processes, even in the In mouse xenograft cancer models, metformin selec-
absence of DNA damage, which might be protective tively kills cancer stem cells, even at low concentrations,
against premalignant to malignant transformation.115 and prolongs tumour remission when combined with
chemotherapy agents,120,121 probably through the regu-
Metformin combined with cytotoxic therapy lation of inflammatory pathways (Box2). Metformin is
Multiple studies have looked into the antineoplastic thought to inhibit cancerous transformation by attenuat-
potential of metformin when used in combination with ing the inflammatory feedback loop, a signalling cascade

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 10 | MARCH 2014 | 151


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

Table 3 | Potential therapeutic targetslessons learned from metformin-related metabolic studies


Theoretical targets or compounds under investigation Insulinresistance Oncology
and/or diabetes
mellitus
Potential renaissance of phenformin use (better absorbed and a probably more potent mitochondrial
inhibitor than metformin; its adverse effect profile is better than that of most cytotoxic drugs129)
Short courses of high-dose metformin84 (mechanism of action of high doses of metformin is unknown)
Novel biguanides
New molecules that are better tolerated and have less adverse effects than current biguanides
Molecules less dependent on active transport than metformin and with more suitable
pharmacokinetics (better absorbed in the gut, better absorbed in the target cells)
Molecules affecting mitochondrial respiratory chain
Tissue-selective (e.g. liver)
Different degree of energy stress
Different inhibitory effect on reactive oxygen species production
Molecules targeting copper ion transport
Metabolic inhibitors and targeted therapies based on tumour metabolic profiles (for example,
DICER, cMYC,98 HIF1, MCT4, and MCT1,149 GLUT1, HKII, phosphoglycerate dehydrogenase,
LDHA,150 6phosphofructo2-kinase (PFKFB3 isoform),151 fatty acid synthase,152 CPT1C153)
P-site of adenylate cyclase21
Other targets specific to glucagon signalling (e.g. glucagon receptor, cells)154
New GLP1 receptor agonists and DPP4 inhibitors (to reduce plasma glucagon levels)
Targeting abnormal metabolism linked to different Gprotein coupled receptors (e.g. hybrid peptides
acting through GIP, glucagon and GLP1 receptors at the same time,155 Gproteincoupled receptor
40 agonist156)
Other targets of insulin signalling (e.g. anti-IGF-1R antibodies or small molecule IGF-1R compounds96)
Other AMPK activators and tissue specific AMPK activators
AMPK inhibitors (for cases where AMPK potentiates tumour metabolism)
Other mTOR inhibitors
Agents targeting appetite and obesity
Anti-inflammatory molecules
New chronotherapeutics (that is, modulators of insulin release to mimic cyclicity; targets against
dysregulated circadian rhythms which may underlie insulin resistance, obesity and can have
association with cancer; for example, CLOCK, BMAL1, PER, CRY)
Targeting complications of other treatments (e.g. steroids157)
Combination therapies (for example, chemotherapeutic agents, other antidiabetics)

mediated by the transcription factor NFB and its down- and type2 diabetes mellitus.21 Molecules that abrogate
stream cytokine interleukin 6 (IL6).122 Interestingly, the cAMP and PKA signalling would bypass the mito
inhibitory effect is more pronounced in cancer stem cells chondrial action of metformin and thus confer a pro-
than noncancer stem cells, as the sensitivity of trans- glycolytic effect without changes in energy charge and
formed cell lines to metformin is determined by the the resulting AMPK activation. The efficacy and safety of
degree of immune-cell-mediated tumour inflammation, such molecules would need to be established. What about
which is reflected by IL6 levels.122 How metformin inhib- the risk of hypoglycaemia or pancreatic cell hyper
its NFB is unclear, but AMPK activation in the pres- plasia, as seen in glucagon knockout mice?57 Could there
ence of a metabolic difference between cancer stem cells be a risk of an undesirable pro-Warburg effect? Could
and noncancer stem cells may play a part.123 The main such molecules affect different enzymatic isoforms of
systemic effect of metformin on cancer is via its bene adenylate cyclase or be tissue-specific? How will speci-
ficial effect on glucose metabolism, whereas the main ficity be established? Glucagon receptor is expressed in
direct effect is via LKB1 and AMPK. More research is various tissues, and the cAMPPKA pathway is involved
needed to dissect the intersection between dysregulated in a plethora of signalling pathways, highlighting the
metabolism and cancer and whether there is a benefit need for a specific targeting strategy. Investigating this
of using metformin as a single agent or incombination. newly proposed pathway beyond hepatoc ytes is of
interest, given that an upregulated, rather than down-
Future therapeutic directions regulated, cAMPPKA pathway, as mediated by GLP1,
The AMP-binding Psite on adenylate cyclase has been is deemed protective of myocardial infarct size. 124
proposed as a new therapeutic target in insulin resistance Metformin-induced activation of AMPK also confers

152 | MARCH 2014 | VOLUME 10  www.nature.com/nrendo


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

cardioprotection;124 however, other frequently prescribed dissociation of AMP from the Psite of adenylate cyclase)
drugs such as statins can activate PKA125 and AMPK126 in and specificity remain undefined.
the heart. Interaction of various pathways and drugs and Laboratory evidence of the antimitotic action of met-
their contribution to the pleiotropic effects attributed to formin is promising, although results from epidemio-
metformin will probably remain a topic of debate in the logical studies remain controversial.80 The combination
future. The knowledge gained from the molecular mecha- of tumour genetics, patient metabolic profile and the cel-
nisms of metformin action could lead to the development lular microenvironment determine the antitumour effect
of novel therapies across multiple fields in medicine with of metformin treatment. Focussed use of metformin to
more suitable pharmacokinetic and pharmacodynamic specifically target metabolic differences between normal
properties (Table3). and abnormal signalling offers a vast, though challenging,
therapeutic potential. The metabolic similarities between
Conclusions activated proinflammatory cells and cancer cells suggest
Metformin acts as a metabolic inhibitor and alters both that metabolic inhibitors may modulate immune cells.
whole-body and cellular energy metabolism. It is pri The immune system could thus be a mediator of some
marily used in patients with type2 diabetes mellitus, of the pleiotropic benefits of metformin. Many details of
and its main mechanism of action in this disease setting metformin action remain to be discovered, and the
is inhibition of hepatic gluconeogenesis. Metformin risk of harm must be considered when designing new
interacts with complex I in the mitochondrial electron metformin-based therapies. Hopefully, the knowledge
transport chain, thereby lowering cellular ATP levels and gained from dissecting the pathways that metformin acts
causing AMP accumulation. AMP binds to the Psite of on will propel the development of multiple noveltherapies.
adenylate cyclase and inhibits its action in response to
glucagon, thereby disrupting downstream cAMPPKA
Review criteria
signalling. As a result, the activity of enzymes of the
gluconeogenic pathway is inhibited in favour of glyco A PubMed database search was performed using the
lysis. This mechanism is probably the main mode through following terms: metformin, gluconeogenesis,
glycogenolysis, glucagon, inflammation, cancer,
which metformin lowers hepatic glucose output.
AMPK, cAMP, mitochondrium, GLP1, liver,
In addition, the reduction in energy charge leads to phenformin, and hypoglycaemia. No restriction was
AMPK activation and downregulation of gluconeogenic placed on the year the paper was published. However,
gene expression. Although AMPK activation is dis- due to the sheer volume of publications relating to this
pensable for the glucose-lowering effect of metformin, topic, spreading over decades, more weight was given to
it probably triggers important insulin-sensitizing and original articles, meta-analysis and reviews published in
lipid-modulating mechanisms. Much of the regulatory the last 5years. The references given in selected papers
components of this model of metformin action, as well as were scrutinised. Manuscripts written in English, French,
Spanish and Czech were considered.
parameters that determine its kinetics (such as the rate of

1. Bailey, C.J. & Day, C. Traditional plant medicines system in animal models of multiple sclerosis. 16. Hermann, L.S. Metformin: a review of its
as treatments for diabetes. Diabetes Care 12, J.Immunol. 182, 80058014 (2009). pharmacological properties and therapeutic
553564 (1989). 9. Giovannucci, E. etal. Diabetes and cancer: use. Diabetes Metab. 5, 233245 (1979).
2. Cicero, A.F., Tartagni, E. & Ertek, S. Metformin aconsensus report. Diabetes Care 33, 17. Shitara, Y. etal. Role of organic cation/carnitine
and its clinical use: new insights for an old drug 16741685 (2010). transporter 1 in uptake of phenformin and
in clinical practice. Arch. Med. Sci. 8, 907917 10. Selvin, E. etal. Cardiovascular outcomes in trials inhibitory effect on complex I respiration in
(2012). of oral diabetes medications: a systematic mitochondria. Toxicol. Sci. 132, 3242 (2013).
3. Salpeter, S.R., Buckley, N.S., Kahn, J.A. review. Arch. Intern. Med. 168, 20702080 18. Nies, A.T., Koepsell, H., Damme, K. &
&Salpeter, E.E. Meta-analysis: metformin (2008). Schwab,M. Organic cation transporters
treatment in persons at risk for diabetes 11. Boussageon, R. etal. Reappraisal of metformin (OCTs,MATEs), invitro and invivo evidence for
mellitus. Am. J.Med. 121, 149157 (2008). efficacy in the treatment of type2 diabetes: the importance in drug therapy. Handb. Exp.
4. Glueck, C.J. etal. Metformin reduces weight, ameta-analysis of randomised controlled trials. Pharmacol. 201, 105167 (2011).
centripetal obesity, insulin, leptin, and low-density PLoSMed. 9, e1001204 (2012). 19. Gunton, J.E., Delhanty, P.J., Takahashi, S. &
lipoprotein cholesterol in nondiabetic, morbidly 12. Hemmingsen, B. etal. Comparison of metformin Baxter, R.C. Metformin rapidly increases insulin
obese subjects with body mass index greater and insulin versus insulin alone for type2 receptor activation in human liver and signals
than 30. Metabolism 50, 856861 (2001). diabetes: systematic review of randomised preferentially through insulin-receptor
5. Gokcel, A. etal. Evaluation of the safety and clinical trials with meta-analyses and trial substrate2.J.Clin. Endocrinol. Metab. 88,
efficacy of sibutramine, orlistat and metformin in sequential analyses. Br. Med. J. 344, e1771 13231332 (2003).
the treatment of obesity. Diabetes Obes. Metab. (2012). 20. Bailey, C.J. & Turner, R.C. Metformin. N.Engl.
4, 4955 (2002). 13. Shaw, R.J. etal. The kinase LKB1 mediates J.Med. 334, 574579 (1996).
6. Kita, Y. etal. Metformin prevents and reverses glucose homeostasis in liver and therapeutic 21. Miller, R.A. etal. Biguanides suppress hepatic
inflammation in a non-diabetic mouse model of effects of metformin. Science 310, 16421646 glucagon signalling by decreasing production of
nonalcoholic steatohepatitis. PLoSONE 7, (2005). cyclic AMP. Nature 494, 256260 (2013).
e43056 (2012). 14. McIntyre, H.D. etal. Metformin increases 22. Alengrin, F., Grossi, G., Canivet, B. &
7. Andrews, M., Soto, N. & Arredondo, M. Effect of insulin sensitivity and basal glucose clearance DolaisKitabgi, J. Inhibitory effects of metformin
metformin on the expression of tumor necrosis in type2 (non-insulin dependent) diabetes on insulin and glucagon action in rat
factor-, Toll like receptors 2/4 and C reactive mellitus. Aust.NZJ.Med. 21, 714719 hepatocytes involve post-receptor alterations.
protein in obese type2 diabetic patients (1991). DiabetesMetab. 13, 591597 (1987).
[Spanish]. Rev. Med. Chil. 140, 13771382 15. Czyzyk, A., Tawecki, J., Sadowski, J., 23. Fulgencio, J.P., Kohl, C., Girard, J. &
(2012). Ponikowska,I. & Szczepanik, Z. Effect of Pegorier,J.P. Effect of metformin on fatty acid
8. Nath, N. etal. Metformin attenuated the biguanides on intestinal absorption of glucose. and glucose metabolism in freshly isolated
autoimmune disease of the central nervous Diabetes 17, 492498 (1968). hepatocytes and on specific gene expression in

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 10 | MARCH 2014 | 153


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

cultured hepatocytes. Biochem. Pharmacol. 62, dipeptidyl peptidase4 inhibitor, and metformin 61. Zang, M. etal. AMP-activated protein kinase is
439446 (2001). on glycemic control in patients with type2 required for the lipid-lowering effect of metformin
24. Consoli, A. & Nurjhan, N. Contribution of diabetes. Diabetes Care 30, 19791987 (2007). in insulin-resistant human HepG2 cells. J.Biol.
gluconeogenesis to overall glucose output in 42. Defronzo, R.A. & Abdul-Ghani, M.A. Preservation Chem. 279, 4789847905 (2004).
diabetic and nondiabetic men. Ann. Med. 22, of -cell function: the key to diabetes prevention. 62. Lochhead, P.A., Salt, I.P., Walker, K.S.,
191195 (1990). J.Clin. Endocrinol. Metab. 96, 23542366 (2011). Hardie,D.G. & Sutherland, C.
25. Stumvoll, M., Nurjhan, N., Perriello, G., Dailey, G. 43. Detaille, D., Guigas, B., Leverve, X., 5aminoimidazole4-carboxamide riboside mimics
& Gerich, J.E. Metabolic effects of metformin in Wiernsperger,N. & Devos, P. Obligatory role of the effects of insulin on the expression of the 2
noninsulindependent diabetes mellitus. N.Engl. membrane events in the regulatory effect of key gluconeogenic genes PEPCK and
J. Med. 333, 550554 (1995). metformin on the respiratory chain function. glucose6phosphatase. Diabetes 49, 896903
26. Lutz, T.A., Estermann, A., Haag, S. & Scharrer, E. Biochem. Pharmacol. 63, 12591272 (2002). (2000).
Depolarization of the liver cell membrane by 44. El-Mir, M.Y. etal. Dimethylbiguanide inhibits cell 63. Foretz, M. etal. Short-term overexpression of a
metformin. Biochim. Biophys. Acta 1513, respiration via an indirect effect targeted on the constitutively active form of AMP-activated
176184 (2001). respiratory chain complex I. J.Biol. Chem. 275, protein kinase in the liver leads to mild
27. Otto, M., Breinholt, J. & Westergaard, N. 223228 (2000). hypoglycemia and fatty liver. Diabetes 54,
Metformin inhibits glycogen synthesis and 45. Owen, M.R., Doran, E. & Halestrap, A.P. 13311339 (2005).
gluconeogenesis in cultured rat hepatocytes. Evidence that metformin exerts its anti-diabetic 64. He, L. etal. Metformin and insulin suppress
Diabetes Obes. Metab. 5, 189194 (2003). effects through inhibition of complex 1 of the hepatic gluconeogenesis through
28. Fischer, Y., Thomas, J., Rosen, P. & mitochondrial respiratory chain. Biochem. J. phosphorylation of CREB binding protein. Cell
Kammermeier, H. Action of metformin on 348(Pt 3), 607614 (2000). 137, 635646 (2009).
glucose transport and glucose transporter 46. Miller, R.A. & Birnbaum, M.J. An energetic tale 65. Lee, J.M. etal. AMPK-dependent repression of
GLUT1 and GLUT4 in heart muscle cells from of AMPK-independent effects of metformin. hepatic gluconeogenesis via disruption of CREB.
healthy and diabetic rats. Endocrinology 136, J.Clin. Invest. 120, 22672270 (2010). CRTC2 complex by orphan nuclear receptor small
412420 (1995). 47. Zhou, G. etal. Role of AMP-activated protein heterodimer partner. J.Biol. Chem. 285,
29. Fantus, I.G. & Brosseau, R. Mechanism of kinase in mechanism of metformin action. J.Clin. 3218232191 (2010).
action of metformin: insulin receptor and Invest. 108, 11671174 (2001). 66. Caton, P.W. etal. Metformin suppresses hepatic
postreceptor effects invitro and invivo. J.Clin. 48. Logie, L. etal. Cellular responses to the metal- gluconeogenesis through induction of SIRT1 and
Endocrinol. Metab. 63, 898905 (1986). binding properties of metformin. Diabetes 61, GCN5. J.Endocrinol. 205, 97106 (2010).
30. Stith, B.J. etal. The antidiabetic drug metformin 14231433 (2012). 67. Samuel, V.T. etal. Fasting hyperglycemia is not
elevates receptor tyrosine kinase activity and 49. Ouyang, J., Parakhia, R.A. & Ochs, R.S. associated with increased expression of PEPCK
inositol 1,4, 5trisphosphate mass in Xenopus Metformin activates AMP kinase through or G6Pc in patients with Type2 diabetes. Proc.
oocytes. Endocrinology 137, 29902999 inhibition of AMP deaminase. J.Biol. Chem. 286, Natl Acad. Sci. USA. 106, 1212112126
(1996). 111 (2011). (2009).
31. Prager, R. & Schernthaner, G. Insulin receptor 50. Muller, S. etal. Action of metformin on 68. Burgess, S.C. etal. Cytosolic
binding to monocytes, insulin secretion, and erythrocyte membrane fluidity invitro and invivo. phosphoenolpyruvate carboxykinase does not
glucose tolerance following metformin Eur. J.Pharmacol. 337, 103110 (1997). solely control the rate of hepatic
treatment. Results of a double-blind cross-over 51. Quesada, I., Tuduri, E., Ripoll, C. & Nadal, A. gluconeogenesis in the intact mouse liver.
study in typeII diabetics. Diabetes 32, Physiology of the pancreatic -cell and glucagon CellMetab. 5, 313320 (2007).
10831086 (1983). secretion: role in glucose homeostasis and 69. Foretz, M. etal. Metformin inhibits hepatic
32. Jackson, R.A. etal. Mechanism of metformin diabetes. J.Endocrinol. 199, 519 (2008). gluconeogenesis in mice independently of the
action in noninsulindependent diabetes. 52. Herzig, S. etal. CREB regulates hepatic LKB1/AMPK pathway via a decrease in hepatic
Diabetes 36, 632640 (1987). gluconeogenesis through the coactivator PGC1. energy state. J.Clin. Invest. 120, 23552369
33. Giugliano, D. etal. Metformin improves glucose, Nature 413, 179183 (2001). (2010).
lipid metabolism, and reduces blood pressure in 53. Yoon, J.C. etal. Control of hepatic 70. Ros, S. & Schulze, A. Balancing glycolytic flux:
hypertensive, obese women. Diabetes Care 16, gluconeogenesis through the transcriptional the role of 6phosphofructo2-kinase/fructose
13871390 (1993). coactivator PGC1. Nature 413, 131138 (2001). 2,6bisphosphatases in cancer metabolism.
34. United Kingdom Prospective Diabetes Study 54. Altarejos, J.Y. & Montminy, M. CREB and the Cancer Metab. 1, 8 (2013).
(UKPDS). 13: Relative efficacy of randomly CRTC co-activators: sensors for hormonal and 71. Collier, C.A., Bruce, C.R., Smith, A.C.,
allocated diet, sulphonylurea, insulin, or metabolic signals. Nat. Rev. Mol. Cell Biol. 12, Lopaschuk, G. & Dyck, D.J. Metformin counters
metformin in patients with newly diagnosed non- 141151 (2011). the insulin-induced suppression of fatty acid
insulin dependent diabetes followed for three 55. Wang, Y. etal. Inositol1,4, 5trisphosphate oxidation and stimulation of triacylglycerol
years. Br. Med. J. 310, 8388 (1995). receptor regulates hepatic gluconeogenesis in storage in rodent skeletal muscle. Am. J.Physiol.
35. Fruehwald-Schultes, B. etal. Metformin does not fasting and diabetes. Nature 485, 128132 Endocrinol. Metab. 291, E182E189 (2006).
adversely affect hormonal and symptomatic (2012). 72. Dresner, A. etal. Effects of free fatty acids on
responses to recurrent hypoglycemia. J.Clin. 56. Johnson, R.A., Yeung, S.M., Stubner, D., glucose transport and IRS1associated
Endocrinol. Metab. 86, 41874192 (2001). Bushfield, M. & Shoshani, I. Cation and structural phosphatidylinositol 3kinase activity. J.Clin.
36. Cho, Y.M. & Kieffer, T.J. New aspects of an old requirements for P site-mediated inhibition of Invest. 103, 253259 (1999).
drug: metformin as a glucagon-like peptide 1 adenylate cyclase. Mol. Pharmacol. 35, 681688 73. Patane, G. etal. Metformin restores insulin
(GLP1) enhancer and sensitiser. Diabetologia (1989). secretion altered by chronic exposure to free
54, 219222 (2011). 57. Gelling, R.W. etal. Lower blood glucose, fatty acids or high glucose: a direct metformin
37. Maida, A., Lamont, B.J., Cao, X. & Drucker, D.J. hyperglucagonemia, and pancreatic cell effect on pancreatic -cells. Diabetes 49,
Metformin regulates the incretin receptor axis hyperplasia in glucagon receptor knockout mice. 735740 (2000).
via a pathway dependent on peroxisome Proc. Natl Acad. Sci. USA 100, 14381443 74. Vincent, M.F., Marangos, P.J., Gruber, H.E. &
proliferator-activated receptor- in mice. (2003). Van den Berghe, G. Inhibition by AICA riboside
Diabetologia 54, 339349 (2011). 58. Hawley, S.A., Gadalla, A.E., Olsen, G.S. & ofgluconeogenesis in isolated rat hepatocytes.
38. Lenhard, J.M., Croom, D.K. & Minnick, D.T. Hardie, D.G. The antidiabetic drug metformin Diabetes 40, 12591266 (1991).
Reduced serum dipeptidyl peptidase-IV after activates the AMP-activated protein kinase 75. Evans, J.M., Donnelly, L.A., Emslie-Smith, A.M.,
metformin and pioglitazone treatments. Biochem. cascade via an adenine nucleotide-independent Alessi, D.R. & Morris, A.D. Metformin and
Biophys. Res. Commun. 324, 9297 (2004). mechanism. Diabetes 51, 24202425 (2002). reduced risk of cancer in diabetic patients.
39. Lindsay, J.R. etal. Inhibition of dipeptidyl 59. Fryer, L.G., Parbu-Patel, A. & Carling, D. The anti- Br.Med. J. 330, 13041305 (2005).
peptidase IV activity by oral metformin in type2 diabetic drugs rosiglitazone and metformin 76. U.S. National Library of Medicine.
diabetes. Diabet. Med. 22, 654657 (2005). stimulate AMP-activated protein kinase through ClinicalTrials.gov [online] (2013).
40. Yasuda, N. etal. Enhanced secretion of glucagon- distinct signaling pathways. J.Biol. Chem. 277, 77. Lee, J.H. etal. The effects of metformin on the
like peptide 1by biguanide compounds. Biochem. 2522625232 (2002). survival of colorectal cancer patients with
Biophys. Res. Commun. 298, 779784 (2002). 60. Hawley, S.A. etal. Calmodulin-dependent protein diabetes mellitus. Int. J.Cancer 131, 752759
41. Goldstein, B.J., Feinglos, M.N., Lunceford, J.K., kinase kinase- is an alternative upstream (2012).
Johnson, J. & Williams-Herman, D.E. Effect of kinase for AMP-activated protein kinase. 78. He, X. etal. Metformin and thiazolidinediones are
initial combination therapy with sitagliptin, a CellMetab. 2, 919 (2005). associated with improved breast cancer-specific

154 | MARCH 2014 | VOLUME 10  www.nature.com/nrendo


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

survival of diabetic women with HER2+ breast 97. Liang, J. etal. The energy sensing LKB1-AMPK chemotherapeutic agents. J.Clin. Endocrinol.
cancer. Ann. Oncol. 23, 17711780 (2012). pathway regulates p27(kip1) phosphorylation Metab. 97, E510E520 (2012).
79. Lega, I.C. etal. Association Between Metformin mediating the decision to enter autophagy or 117. Song, C.W. etal. Metformin kills and
Therapy and Mortality After Breast Cancer: apoptosis. Nat. Cell Biol. 9, 218224 (2007). radiosensitizes cancer cells and preferentially
APopulation-Based Study. Diabetes Care 36, 98. Blandino, G. etal. Metformin elicits anticancer kills cancer stem cells. Sci. Rep. 2, 362 (2012).
30183026 (2013). effects through the sequential modulation of 118. Dong, L. etal. Metformin sensitizes endometrial
80. Suissa, S. & Azoulay, L. Metformin and the risk DICER and cMYC. Nat. Commun. 3, 865 (2012). cancer cells to chemotherapy by repressing
ofcancer: time-related biases in observational 99. Dandapani, M. & Hardie, D.G. AMPK: opposing glyoxalase I expression. J.Obstet. Gynaecol. Res.
studies. Diabetes Care 35, 26652673 (2012). the metabolic changes in both tumour cells and 38, 10771085 (2012).
81. Niraula, S. etal. Influence of concurrent inflammatory cells? Biochem. Soc. Trans. 41, 119. Janjetovic, K. etal. Metformin reduces cisplatin-
medications on outcomes of men with prostate 687693 (2013). mediated apoptotic death of cancer cells through
cancer included in the TAX 327 study. Can. Urol. 100. Warburg, O. Metabolism of tumours. Biochem.Z. AMPK-independent activation of Akt.
Assoc. J. 7, E74E81 (2013). 142, 317333 (1923). Eur.J.Pharmacol. 651, 4150 (2011).
82. Hosono, K. etal. Metformin suppresses 101. Thomas, G.V. etal. Hypoxia-inducible factor 120. Iliopoulos, D., Hirsch, H.A. & Struhl, K. Metformin
colorectal aberrant crypt foci in a short-term determines sensitivity to inhibitors of mTOR in decreases the dose of chemotherapy for
clinical trial. Cancer Prev. Res. (Phila.) 3, kidney cancer. Nat. Med. 12, 122127 (2006). prolonging tumor remission in mouse xenografts
10771083 (2010). 102. Hardie, D.G. & Alessi, D.R. LKB1 and AMPK and involving multiple cancer cell types. Cancer Res.
83. Hadad, S. etal. Evidence for biological effects of the cancer-metabolism linkten years after. 71, 31963201 (2011).
metformin in operable breast cancer: a pre- BMC.Biol. 11, 36 (2013). 121. Hirsch, H.A., Iliopoulos, D., Tsichlis, P.N. &
operative, windowofopportunity, randomized trial. 103. Faubert, B. etal. AMPK is a negative regulator Struhl, K. Metformin selectively targets cancer
Breast Cancer Res. Treat. 128, 783794 (2011). of the Warburg effect and suppresses tumor stem cells, and acts together with chemotherapy
84. Pollak, M.N. Investigating metformin for cancer growth invivo. Cell Metab. 17, 113124 to block tumor growth and prolong remission.
prevention and treatment: the end of the (2013). Cancer Res. 69, 75077511 (2009).
beginning. Cancer Discov. 2, 778790 (2012). 104. Algire, C., Amrein, L., Zakikhani, M., Panasci, L. 122. Hirsch, H.A., Iliopoulos, D. & Struhl, K. Metformin
85. Emami, R.A., Fisel, P., Nies, A.T., Schaeffeler, E. &Pollak, M. Metformin blocks the stimulative inhibits the inflammatory response associated
& Schwab, M. Metformin and cancer: from the effect of a high-energy diet on colon carcinoma with cellular transformation and cancer stem cell
old medicine cabinet to pharmacological pitfalls growth invivo and is associated with reduced growth. Proc. Natl Acad. Sci. USA 110, 972977
and prospects. Trends Pharmacol. Sci. 34, expression of fatty acid synthase. Endocr. Relat. (2013).
126135 (2013). Cancer 17, 351360 (2010). 123. Anastasiou, D. Metformin: a case of divide and
86. Nies, A.T., Damme, K., Schaeffeler, E. & 105. Buzzai, M. etal. Systemic treatment with the conquer. Breast Cancer Res. 15, 306 (2013).
Schwab,M. Multidrug and toxin extrusion antidiabetic drug metformin selectively impairs 124. Ye, Y., Perez-Polo, J.R., Aguilar, D. & Birnbaum, Y.
proteins as transporters of antimicrobial drugs. p53-deficient tumor cell growth. Cancer Res. 67, The potential effects of anti-diabetic medications
Expert Opin. Drug Metab. Toxicol. 8, 15651577 67456752 (2007). on myocardial ischemia-reperfusion injury.
(2012). 106. Huang, X. etal. Important role of the LKB1-AMPK BasicRes. Cardiol. 106, 925952 (2011).
87. Gale, E.A. Insulin glargine and cancer: another pathway in suppressing tumorigenesis in PTEN- 125. Manickavasagam, S. etal. The cardioprotective
side to the story? Lancet 374, 521 (2009). deficient mice. Biochem. J. 412, 211221 effect of a statin and cilostazol combination:
88. Tomimoto, A. etal. Metformin suppresses (2008). relationship to Akt and endothelial nitric oxide
intestinal polyp growth in ApcMin/+ mice. 107. Jeon, S.M., Chandel, N.S. & Hay, N. AMPK synthase activation. Cardiovasc. Drugs Ther. 21,
CancerSci. 99, 21362141 (2008). regulates NADPH homeostasis to promote 321330 (2007).
89. Pierotti, M.A. etal. Targeting metabolism for tumour cell survival during energy stress. Nature 126. Hermida, N. etal. HMGCoA reductase inhibition
cancer treatment and prevention: metformin, an 485, 661665 (2012). reverses myocardial fibrosis and diastolic
old drug with multi-faceted effects. Oncogene 32, 108. Mendoza, E.E. etal. Control of glycolytic flux by dysfunction through AMP-activated protein
14751487 (2013). AMP-activated protein kinase in tumor cells kinase activation in a mouse model of metabolic
90. Bar-Peled, L., Schweitzer, L.D., Zoncu, R. & adapted to low pH. Transl. Oncol. 5, 208216 syndrome. Cardiovasc. Res. 99, 4454 (2013).
Sabatini, D.M. Ragulator is a GEF for the rag (2012). 127. Goodarzi, M.O. & Bryer-Ash, M. Metformin
GTPases that signal amino acid levels to 109. Algire, C. etal. Diet and tumor LKB1 expression revisited: re-evaluation of its properties and role
mTORC1. Cell 150, 11961208 (2012). interact to determine sensitivity to anti-neoplastic in the pharmacopoeia of modern antidiabetic
91. Ece, H. etal. Use of oral antidiabetic drugs effects of metformin invivo. Oncogene 30, agents. Diabetes Obes. Metab. 7, 654665
(metformin and pioglitazone) in diabetic patients 11741182 (2011). (2005).
with breast cancer: how does it effect serum 110. Kalender, A. etal. Metformin, independent of 128. Sogame, Y., Kitamura, A., Yabuki, M. & Komuro, S.
Hif1 and 8Ohdg levels? Asian Pac. J.Cancer AMPK, inhibits mTORC1 in a rag GTPase- A comparison of uptake of metformin and
Prev. 13, 51435148 (2012). dependent manner. Cell Metab. 11, 390401 phenformin mediated by hOCT1 in human
92. Wang, Y. etal. Effects of antidiabetic drug (2010). hepatocytes. Biopharm. Drug Dispos. 30,
metformin on human breast carcinoma cells with 111. Efeyan, A. etal. Regulation of mTORC1 by the 476484 (2009).
different estrogen receptor expressing invitro. Rag GTPases is necessary for neonatal 129. Segal, E.D. etal. Relevance of the OCT1
Xi.Bao. Yu Fen. Zi. Mian. Yi. Xue. Za Zhi. 27, autophagy and survival. Nature 493, 679683 transporter to the antineoplastic effect of
253256 (2011). (2013). biguanides. Biochem. Biophys. Res. Commun.
93. Inoki, K., Li, Y., Zhu, T., Wu, J. & Guan, K.L. 112. Zhu, Z., Jiang, W., Thompson, M.D., 414, 694699 (2011).
TSC2is phosphorylated and inhibited by Akt McGinley,J.N. & Thompson, H.J. Metformin as 130. Fasshauer, M., Paschke, R. & Stumvoll, M.
andsuppresses mTOR signalling. Nat. Cell Biol. an energy restriction mimetic agent for breast Adiponectin, obesity, and cardiovascular disease.
4, 648657 (2002). cancer prevention. J. Carcinog. 10, 17 (2011). Biochimie 86, 779784 (2004).
94. Gwinn, D.M. etal. AMPK phosphorylation of 113. Algire, C. etal. Metformin reduces endogenous 131. Sogame, Y., Kitamura, A., Yabuki, M., Komuro, S.
raptor mediates a metabolic checkpoint. Mol. Cell reactive oxygen species and associated DNA & Takano, M. Transport of biguanides by human
30, 214226 (2008). damage. Cancer Prev. Res. (Phila.) 5, 536543 organic cation transporter OCT2.
95. Ning, J. & Clemmons, D.R. AMP-activated protein (2012). Biomed.Pharmacother. 67, 425430 (2013).
kinase inhibits IGFI signaling and protein 114. Zhou, K. etal. Common variants near ATM are 132. Gan, S.C., Barr, J., Arieff, A.I. & Pearl, R.G.
synthesis in vascular smooth muscle cells via associated with glycemic response to metformin Biguanide-associated lactic acidosis. Case report
stimulation of insulin receptor substrate 1 S794 in type2 diabetes. Nat. Genet. 43, 117120 and review of the literature. Arch. Intern. Med.
and tuberous sclerosis 2 S1345 (2011). 152, 23332336 (1992).
phosphorylation. Mol. Endocrinol. 24, 115. Vazquez-Martin, A., Oliveras-Ferraros, C., Cufi, S., 133. Lemke, T.L. & Williams, D.A. Foyes Principles of
12181229 (2010). Martin-Castillo, B. & Menendez, J.A. Metformin Medicinal Chemistry. 5th edn (Lippincott, Williams
96. Vazquez-Martin, A., Oliveras-Ferraros, C., activates an ataxia telangiectasia mutated (ATM)/ and Wilkins, 1995).
Del,B.S., Martin-Castillo, B. & Menendez, J.A. Chk2-regulated DNA damage-like response. 134. electronic Medicines Compendium (eMC). SPC
Ifmammalian target of metformin indirectly is CellCycle 10, 14991501 (2011). Glucophage 500mg and 850mg film coated
mammalian target of rapamycin, then the insulin- 116. Chen, G., Xu, S., Renko, K. & Derwahl, M. tablets [online], http://www.medicines.org.uk/
like growth factor1 receptor axis will audit the Metformin inhibits growth of thyroid carcinoma emc/medicine/1043 (2010).
efficacy of metformin in cancer clinical trials. cells, suppresses self-renewal of derived cancer 135. Luft, D., Schmulling, R.M. & Eggstein, M.
J.Clin. Oncol. 27, e207e209 (2009). stem cells, and potentiates the effect of Lactic acidosis in biguanide-treated diabetics:

NATURE REVIEWS | ENDOCRINOLOGY VOLUME 10 | MARCH 2014 | 155


2014 Macmillan Publishers Limited. All rights reserved
REVIEWS

areview of 330 cases. Diabetologia 14, 7587 153. Zaugg, K. etal. Carnitine palmitoyltransferase 171. Chakrabarti, R., Hocking, E.D. & Fearnley, G.R.
(1978). 1C promotes cell survival and tumor growth Fibrinolytic effect of metformin in coronary-artery
136. Kajbaf, F., Arnouts, P., de Broe,M. & Lalau, J.D. under conditions of metabolic stress. disease. Lancet 2, 256259 (1965).
Metformin therapy and kidney disease: GenesDev. 25, 10411051 (2011). 172. Nagi, D.K. & Yudkin, J.S. Effects of metformin
a review of guidelines and proposals for 154. DAlessio, D. The role of dysregulated glucagon on insulin resistance, risk factors for
metformin withdrawal around the world. secretion in type2 diabetes. Diabetes Obes. cardiovascular disease, and plasminogen
Pharmacoepidemiol. Drug Saf. 22, 10271035 Metab. 13 (Suppl. 1), 126132 (2011). activator inhibitor in NIDDM subjects. A study of
(2013). 155. Bhat, V.K., Kerr, B.D., Flatt, P.R. & Gault, V.A. two ethnic groups. Diabetes Care 16, 621629
137. Brown, J.B., Pedula, K., Barzilay, J., Anovel GIP-oxyntomodulin hybrid peptide acting (1993).
Herson,M.K. & Latare, P. Lactic acidosis rates through GIP, glucagon and GLP1 receptors 173. Mather, K.J., Verma, S. & Anderson, T.J.
in type2 diabetes. Diabetes Care 21, exhibits weight reducing and anti-diabetic Improved endothelial function with metformin in
16591663 (1998). properties. Biochem. Pharmacol. 85, type2 diabetes mellitus. J.Am. Coll. Cardiol. 37,
138. UKPDS. Effect of intensive blood-glucose control 16551662 (2013). 13441350 (2001).
with metformin on complications in overweight 156. Ou, H.Y. etal. Multiple mechanisms of GW9508, 174. Matsumoto, K. etal. Metformin attenuates
patients with type2 diabetes (UKPDS 34). a selective G protein-coupled receptor 40 progression of carotid arterial wall thickness in
UKProspective Diabetes Study (UKPDS) Group. agonist, in the regulation of glucose homeostasis patients with type2 diabetes. Diabetes Res. Clin.
Lancet 352, 854865 (1998). and insulin sensitivity. Am. J. Physiol. Endocrinol. Pract. 64, 225228 (2004).
139. Cubeddu, L.X. etal. Effects of metformin on Metab. 304, E668E676 (2013). 175. Preiss, D. etal. Metformin for non-diabetic
intestinal 5hydroxytryptamine (5-HT) release 157. Christ-Crain, M. etal. AMP-activated protein patients with coronary heart disease (the
and on 5HT3 receptors. Naunyn Schmiedebergs kinase mediates glucocorticoid-induced CAMERA study): a randomised controlled trial.
Arch. Pharmacol. 361, 8591 (2000). metabolic changes: a novel mechanism in Lancet Diabet. Endocrinol. http://dx.doi.org/
140. Donnelly, L.A., Morris, A.D. & Pearson, E.R. Cushings syndrome. FASEB J. 22, 16721683 10.1016/S2213-8587(13)70152-9.
Adherence in patients transferred from (2008). 176. Arai, M. etal. Metformin, an antidiabetic agent,
immediate release metformin to a sustained 158. Lv, W.S. etal. The effect of metformin on food suppresses the production of tumor necrosis
release formulation: a population-based study. intake and its potential role in hypothalamic factor and tissue factor by inhibiting early growth
Diabetes Obes. Metab. 11, 338342 (2009). regulation in obese diabetic rats. Brain Res. response factor1 expression in human
141. Appleyard, M.V. etal. Phenformin as prophylaxis 1444, 1119 (2012). monocytes invitro. J.Pharmacol. Exp. Ther. 334,
and therapy in breast cancer xenografts. 159. Stevanovic, D. etal. Intracerebroventricular 206213 (2010).
Br.J.Cancer 106, 11171122 (2012). administration of metformin inhibits ghrelin- 177. de Jager,J. etal. Effects of short-term treatment
142. Dilman, V.M. etal. Metabolic induced Hypothalamic AMP-kinase signalling and with metformin on markers of endothelial function
immunodepression and metabolic food intake. Neuroendocrinol. 96, 2431 (2012). and inflammatory activity in type2 diabetes
immunotherapy: an attempt of improvement in 160. Yang, Y., Atasoy, D., Su, H.H. & Sternson, S.M. mellitus: a randomized, placebo-controlled trial.
immunologic response in breast cancer patients Hunger states switch a flip-flop memory circuit J.Intern. Med. 257, 100109 (2005).
by correction of metabolic disturbances. via a synaptic AMPK-dependent positive 178. Hattori, Y., Suzuki, K., Hattori, S. & Kasai, K.
Oncology 39, 1319 (1982). feedback loop. Cell 146, 9921003 (2011). Metformin inhibits cytokine-induced nuclear
143. Pollak, M. Potential applications for biguanides 161. Seo, S., Ju, S., Chung, H., Lee, D. & Park, S. factor B activation via AMP-activated protein
in oncology. J.Clin. Invest. 123, 36933700 Acute effects of glucagon-like peptide1 on kinase activation in vascular endothelial cells.
(2013). hypothalamic neuropeptide and AMP activated Hypertension 47, 11831188 (2006).
144. Tzvetkov, M.V. etal. The effects of genetic kinase expression in fasted rats. Endocr. J. 55, 179. Isoda, K. etal. Metformin inhibits
polymorphisms in the organic cation 867874 (2008). proinflammatory responses and nuclear factor-B
transporters OCT1, OCT2, and OCT3 on the renal 162. Andrews, Z.B. etal. UCP2 mediates ghrelins in human vascular wall cells. Arterioscler. Thromb.
clearance of metformin. Clin. Pharmacol. Ther. action on NPY/AgRP neurons by lowering free Vasc. Biol. 26, 611617 (2006).
86, 299306 (2009). radicals. Nature 454, 846851 (2008). 180. Pearce, E.L. etal. Enhancing CD8 Tcell memory
145. Chen, L. etal. Role of organic cation transporter3 163. Kola, B. etal. Cannabinoids and ghrelin have by modulating fatty acid metabolism. Nature 460,
(SLC22A3) and its missense variants in the both central and peripheral metabolic and 103107 (2009).
pharmacologic action of metformin. cardiac effects via AMP-activated protein kinase. 181. Yuan, H. etal. Antidiabetic drug metformin
Pharmacogenet. Genomics 20, 687699 (2010). J.Biol. Chem. 280, 2519625201 (2005). alleviates endotoxin-induced fulminant liver injury
146. Nakamichi, N. etal. Involvement of carnitine/ 164. Chau-Van, C., Gamba, M., Salvi, R., in mice. Int. Immunopharmacol. 12, 682688
organic cation transporter OCTN1/SLC22A4 in Gaillard,R.C. & Pralong, F.P. Metformin inhibits (2012).
gastrointestinal absorption of metformin. adenosine 5'monophosphateactivated kinase 182. Liu, L., Zhang, C., Hu, Y. & Peng, B. Protective
J.Pharmacol. Sci. 102, 34073417 (2013). activation and prevents increases in effect of metformin on periapical lesions in rats
147. Becker, M.L. etal. Genetic variation in the neuropeptide Y expression in cultured by decreasing the ratio of receptor activator of
multidrug and toxin extrusion 1 transporter hypothalamic neurons. Endocrinology 148, nuclear factor B ligand/osteoprotegerin.
protein influences the glucose-lowering effect 507511 (2007). J.Endod. 38, 943947 (2012).
ofmetformin in patients with diabetes: 165. Lee, C.K. etal. Intracerebroventricular injection 183. Kalariya, N.M., Shoeb, M., Ansari, N.H.,
apreliminary study. Diabetes 58, 745749 of metformin induces anorexia in rats. Srivastava, S.K. & Ramana, K.V. Antidiabetic
(2009). DiabetesMetab. J. 36, 293299 (2012). drug metformin suppresses endotoxin-induced
148. Zhou, M., Xia, L. & Wang, J. Metformin transport 166. Hermann, L.S. etal. Therapeutic comparison of uveitis in rats. Invest. Ophthalmol. Vis. Sci. 53,
by a newly cloned proton-stimulated organic metformin and sulfonylurea, alone and in various 34313440 (2012).
cation transporter (plasma membrane combinations. A double-blind controlled study. 184. Kang, K.Y. etal. Metformin downregulates Th17
monoamine transporter) expressed in human Diabetes Care 17, 11001109 (1994). cells differentiation and attenuates murine
intestine. Drug Metab. Dispos. 35, 19561962 167. Chu, N.V. etal. Differential effects of metformin autoimmune arthritis. Int. Immunopharmacol. 16,
(2007). and troglitazone on cardiovascular risk factors in 8592 (2013).
149. Morris, J.C. etal. Targeting hypoxic tumor cell patients with type2 diabetes. Diabetes Care 25, 185. Park, C.S. etal. Metformin reduces airway
viability with carbohydrate-based carbonic 542549 (2002). inflammation and remodeling via activation
anhydrase IX and XII inhibitors. J.Med. Chem. 168. Gomez-Diaz, R.A. etal. Metformin decreases ofAMP-activated protein kinase.
54, 69056918 (2011). plasma resistin concentrations in pediatric Biochem.Pharmacol. 84, 16601670 (2012).
150. Hamanaka, R.B. & Chandel, N.S. Targeting patients with impaired glucose tolerance:
glucose metabolism for cancer therapy. J. Exp. aplacebo-controlled randomized clinical trial. Acknowledgements
Med. 209, 211215 (2012). Metabolism 61, 12471255 (2012). We would like to thank Professor A. B. Grossman,
151. Clem, B.F. etal. Targeting 6Phosphofructo2- 169. Singh, S., Akhtar, N. & Ahmad, J. Plasma Oxford Centre for Diabetes, Endocrinology and
Kinase (PFKFB3) as a Therapeutic Strategy adiponectin levels in women with polycystic Metabolism, for his expert review of this manuscript.
against Cancer. Mol. Cancer Ther. 12, ovary syndrome: impact of metformin treatment I.Pernicova is supported by a Project Grant from the
14611470 (2013). in a case-control study. Diabetes Metab. Syndr. 6, Barts Charity.
152. Zhou, W. etal. Fatty acid synthase inhibition 207211 (2012).
triggers apoptosis during S phase in human 170. Fanghanel, G. etal. Effects of metformin on Author contributions
cancer cells. Cancer Res. 63, 73307337 fibrinogen levels in obese patients with type2 The authors contributed equally to all aspects
(2003). diabetes. Rev. Invest. Clin. 50, 389394 (1998). ofthemanuscript.

156 | MARCH 2014 | VOLUME 10  www.nature.com/nrendo


2014 Macmillan Publishers Limited. All rights reserved

Vous aimerez peut-être aussi