Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

diabetes research and clinical practice 160 (2020) 108025

Contents available at ScienceDirect

Diabetes Research
and Clinical Practice
journal homepage: www.elsevier.com/locat e/dia bre s

Metformin: An old drug against old age and


associated morbidities

Teresa Salvatore a, Pia Clara Pafundi b, Floriana Morgillo c, Raimondo Di Liello c,


Raffaele Galiero b, Riccardo Nevola b, Raffaele Marfella b, Lucio Monaco b, Luca Rinaldi b,
Luigi Elio Adinolfi b, Ferdinando Carlo Sasso b,*
a
Unit of Internal Medicine, Department of Precision Medicine, University of Campania ‘‘Luigi Vanvitelli”, Via Pansini, 5, 80131 Naples, Italy
b
Unit of Internal Medicine, Department of Advanced Medical and Surgical Sciences, University of Campania ‘‘Luigi Vanvitelli”, Piazza
Miraglia, 2, 80138 Naples, Italy
c
Division of Medical Oncology, Department of Precision Medicine, University of Campania ‘‘Luigi Vanvitelli”, Via Pansini, 5, 80131 Naples,
Italy

A R T I C L E I N F O A B S T R A C T

Article history: Metformin represents a striking example of a ‘‘historical nemesis” of a drug. About 40 years
Received 9 December 2019 after its marketing in Europe, once demonstrated its efficacy and safety, metformin was
Received in revised form registered also in the U.S. A few years later, it has become a mainstay in T2DM treatment,
2 January 2020 according to all international Scientific Societies guidelines.
Accepted 13 January 2020 Today, despite the advent of new innovative drugs, metformin still persists as a first-
Available online 16 January 2020 choice drug in T2DM.
This success is largely justified. In fact, over the years, also positive effects on health
increased. In particular, evidence has been accumulated on a beneficial impact against
Keywords:
many other aging-related morbidities (obesity, metabolic syndrome, cardiovascular dis-
Metformin
ease, cancer, cognitive decline and mortality). This literature review describes preclinical
Aging
and clinical evidence favoring the ‘‘anti-aging” therapeutic potential of metformin outside
Obesity
of T2DM. The rationale to the use of metformin as part of a combined therapy in a variety of
Mortality
clinical settings, allowing for a reduction of the chemotherapy dose in cancer patients, has
Cardiovascular diseases
also been discussed. In particular, the focus was on metformin action on RAS/RAF/MAPK
Cancer
pathway.
In the end, the real challenge for metformin could be to fully demonstrate beneficial
effects on health even in non-diabetic subjects.
Ó 2020 Elsevier B.V. All rights reserved.

* Corresponding authors at: University of Campania ‘‘Luigi Vanvitelli”, Department of Advanced Medical and Surgical Sciences, Italy.
E-mail addresses: teresa.salvatore@unicampania.it (T. Salvatore), piaclara.pafundi@unicampania.it (P.C. Pafundi), floriana.morgil-
lo@unicampania.it (F. Morgillo), diliello90@gmail.com (R. Di Liello), raffaele_ga@outlook.it (R. Galiero), riccardo.nevola@unicampania.it
(R. Nevola), raffaele.marfella@unicampania.it (R. Marfella), monacolucio@live.it (L. Monaco), luca.rinaldi@unicampania.it (L. Rinaldi),
luigielio.adinolfi@unicampania.it (L.E. Adinolfi), ferdinando.sasso@unicampania.it (F.C. Sasso).
https://doi.org/10.1016/j.diabres.2020.108025
0168-8227/Ó 2020 Elsevier B.V. All rights reserved.
2 diabetes research and clinical practice 160 (2020) 108025

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Benefits of metformin on Age-Related morbidities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1. Obesity and associated conditions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.2. Cardiovascular disease and mortality . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3. Anticancer properties. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
4. Cognitive decline . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
5. Anti-Aging potential of metformin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
7. Authors’ contributions statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Declaration of Competing Interest. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
Appendix A. Supplementary material . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8

1. Introduction

Metformin is one of the oldest and most widely prescribed than diabetes, including obesity, metabolic syndrome, cardio-
anti-hyperglycemic drug worldwide. Its history begins in the vascular disease, cancer, cognitive decline and mortality.
1920s, when compounds related to the active agents in the Some of these benefits have been known since long time
Galega officinalis, the French lilac, were accidentally noted (e.g., metabolic disorders and atherosclerotic burden) [3,4].
to have anti-hyperglycemic properties. Firstly introduced in Others have emerged later, i.e. anti-cancer [5] and anti-
Europe in 1957, the drug was registered in U.S. much later, neurodegeneration properties [6]. As a result, metformin
in 1995, after its efficacy and safety were confirmed and pub- appears like a gerontoprotector agent that may substantially
lished. A few years after its introduction into the American extend lifespan and lifetime free from diseases in diabetic
market, metformin has become a mainstay in the treatment subjects taking the drug, even compared with non-diabetic
of type 2 diabetes mellitus (T2DM) thanks to the support of people [7].
UKPDS results [1]. Still today, the guidelines on diabetes man- In this review we provide a brief picture of current preclin-
agement indicate that the drug should be continued as long ical and clinical evidences about the anti-aging therapeutic
as tolerated and not contraindicated, in monotherapy as well potential of metformin outside of T2DM, with a short discus-
as in combination with other oral and subcutaneous antihy- sion on the underlying hypothesized pathophysiological
perglycemic drugs. mechanisms.
Metformin is a multifaceted drug, with multiple sites of
action and molecular mechanisms [2]. Its anti- 2. Benefits of metformin on Age-Related
hyperglycemic effects are mainly produced by the either morbidities
direct or indirect reduction of hepatic glucose production
and, according to several studies, by an increase in insulin 2.1. Obesity and associated conditions
sensitivity of peripheral tissues. Moreover, for time gut has
been recognized as a target organ for metformin where the The excess of body fat must be considered to right reason an
drug increases glucose consumption, stimulates GLP-1 pro- age-related disease as overweight and obesity have been
duction and modifies the microbiome. demonstrated to markedly increase worldwide as individuals
The complex I of the mitochondrial respiratory chain is age [8]. It is widely accepted that obesity increases the risk of
the ‘‘classic” molecular target of metformin. Its inhibition cardiovascular diseases (e.g., atherosclerotic disease, chronic
prevents mitochondrial ATP production and increases cyto- heart failure, and arterial hypertension), type 2 diabetes, var-
plasmic ADP:ATP and AMP:ATP ratios, thus leading to activa- ious types of cancers, and sleep apnea/sleep-disordered
tion of AMP-activated protein kinase (AMPK). AMPK is an breathing. However, whereas weight excess is associated with
important energy sensing enzyme which regulates cellular a higher mortality risk in young and middle-aged individuals,
energy status by enhancing insulin sensitivity (through a similar relationship is not observed in adults aged 60 to
effects on fat metabolism) and lowering cAMP, thus reducing 75 years [9]. This phenomenon, known as the ‘‘obesity sur-
the expression of gluconeogenic enzymes. AMPK- vival paradox”, represents an unresolved matter of debate,
independent mechanisms of action may include inhibition which needs further clarifications, particularly a more accu-
of fructose-1,6-bisphosphatase by AMP. rate risk status stratification based on comorbidities, sex,
Over the years, metformin has been shown to exert an body fat distribution, preserved amount of lean mass, nutri-
increasingly wide range of positive pleiotropic effects on tional status, cardiorespiratory fitness, socioeconomic envi-
health, in addition to those associated with an improvement ronment, etc. [10].
in glycemia. In particular, data has accumulated on a benefi- The belief in a protective role of excess adiposity from
cial impact against many aging-related morbidities other health disorders in the elderly may be a simplistic way of
diabetes research and clinical practice 160 (2020) 108025 3

dealing with the issue if considering that a high risk of dis- obesity and prediabetes. Metformin monotherapy may signif-
ability and falls, as well as a low quality of life, may result icantly improve dyslipidemia on statin-naı̈ve people with
from reduced physical function. In addition, individuals over newly diagnosed T2DM [21] and a meta-analysis suggests that
80 years old are characterized by a high prevalence of ‘‘sar- biguanide could effectively lower systolic blood pressure in
copenic obesity” (8.4% in women and 13.5% in men) [11]. This nondiabetic patients, especially in those with IGT or obesity
clinical condition combining sarcopenia and obesity, and not [22].
easily identifiable by anthropometric measures, is worse than
the other forms of obesity associated with an increased all- 2.2. Cardiovascular disease and mortality
cause mortality [12].
Metformin is one of the few oral agents preventing weight Metformin is also one of the few anti-hyperglycemic drugs
gain or even favoring weight loss in T2DM patients. Due to the with significant results in cardiovascular events reduction
progressive nature of disease, many diabetic subjects will and diabetes-related mortality. These benefits are mostly sup-
need insulin treatment, a regimen that may induce weight ported by the seminal trial UK Prospective Diabetes Study
gain in various ways (anabolic effect of hormone, correction (UKPDS 34), in which a small subgroup of overweight patients
of glycosuria, and increase in dietary intake to prevent/cor- randomized to metformin displayed a risk reduction of 39%
rect episodes of hypoglycemia). In this setting, the HOME trial for nonfatal myocardial infarct, 42% for diabetes-related
has indicated that continuing metformin beyond the initia- death and 36% for all-cause mortality, compared to conven-
tion of insulin therapy may prevent insulin-induced weight tional dietary measures [1]. Remarkably, beneficial effects
gain [13]. were still appreciable throughout the 10-year post-trial mon-
It is usually thought that weight loss associated to met- itoring, despite similarities in glycated hemoglobin levels [23].
formin therapy is determined by a reduction in systemic insu- Besides UKPDS, only two small randomized, placebo-
lin requirement and gastrointestinal side effects such as controlled trial were published. The first conducted on
nausea, diarrhea, and dysgeusia. Currently, emerging evi- insulin-treated diabetic patients showed a 40% reduction of
dence suggests multiple mechanisms underlying this effect macrovascular events with the addition of metformin vs pla-
mediated by modification of gut microbiome and modulation cebo [13]. In the other, T2DM patients with coronary artery
of hypothalamic appetite regulatory centers, both directly or disease (CAD) obtained a 46% reduction of primary cardiovas-
indirectly, i.e. through the gut-brain axis [14]. cular composite endpoint when treated with metformin vs
Some studies only reported in abstract form associate glipizide [24].
metformin treatment with a redistribution of fat from central Solid evidence of cardiovascular protection also derives
to subcutaneous depots and with a reduction in the thickness from large observational studies in people with early- and
of epicardial adipose tissue [15]. late-stage diabetes, showing improved outcomes with met-
The role of metformin as a primary treatment for obesity formin as compared with sulfonylurea monotherapy, both
is still equivocal and its current use as weight loss agent out- for cardiovascular events and all-cause mortality. This advan-
side of T2DM remains off-label. However, the American Dia- tage over sulfonylureas is confirmed by a recently published
betes Association (ADA) suggests taking into consideration updated meta-analysis including over one million patients
the prescription of metformin also to prediabetic patients with CAD, in which a higher reduction in incidence rate of
with a BMI > 35 kg/m2 and/or other additional risk factors cardiovascular events and all-cause and cardiovascular mor-
for diabetes. This recommendation mainly arises from the tality was shown with metformin rather than with sulfony-
impressive findings of the Diabetes Prevention Program lureas [25].
(DPP) [16], a study involving overweight patients with An interesting finding is reported by a retrospective obser-
impaired glucose tolerance (IGT) and fasting plasma glucose vational study on a population of about 80,000 T2DM subjects,
95–125 mg/dL. During the 2-year double-blind period, subjects showing that metformin therapy determined not only a
assigned to the metformin arm experienced significant body longer survival than in diabetic patients not treated with met-
weight and waist circumference reductions, in addition to a formin, as well as in their matched non‐diabetic controls,
31% reduction in T2DM incidence compared to placebo. despite the higher BMI and greater comorbidities at baseline
Patients initially randomized to the drug maintained these [26].
benefits, both for weight loss and diabetes incidence, also Clinical literature is rich in data suggesting a substantial
after a follow-up >15 years [17]. benefit from metformin in chronic heart failure (CHF) popula-
As widely reviewed by Hostalek et al. [18], a strong evi- tion, even on patients with CHF in III and IV NYHA class. A
dence from a number of clinical trials other than DPP, as well retrospective cohort study on 16,417 Medicare beneficiaries
as the clinical experience, support both efficacy and safety with diabetes hospitalized for CHF showed that patients dis-
profiles of metformin for diabetes prevention. To this concern, charged on metformin experimented a reduced mortality
it should be remarked that aging is associated with major (13%) and readmission rates (8%) [27]. In another retrospective
changes in glucose metabolism, with a progressive decline study on a large British database, metformin therapy in out-
in glucose tolerance from the third through the ninth decade patients with newly diagnosed T2DM and CHF significantly
of life [19] and a rise of blood glucose levels per decade after reduced mortality rates with respect to diet and lifestyle
50 years by 0.06 mmol/L at fasting and by 0.05 mmol/L 2- changes [28]. In a systematic review of observational studies
hours after an oral glucose tolerance test [20]. involving 34,000 patients with diabetes and CHF, a 20% lower
Data from the literature support a positive effect of met- mortality in metformin users was recorded, mostly compared
formin even on metabolic syndrome components other than with sulfonylureas [29].
4 diabetes research and clinical practice 160 (2020) 108025

Finally, data collected on T2DM patients with established formin efficacy in reducing both mortality and cardiovascular
atherothrombosis participating to the REACH (Reduction of morbidity in patients with pre-diabetes and established car-
Atherothrombosis for Continued Health) Registry [30] deserve diovascular disease.
to be mentioned. In this study, metformin used in secondary
prevention resulted associated with a 24% lower mortality 3. Anticancer properties
rate. Such a finding was consistent among subsets of patients
in whom biguanide was not recommended, noticeably those Pharmaco-epidemiology has provided several lines of evi-
with a history of congestive heart failure (31%), >65 years dence suggesting a protective role of metformin on malignan-
(23%), and with an estimated creatinine clearance of 30 to cies development, which impacts differently based on the
60 mL/min/1.73 m2 (36%). The REACH registry results have type of cancer, with promising results with some and mini-
thus helped to reconsider the traditional contraindications mal effects on others [37]. Nevertheless, these considerations
to the use of metformin and the concerns about the risk of arise from observational retrospective studies, most of which
lactic acidosis, especially in the elderly [31,32]. deprived of important covariates (e.g., BMI and glycemic con-
Though the extensive literature, as well as the positive trol), and with several methodological limitations. These lat-
results recently reported in landmark large-scale randomized ter, along with an insufficient follow-up in most of cases,
CV outcome trials (CVOT) with glucagon-like peptide-1 recep- render the relationship between metformin and cancer inci-
tor agonists (GLP-1 RAs) and sodium-glucose cotransporter dence still object of debate [38].
type 2 inhibitors (SGLT2), the most recent 2019 ESC guidelines Few groups have studied the antiproliferative effects of
on diabetes, prediabetes and CVD [33] recommend these metformin as single agent in different cancer settings. Anti-
glucose-lowering agents in drug-naı̈ve patients at high/very cancer effects of metformin are associated with both direct
high CV risk instead of metformin. (insulin-independent) and indirect (insulin-dependent)
Metformin would be reserved as first choice only to over- actions of the drug. Two main mechanisms have been pro-
weight T2DM patients without CVD and at moderate CV risk, posed: (i) the correction of hyperinsulinemia and (ii) the
or added in second line only if HbA1c is not at target. Scheen reduced energy consumption in neoplastic cells [37]
suggests several reasons to challenge this downgrading of (Fig. 1). Both mechanisms involve the activation of AMPK,
metformin use, first of all the lack of a meta-analysis on which inhibits the mammalian target of rapamycin (mTOR)
CVOTs of both SGLT2i and GLP-1RAs separating T2DM signaling pathway, whose role in tumorigenesis is well-
patients treated (about three quarters of the enrolled popula- known [39].
tion) or not with metformin as background therapy [34]. In our previous experience, treatment with metformin
Moreover, we cannot ignore the huge amount of data on both resulted in a dose-dependent inhibition of growth in selected
animal and human studies, proving the cardiovascular pro- non-small cell lung cancer (NSCLC) cell lines, which also
tection exerted by metformin. showed a significant reduction in the anchorage-
As recently reviewed by Zilov et al. [15], this beneficial independent colony-forming ability, with a nonsignificant
effect derives from an improvement either of glycemic con- antiproliferative effect in other NSCLC cell lines [40–42]. The
trol and of other traditional risk factors (i.e., body fat and lipid reason for the different response to metformin among the
profile), but also from the correction of a series of disorders NSCLC cell lines examined may depend on LKB1 gene inacti-
such as haemostasis, inflammation and oxidative stress, as vation, a common event in NSCLC, especially in lung adeno-
well as from the direct inhibition of atherosclerotic plaque carcinoma cells [43]. In this sense, loss of function of LKB1
development. In particular, as shown by the major part of may reduce the sensitivity to metformin. However, several
controlled trials on T2DM patients, metformin seems to exert preclinical studies suggest the existence of an AMPK-
a protective effect against endothelial dysfunction which rep- independent mechanism of action of metformin [2].
resents an early step both in the initiation and progression of Several studies have assessed the risk of cancer in diabetic
atherosclerosis. A recently published trial, the CODICE multi- patients, as well as any protective role of metformin on can-
center prospective study, has assessed in prediabetic patients cer development, whilst there is less evidence about how
with stable angina and nonobstructive coronary stenosis the antidiabetic drugs act on the outcome after anticancer ther-
effect of metformin therapy on coronary endothelial function apy. One study by Tan BX et al. on 99 patients with NSCLC
and major adverse cardiac events (MACEs) at 24 months of assessed both metformin and insulin influence on
follow-up [35]. The findings demonstrated a reduction of car- chemotherapy response rates and survival [44]. In particular,
diovascular events of 40% and a significantly lower percent- chemotherapy with metformin produced higher results com-
age of endothelial dysfunction (left anterior descending pared to insulin and other compounds, both in terms of pro-
coronary diameter at baseline and after acetylcholine infu- gression, free-survival and overall survival.
sion) in prediabetics treated with metformin 850 mg twice a Evidence of increased antitumor effects by the concomi-
day as compared with untreated prediabetics. tant treatment with metformin and chemotherapeutics (e.g.,
This topic is as well the aim of an ongoing randomized cisplatin, paclitaxel or doxorubicin) arises from in vivo studies
clinical trial exploring primary prevention of cardiovascular on breast, prostate and lung cancer models [45–48].
events by metformin in people with hyperglycemia below The reason for such a synergism is supported by several
the threshold diagnosis for diabetes [36]. Moreover, the Inves- hypotheses. Metformin has been demonstrated to decrease cel-
tigation of Metformin in Pre-Diabetes on Atherosclerotic Car- lular thymidine phosphorylase (TP) and excision repair cross-
diovascular OuTcomes (VA-IMPACT; https://clinicaltrials.gov/ complementation 1 (ERCC1) protein and mRNA levels by down
ct2/show/NCT02915198) aims to provide insights into met- regulating phosphorylated MEK1-ERK1/2 protein levels, in both
diabetes research and clinical practice 160 (2020) 108025 5

Fig. 1 – Direct cardioprotective effects of metformin. AMPK, adenosine monophosphate activated protein kinase; eNOS,
endothelial nitric oxide synthase; ER, endoplasmic reticulum; HIF-1, hypoxia-inducible factor-1; LKB1, liver kinase B1; mTOR,
mammalian target of rapamycin; NADPH, nicotinamide adenine dinucleotide phosphate; NFkB, nuclear factor-kappa B; ROS,
reactive oxygen species; TGF-b1 transforming growth factor-b1.

a dose- and time-dependent manner [45]. These two enzymes agent metformin treatment could enhance proliferating sig-
are involved in protection from apoptosis and DNA damage nals through the RAS/RAF/MAPK pathway. Such action could
recognition and repair, respectively. Both are considered nega- in turn, induce cell proliferation in those cell lines with con-
tive prognostic factors and negatively correlate with the efficacy stitutively activating Ras mutations. In this scenario, met-
of chemotherapy. Similarly, metformin has been demonstrated formin enhances MEK-Is antitumor activity in human
to enhance paclitaxel cytotoxicity in human lung cancer cells LKB1-wild-type NSCLC cell lines by downregulating GLI1
(H1650 and H1703) by inhibiting the paclitaxel-induced p38 and reducing the NF-kB (p65)-mediated transcription of
MAPK-mediated ERCC1 expression [46,47]. MMP-2 and MMP-9 [49]. Similarly, a recent work by Martin
These observations provide an experimental rationale to et al. showed that melanoma cells driven by oncogenic BRAF
the use of metformin as part of a combined therapy in a vari- are resistant to the growth-inhibitory effects of metformin,
ety of clinical settings, and allows for a reduction of the which in turn increases ERK activity and upregulates the
chemotherapy dose in cancer patients. The evidence of a sig- VEGF-A protein. Likewise to the synergism identified
nificant synergism of metformin with chemotherapy in lung between gefitinib and metformin, Martin et al. also identified
cancer models led to examine the effects of a combined treat- another unexpected ‘synthetic lethality’ whereby metformin
ment of metformin with gefitinib, a selective EGFR tyrosine and VEGF inhibitors cooperate to suppress BRAF mutant
kinase inhibitor (EGFR-TKI) on NSCLC cell lines. To this end, tumor growth [50].
a panel of human NSCLC cell lines with a defined spectrum Actually, it is mandatory to use caution to extrapolate
of sensitivity to EGFR inhibition by gefitinib was used, includ- from in vitro to in vivo effects, due to a potential difference
ing an in vitro model of acquired resistance to gefitinib devel- between high pharmacological concentrations in vitro com-
oped from the sensitive human CALU-3 lung cancer cells [40]. pared to lower physiological concentrations used in clinical
Combination of metformin with gefitinib strongly reduced setting.
proliferation and induced apoptosis of NSCLC cell lines which Moreover, future studies in cancer patients, alongside with
had harbored wild-type LKB1 gene. Such effects were also extensive preclinical investigations, are thus required to bet-
shown in NSCLC cell lines resistant to the EGFR TKI, suggest- ter investigate metformin effect on the RAS/RAF/MAPK path-
ing a potential metformin-induced reversal of resistance to way and the best setting in which to use metformin combined
gefitinib in some cancer cell lines. The effects were associated with molecularly targeted agents.
with a pronounced decrease in the levels of MAPK and Akt
proteins phosphorylation, key intracellular mediators of 4. Cognitive decline
growth factor-activated cell survival and proliferation signals.
The combined treatment also affected mTOR signaling, as In the last decade, emerging evidence has accumulated also
suggested by the sustained inhibition of S6 and p70S6K suggesting an exciting metformin property of counteracting
phosphorylation. neurodegenerative diseases.
To note, single-agent metformin treatment caused an It is reported that higher glucose levels may represent a
unexpected paradoxical increase in activated phosphory- risk factor for dementia, even among persons without dia-
lated MAPK (P-MAPK) levels, as a result of an increased B- betes [51]. On the contrary, individuals affected by Alzhei-
RAF and C-RAF association. This observation could be thera- mer’s disease (AD), the major neurocognitive disorder, are
peutically relevant, since it has been shown that, while characterized by an increased likelihood of T2DM develop-
exerting antiproliferative and proapoptotic effects, single ment [52]. This epidemiological link, along with several bio-
6 diabetes research and clinical practice 160 (2020) 108025

logical and clinical data, have provided convincing evidence enrollment, provided a robust evidence that metformin use
that the pathogenesis of these two disorders may share a ser- for about 6 years is associated with an only modestly lower
ies of common mechanisms (e.g., impaired insulin signaling, (8%) risk of incident dementia than sulfonylurea, but no effect
altered glucose metabolism, inflammation, increased oxida- after 75 years [60]. Other publications report no association
tive stress, and premature senescence), which may represent with metformin therapy or rather an opposite effect of
the rationale for anti-hyperglycemic drugs (e.g., metformin) increasing dementia. In a cohort study from Taiwan’s
use in dementia [53]. National Health Insurance Research Database, metformin
Laboratory research studies on spontaneously obese or users showed, during a 12-yrs follow-up, not only an
high fat fed murine models with insulin resistance report a increased risk for all-cause dementia, but also a higher inci-
reduction by metformin in cognitive deficits [54]. dence of Parkinson’s disease [61].
In humans, although several publications favor an appre- Further large-scale long-term controlled trials are needed
ciable metformin benefit, epidemiological data are still to finally establish an eventual beneficial role of metformin
ambiguous. In a large observational study on about 70.000 in neurodegenerative disorders development.
subjects aged 65 or over, non-dement and non-diabetic at At same time, we also need further investigations into the
baseline, new-onset T2DM was associated with an increased molecular mechanisms possibly mediating this protective
risk of dementia, which resulted weaker in patients treated effect, for which the activation of AMPK-dependent pathways
with metformin rather than other antidiabetic medications with resulting inhibition of mTor and PI3K-Akt signaling, has
[55]. A best neuroprotection offered by metformin vs sulfony- been hypothesized as main actor [6] (Fig. 2). As shown by
lureas is supported by a retrospective study on 28,640 older postmortem brain examination, the AD neuropathological
veterans, new users of metformin or sulfonylureas, showing hallmark is the presence of: (1) extracellular amyloid plaques
a little (11%) though significant lower risk of dementia in resulting from the aggregation of amyloid-b (Ab) peptides; (2)
patients taking biguanide and aged <75 years with no differ- intracellular neurofibrillary tangles (NFT) made by hyper-
ence in patients aged 75 years [56]. In the population- phosphorylated tau protein and (3) neuronal loss [55]. Met-
based Singapore Longitudinal Aging Study, metformin admin- formin may affect all these pathological conditions through
istration in a very low sample size (365 T2DM patients) mechanisms related to its anti-hyperglycemic actions, i.e.
resulted associated with a very impressive lower risk of cogni- correction of insulin-resistance (and therefore of hyperinsu-
tive decline (51%; until 73% for treatment duration >6 years), linemia) and hyperglycemia, as described below.
even after adjusting for vascular and non-vascular risk factors Since the insulin-degrading enzyme acts on both insulin
[57]. The matter of treatment duration has emerged even in a and Ab, but more selective for insulin, high levels of insulin
recently published study on few thousand of elderly veterans in the brain may deprive Ab of its main clearance mechanism
with T2DM in which only a metformin exposure longer than [62]. Again, insulin seems to increase tau phosphorylation
2 years showed a significant beneficial influence [58]. and cleavage, thus leading to NFT formation and accumula-
As suggested by a pilot trial on 80 subjects with amnestic tion, as shown by both in vitro [63] and in vivo experiments
mild cognitive impairment followed for 12 months, met- [64].
formin seems also to improve cognition in either overweight Chronic hyperglycemia is mainly associated with
or obese individuals without treated diabetes, though more advanced glycation end-products (AGEs) accumulation, which
likely affected by prediabetes (HbA1c: 6.1% + 0.8 in 40 met- induces Ab and tau proteins glycation and lead to Ab aggrega-
formin subjects vs. 6.1 + 0.5 in 40 placebo subjects) [59]. On tion and NFT formation [65]. Hyperglycaemia also increases
the other hand, a recent study on a large population free of the production of reactive oxygen and nitrogen species, thus
dementia and anti-hyperglycemic therapy for 2 years before determining oxidative stress. This event negatively reflects on

Fig. 2 – Potential protective mechanisms of metformin against Alzheimer’s disease.


diabetes research and clinical practice 160 (2020) 108025 7

neuronal cells, given the brain high susceptibility to an oxida- 65–79 yrs. were enrolled to evaluate the time until appearance
tive imbalance due to its high-energy demand and oxygen of any ageing-related morbidity by taking metformin.
consumption and abundance of easily peroxidable polyunsat- The recent publication of the results of Metformin in Long-
urated fatty acids [66]. evity Study (MILES; https://clinicaltrials.gov/ct2/show/
Neuronal damage in AD also derives from mitochondrial NCT02915198) provided the first direct evidence that met-
structural and functional abnormalities, neuroinflammation, formin exerts tissue-specific effects on the expression of
and senescence [67]. Even these processes may be relieved metabolic and non-metabolic human genes implicated in
by metformin, as the drug inhibits NF-kB signaling and proin- ageing [72]. Besides this interesting suggestion, the met-
flammatory cytokines [68] and regulates mitochondrial bio- formin anti-aging potential currently remains a highly
genesis and autophagy by mTOR inhibition [54]. As reported debated topic, as it is unclear whether the ability of drug to
in the literature, various others mechanisms may contribute. target multiple pathways of aging reflects downstream conse-
As an example, metformin has been found to remarkably quences of a primary action on a single mechanism or
decrease, via AMPK, the expression and activity of Beta- whether it involves direct effects on multiple aging regulators
secretase 1 protein involved in the cleavages of the amyloid (Fig. 3).
precursor protein and generation of Ab [69], as well as to Some of the nine alterations indicated by López-Otı́n et al.
reduce tau phosphorylation via mTOR/PP2A [70]. as the hallmarks of the aging process (genomic instability,
telomere attrition, epigenetic alterations, loss of proteostasis,
5. Anti-Aging potential of metformin deregulated nutrient sensing, mitochondrial dysfunction, cel-
lular senescence, stem cell exhaustion, and altered intercellu-
Growing experimental and clinical data have recently turned lar communication) may be modulated by metformin [73]. An
the spotlight on metformin as a potential therapeutic agent to example may be represented by cellular senescence, an irre-
slow ageing. versible cellular growth arrest resistant to apoptosis and asso-
The life-extending capability of the drug has been revealed ciated to secretion of a variety of bioactive factors inducing, in
in a number of recent experimental studies on various non- turn, senescence. Metformin has been described to control
diabetic animal models such as rodents and invertebrates, this process through limitation of ceramide harmful effects
i.e. Drosophila melanogaster and mostly Caenorhabditis elegans. [74] or inhibition of genes coding for multiple inflammatory
In humans, the anti-aging effect is reported in a population cytokines [75]. Even if it is only an untested hypothesis, met-
of diabetic subjects taking metformin who exhibits a signifi- formin may exert some of its health-promoting effects
cantly lower (7%) all-cause mortality than non-diabetics [7]. through epigenomic alterations, i.e. by modulation of micro-
The ongoing clinical trial TAME (Targeting Aging with Met- biota, as it has been reported in C. elegans [76] and in many
formin) [71] is testing metformin as a tool to target ageing in human age-related disorders (e.g., T2DM, obesity, and cancer)
non-diabetic people. To this purpose, healthy subjects aged [77].

Fig. 3 – Potential mechanisms of metformin anti-aging properties.


8 diabetes research and clinical practice 160 (2020) 108025

A series of other pathological mechanisms implied in the and healthier without manifesting negative pleiotropic
aging process seem to be modulated by metformin. Inflam- actions as well as patients with or at risk for T2DM. Moreover,
mation may be suppressed by inhibition of NF-jB transcrip- we do not even have reliable biomarkers which could objec-
tion factor via AMPK-dependent and AMPK-independent tively monitor the healthy aging process, and we do not know
pathways [68]. In addition, also autophagy, a form of pro- the optimal anti-aging dosage of the drug.
grammed cell death highly implicated in shortened lifespan Given these ambiguities, further findings from laboratory
and development of aging-associated diseases, may be research and new clinical trials such as TAME [71] could pro-
enhanced [78]. Metformin may even correct the imbalance vide definitive answers to the ongoing debate on metformin
in the amount of endogenous reactive oxygen species pro- role as gerontoprotector. In the meantime, it is worthwhile
duced by mitochondria to antioxidant defense [79], with a to point out that: (1) metformin should be the preferred treat-
resulting decrease of oxidant stress responsible of proteins, ment option for T2DM, even in older patients; (2) the drug
lipids and nucleotide damage. Expression of SIRT1, a member may represent a therapeutic chance even for subjects with
of the class III (NAD+-dependent) histone deacetylases con- prediabetes and (3) an excessive caution due to the fear of
tributing to enhanced healthy aging, is induced by metformin side effects (i.e. lactic acidosis) may deprive a substantial
as a downstream consequence of AMPK mediated NAD+ ele- number of diabetic individuals from its wide potential bene-
vation [80]. fits [32].
To sum up, many of the above described actions could be
traced to the metformin direct inhibitory action on mitochon- 7. Authors’ contributions statement
drial respiratory complex I and elevation of cellular AMP
levels that allosterically primes AMPK activation, a principal Conception and design: TS, FCS. Development of methodol-
energy sensor that switches on catabolic pathways breaking ogy: TS, FCS, PCP. Acquisition of data: RG, LM, RDL, RN. Anal-
down complex molecules to produce more ATP [2]. This is ysis and interpretation of data: PCP. Writing, review, and/or
what most likely happens in Caenorhabditis elegans, where revision of the manuscript: TS, PCP, FM, RDL, RG, RM, LR,
metformin-mediated lifespan extension seems to be geneti- LM, LEA, RN, FCS. Study supervision: TS, FCS, PCP, FM. All
cally dependent upon AMPK and its upstream effects [81]. authors read and approved the final draft of the manuscript.
The inhibition of mTOR complex, another hub for energy
and nutrient sensing that switches on anabolism and Funding
increases cell growth when activated, has been shown to
extend lifespan across multiple model systems [82]. Met- The authors received no funding from an external source.
formin suppresses mTOR function, both through AMPK-
independent [83] or AMPK-dependent mechanisms [84]. Wu Declaration of Competing Interest
et al. recently observed that the drug inhibits mTOR by acting
on the transport through the nuclear pore complex [85], The authors declared that there is no conflict of interest.
which has been shown as increased in aging [86].
An interesting unifying molecular mechanism has been
Appendix A. Supplementary material
suggested by Chen et al. [87]. Based on biochemical and
genetic experiments involving Caenorhabditis elegans and
Supplementary data to this article can be found online at
human cells, the authors found that metformin regulates
https://doi.org/10.1016/j.diabres.2020.108025.
AMPK activation and mTORC1 suppression, two metabolic
pathways that both converge on lysosomes, membrane-
bound cellular organelles involved in nutrient sensing and
R E F E R E N C E S
recycling.

6. Conclusions [1] UK Prospective Diabetes Study Group. Effect of intensive


blood-glucose control with metformin on complications in
Since the latest version of the International Classification of overweight patients with type 2 diabetes (UKPDS 34). UK
Diseases (ICD-11, code ‘Ageing-related’ XT9T) has formally Prospective Diabetes Study (UKPDS) Group. Lancet 1998;352
recognized ageing as a disease, now therapeutic strategies (9131):854–65.
targeting this condition should be developed. [2] Rena G, Hardie DG, Pearson ER. The mechanisms of action of
To this aim, metformin may represent an interesting metformin. Diabetologia 2017;60:1577–85.
[3] Zimmet P, Collier G. Clinical efficacy of metformin against
option, since this drug has long been approved for human
insulin resistance parameters: sinking the iceberg. Drugs
use, with an excellent safety record for over six decades and 1999;58(Suppl 1):21–8.
low costs. Moreover, in patients with T2DM, metformin can [4] Anfossi G, Russo I, Bonomo K, Trovati M. The cardiovascular
be combined with any other anti-hyperglycemic agent, both effects of metformin: further reasons to consider an old drug
orally and parenterally administered, and therefore contin- as a cornerstone in the therapy of type 2 diabetes mellitus.
ued for life, unless contraindications. Curr Vasc Pharmacol 2010 May;8(3):327–37.
[5] Vancura A, Bu P, Bhagwat M, Zeng J, Vancurova I. Metformin
The experimental and clinical data collected till now are
as an Anticancer Agent. Trends Pharmacol Sci 2018;39
very encouraging. However, it remains still unknown whether
(10):867–78. https://doi.org/10.1016/j.tips.2018.07.006.
non-diabetic individuals taking metformin will live longer
diabetes research and clinical practice 160 (2020) 108025 9

[6] Markowicz-Piasecka M, Sikora J, Szydłowska A, Skupień A, [23] Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-
Mikiciuk-Olasik E, Huttunen KM. Metformin – a future year follow-up of intensive glucose control in type 2 diabetes.
therapy for neurodegenerative diseases: theme: drug N Engl J Med 2008;359(15):1577–89.
discovery, development and delivery in Alzheimer’s disease [24] Hong J, Zhang Y, Lai S, Lv A, Su Q, Dong Y, et al. Effects of
guest editor: Davide Brambilla. Pharm Res 2017;34 metformin versus glipizide on cardiovascular outcomes in
(12):2614–27. patients with type 2 diabetes and coronary artery disease.
[7] Campbell JM, Bellman SM, Stephenson MD, Lisy K. Metformin Diabetes Care 2013;36:1304–13.
reduces all-cause mortality and diseases of ageing [25] Han Y, Xie H, Liu Y, Gao P, Yang X, Shen Z. Effect of metformin
independent of its effect on diabetes control: a systematic on all-cause and cardiovascular mortality in patients with
review and meta-analysis. Ageing Res. Rev. 2017;40:31–44. coronary artery diseases: a systematic review and an updated
https://doi.org/10.1016/j.arr.2017.08.003. meta-analysis. Cardiovasc Diabetol. 2019;18(1):96.
[8] GBD 2015 Obesity Collaborators, Afshin A, Forouzanfar MH, [26] Bannister CA, Holden SE, Jenkins-Jones S, Morgan CL, Halcox
Reitsma MB, Sur P, Estep K, Lee A, et al. Health effects of JP, Schernthaner G, et al. Can people with type 2 diabetes live
overweight and obesity in 195 countries over 25 years. N Engl longer than those without? A comparison of mortality in
J Med 2017;377(1):13–27. people initiated with metformin or sulphonylurea
[9] Oreopoulos Antigone, Kalantar-Zadeh Kamyar, Sharma Arya monotherapy and matched, non-diabetic controls. Diabetes
M, Fonarow Gregg C. The obesity paradox in the elderly: Obes Metab 2014;16(11):1165–73.
potential mechanisms and clinical implications. Clin Geriatr [27] Masoudi FA, Inzucchi SE, Wang Y, Havranek EP, Foody JM,
Med 2009;25(4):643–59. https://doi.org/10.1016/j. Krumholz HM. Thiazolidinediones, metformin, and
cger.2009.07.005. outcomes in older patients with diabetes and heart failure:
[10] Carbone S, Lavie CJ. An opposing point of view on the obesity an observational study. Circulation 2005;111(5):583–90.
paradox. Postgrad Med 2019 Jun;131(5):333–4. [28] Aguilar D, Chan W, Bozkurt B, Ramasubbu K, Deswal A.
[11] Morley JE, Baumgartner RN, Roubenoff R, Mayer J, Nair KS. Metformin use and mortality in ambulatory patients with
Sarcopenia. J Lab Clin Med 2001;137:231–43. diabetes and heart failure. Circ Heart Fail. 2011;4(1):53–8.
[12] Yamashita M, Kamiya K, Matsunaga A, Kitamura T, [29] Eurich DT, Weir DL, Majumdar SR, Tsuyuki RT, Johnson JA,
Hamazaki N, Matsuzawa R, et al. Prognostic value of Tjosvold L, et al. Comparative safety and effectiveness of
sarcopenic obesity estimated by computed tomography in metformin in patients with diabetes mellitus and heart
patients with cardiovascular disease and undergoing surgery. failure: systematic review of observational studies involving
J Cardiol 2019 Sep;74(3):273–8. 34,000 patients. Circ Heart Fail. 2013;6(3):395–402.
[13] Kooy A, de Jager J, Lehert P, Bets D, WulffeléMG Donker AJM, [30] Roussel R, Travert F, Pasquet B, Wilson PW, Smith Jr SC, Goto
et al. Long-term effects of metformin on metabolism and S, et al. Reduction of atherothrombosis for continued health
microvascular and macrovascular disease in patients with (REACH) registry investigators. Metformin use and mortality
type 2 diabetes mellitus. Arch Intern Med 2009;169(6):616–25. among patients with diabetes and atherothrombosis. Arch
[14] Yerevanian A, Soukas AA. Metformin: mechanisms in human Intern Med 2010;170:1892–9.
obesity and weight loss. Curr Obes Rep. 2019;8(2):156–64. [31] Scheen AJ, Paquot N. Metformin revisited: a critical review of
[15] Zilov AV, Abdelaziz SI, AlShammary A, Al Zahrani A, Amir A, the benefit-risk balance in at-risk patients with type 2
Assaad Khalil SH, et al. Mechanisms of action of metformin diabetes. Diabetes Metab 2013;39(3):179–90.
with special reference to cardiovascular protection. Diabetes [32] Salvatore T, Pafundi PC, Marfella R, Sardu C, Rinaldi L,
Metab Res Rev. 2019;25:e3173. Monaco L, et al. Metformin lactic acidosis: should we still be
[16] Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, afraid? Diabetes Res Clin Pract 2019 Nov;157:107879).
Lachin JM, Walker EA, et al. Diabetes Prevention Program [33] Cosentino F, Grant PJ, Aboyans V, Bailey CJ, Ceriello A,
Research Group. Reduction in the incidence of type 2 diabetes Delgado V, et al. ESC Guidelines on diabetes, pre-diabetes,
with lifestyle intervention or metformin. N Engl J Med and cardiovascular diseases developed in collaboration with
2002;346(6):393–403. the EASD. Eur Heart J 2019:ehz486. https://doi.org/10.1093/
[17] Diabetes Prevention Program Research Group. Long-term eurheartj/ehz486.
effects of lifestyle intervention or metformin on diabetes [34] Scheen AJ. Challenging 2019 ESC guidelines for the
development and microvascular complications over 15-year management of type 2 diabetes. Diabetes Metab 2019. pii:
follow-up: the Diabetes Prevention Program Outcomes Study. S1262-3636(19)30178-8.
Lancet Diabetes Endocrinol 2015;3(11):866–75. [35] Sardu C, Paolisso P, Sacra C, Mauro C, Minicucci F, Portoghese
[18] Hostalek U, Gwilt M, Hildemann S. Therapeutic use of M, et al. Effects of metformin therapy on coronary
metformin in prediabetes and diabetes prevention. Drugs endothelial dysfunction in patients with prediabetes with
2015;75:1071–94. stable angina and non-obstructive coronary artery stenosis:
[19] Meigs JB, Muller DC, Nathan DM, Blake DR, Andres R. the CODYCE multicenter prospective study. Diabetes Care
Baltimore Longitudinal Study of Aging. The natural history of 2019 Oct;42(10):1946–55.
progression from normal glucose tolerance to type 2 diabetes [36] Griffin SJ, Bethel MA, Holman RR, Khunti K, Wareham N,
in the Baltimore Longitudinal Study of Aging. Diabetes Brierley G, et al. Metformin in non-diabetic hyperglycaemia:
2003;52(6):1475–84. the GLINT feasibility RCT. Health Technol Assess 2018 Apr;22
[20] Scheen AJ. Diabetes mellitus in the elderly: insulin resistance (18):1–64.
and/or impaired insulin secretion? Diabetes Metab [37] Heckman-Stoddard BM, De Censi A, Sahasrabuddhe VV, Ford
2005;31:5S27–34. LG. Repurposing metformin for the prevention of cancer and
[21] Lin SH, Cheng PC, Tu ST, Hsu SR, Cheng YC, Liu YH. Effect of cancer recurrence. Diabetologia 2017;60(9):1639–47.
metformin monotherapy on serum lipid profile in statin- [38] Morgillo F, Sasso FC, Della Corte CM, Festino L, Manzo A,
naı̈ve individuals with newly diagnosed type 2 diabetes Martinelli E, et al. Metformin in lung cancer: rationale for a
mellitus: a cohort study. Peer J. 2018;12(6):e4578. combination therapy. Expert Opin Invest Drugs 2013;22
[22] Zhou L, Liu H, Wen X, Peng Y, Tian Y, Zhao L. Effects of (11):1401–9.
metformin on blood pressure in nondiabetic patients: a [39] Li M, Li X, Zhang H, Lu Y. Molecular mechanisms of
meta-analysis of randomized controlled trials. J Hypertens metformin for diabetes and cancer treatment. Front Physiol
2017;35:18–26. 2018;9:1039.
10 diabetes research and clinical practice 160 (2020) 108025

[40] Morgillo F, Sasso FC, Della Corte CM, Vitagliano D, D’Aiuto E, veterans aged 65 years with diabetes. Neurology 2017;89
Troiani T, et al. Synergistic effects of metformin treatment in (18):1877–85.
combination with gefitinib, a selective EGFR tyrosine kinase [57] Ng TP, Feng L, Yap KB, Lee TS, Tan CH, Winblad B. Long-term
inhibitor, in LKB1 wild-type NSCLC cell lines. Clin Cancer Res metformin usage and cognitive function among older adults
2013;19(13):3508–19. with diabetes. J Alzheimers Dis 2014;41:61–8.
[41] Fasano M, Della Corte CM, Capuano A, Sasso FC, Papaccio F, [58] Shi Q, Liu S, Fonseca VA, Thethi TK, Shi L. Effect of metformin
Berrino L, et al. A multicenter, open-label phase II study of on neurodegenerative disease among elderly adult US
metformin with erlotinib in second-line therapy of stage IV veterans with type 2 diabetes mellitus. BMJ Open. 2019;9(7):
non-small-cell lung cancer patients: treatment rationale and e024954.
protocol dynamics of the METAL trial. Clin Lung Cancer [59] Luchsinger JA, Perez T, Chang H, Mehta P, Steffener J,
2015;16(1):57–9. Pradabhan G, et al. Metformin in amnestic mild cognitive
[42] Morgillo F, Fasano M, Della Corte CM, Sasso FC, Papaccio F, impairment: results of a pilot randomized placebo controlled
Viscardi G, et al. Results of the safety run-in part of the clinical trial. J Alzheimers Dis 2016;51(2):501–14.
METAL (METformin in Advanced Lung cancer) study: a [60] Scherrer JF, Salas J, Floyd JS, Farr SA, Morley JE, Dublin S.
multicentre, open-label phase I-II study of metformin with Metformin and sulfonylurea use and risk of incident
erlotinib in second-line therapy of patients with stage IV dementia. Mayo Clin Proc 2019;94(8):1444–56.
non-small-cell lung cancer. ESMO Open. 2017;2(2):e000132. [61] Kuan YC, Huang KW, Lin CL, Hu CJ, Kao CH. Effects of
[43] Matsumoto S, Iwakawa R, Takahashi K, Kohno T, Nakanishi metformin exposure on neurodegenerative diseases in
Y, Matsuno Y, et al. Prevalence and specificity of LKB1 genetic elderly patients with type 2 diabetes mellitus. Prog Neuro-
alterations in lung cancers. Oncogene 2007;26:5911–8. Psychopharmacol Biol Psychiatry 2017;79:77–83.
[44] Tan BX, Yao WX, Ge J, Peng XC, Du XB, Zhang R, et al. [62] Roriz-Filho J, Sá-Roriz TM, Rosset I, Camozzato AL, Santos
Prognostic influence of metformin as first-line chemotherapy AC, Chaves MLF, et al. brain aging, and cognition. Biochim
for advanced nonsmall cell lung cancer in patients with type Biophys Acta – Mol Basis Dis 2009;1792:432–43.
2 diabetes. Cancer 2011;117:5103–11. [63] Lesort M, Johnson GV. Insulin-like growth factor-1 and
[45] Ko JC, Huang YC, Chen HJ, Tseng SC, Chiu HC, Wo TY, et al. insulin mediate transient site selective increases in tau
Metformin induces cytotoxicity by down-regulating phosphorylation in primary cortical neurons. Neuroscience
thymidine phosphorylase and excision repair cross- 2000;99:305–16.
complementation 1 expression in non-small cell lung cancer [64] Cheng CM, Tseng V, Wang J, Wang D, Matyakhina L, Bondy
cells. Basic Clin Pharmacol Toxicol 2013;113(1):56–65. CA. Tau is hyperphosphorylated in the insulin-like growth
[46] Lin CC, Yeh HH, Huang WL, Yan JJ, Lai WW, Su WP, et al. factor-I null brain. Endocrinology 2005;146:5086–91.
Metformin enhances cisplatin cytotoxicity by suppressing [65] Iannuzzi C, Irace G, Sirangelo I. Differential effects of
Stat3 activity independently of the LKB1-AMPK pathway. Am glycation on protein aggregation and amyloid formation.
J Respir Cell Mol Biol 2013;49(2):241–50. Front Mol Biosci 2014;1:9.
[47] Tseng SC, Huang YC, Chen HJ, Chiu HC, Huang YJ, Wo TY, [66] Mecocci P, Boccardi V, Cecchetti R, Bastiani P, Scamosci M,
et al. Metformin-mediated downregulation of p38 mitogen- Ruggiero C, et al. A long journey into aging, brain aging, and
activated protein kinase-dependent excision repair cross- Alzheimer’s disease following the oxidative stress tracks. J
complementing 1 decreases DNA repair capacity and Alzheimer’s Dis 2018;62:1319–35.
sensitizes human lung cancer cells to paclitaxel. Biochem [67] Kandimalla R, Thirumala V, Reddy PH. Is Alzheimer’s disease
Pharmacol 2013;85:583–94. a Type 3 diabetes? A critical appraisal. Biochim Biophys Acta
[48] Iliopoulos D, Hirsch HA, Struhl K. Metformin decreases the – Mol Basis Dis 2018;1863:1078–89.
dose of chemotherapy for prolonging tumor remission in [68] Saisho Y. Metformin and inflammation: its potential beyond
mouse xenografts involving multiple cancer cell types. glucose-lowering effect. Endocr Metab Immune Disord Drug
Cancer Res 2011;71:3196–201. Targets 2015;15:196–205.
[49] Della Corte CM, Ciaramella V, Di Mauro C, Castellone MD, [69] Hettich MM, Matthes F, Ryan DP, Griesche N, Schroder S, Dorn
Papaccio F, Fasano M, et al. Metformin increases antitumor S, et al. The anti-diabetic drug metformin reduces BACE1
activity of MEK inhibitors through GLI1 downregulation in protein level by interfering with the MID1 complex. PLoS One
LKB1 positive human NSCLC cancer cells. Oncotarget. 2016;7 2014;9:e102420.
(4):4265–78. [70] Kickstein E, Krauss S, Thornhill P, Rutschow D, Zeller R,
[50] Martin MJ, Hayward R, Viros A, Marais R. Metformin Sharkey J, et al. Biguanide metformin acts on tau
accelerates the growth of BRAF V600E-driven melanoma by phosphorylation via mTOR/protein phosphatase 2A (PP2A)
upregulating VEGF-A. Cancer Discov. 2012;2:344–55. signaling. Proc Natl Acad Sci 2010;107:21830–5.
[51] Crane PK, Walker R, Hubbard RA, Li G, Nathan DM, Zeng H, [71] Justice JN, Ferrucci L, Newman AB, Aroda VR, Bahnson JL,
et al. Glucose levels and risk of dementia. N Engl J Med Divers J, et al. (2018) A framework for selection of blood based
2013;369(6):540–8. biomarkers for geroscience-guided clinical trials: report from
[52] Janson J, Laedtke T, Parisi JE, O’Brien Petersen RC, Butler PC. the TAME Biomarkers Workgroup. Geroscience
Increased risk of type 2 diabetes in Alzheimer disease. 2018;40:419–36.
Diabetes 2004;53(2):474–81. [72] Kulkarni AS, Brutsaert EF, Anghel V, Zhang K, Bloomgarden N,
[53] Boccardi V, Murasecco I, Mecocci P. Diabetes drugs in the fight Pollak M, et al. Metformin regulates metabolic and
against Alzheimer’s disease. Ageing Res Rev 2019;54:100936S. nonmetabolic pathways in skeletal muscle and
[54] Rotermund C, Machetanz G, Fitzgerald JC. The therapeutic subcutaneous adipose tissues of older adults. Aging Cell
potential of metformin in neurodegenerative diseases. Front 2018;17(2):e12723. https://doi.org/10.1111/acel.2018.17.issue-
Endocrinol (Lausanne) 2018;19(9):400. 210.1111/acel.12723.
[55] Cheng C, Lin CH, Tsai YW, Tsai CJ, Chou PH, Lan TH. Type 2 [73] Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G.
diabetes and antidiabetic medications in relation to The hallmarks of aging. Cell 2013;153:1194–217.
dementia diagnosis. J Gerontol A Biol Sci Med Sci [74] Jadhav KS, Dungan CM, Williamson DL. Metformin limits
2014;69:1299–305. ceramide-induced senescence in C2C12 myoblasts. Mech
[56] Orkaby AR, Cho K, Cormack J, Gagnon DR, Driver JA. Ageing Dev 2013;134:548–59.
Metformin vs sulfonylurea use and risk of dementia in US
diabetes research and clinical practice 160 (2020) 108025 11

[75] Moiseeva O, Deschenes-Simard X, St-Germain E, Igelmann S, [81] Onken B, Driscoll M. Metformin induces a dietary restriction-
Huot G, Cadar AE, et al. Metformin inhibits the senescence- like state and the oxidative stress response to extend C.
associated secretory phenotype by interfering with IKK/NF- elegans health-span via AMPK, LKB1, and SKN-1. PLoS One
kB activation. Aging Cell 2013;12:489–98. 2010;5:e8758.
[76] Cabreiro F, Au C, Leung KY, Vergara-Irigaray N, Cocheme´ HM, [82] Soukas AA, Hao H, Wu L. Metformin as anti-aging therapy: is
Noori T, et al. Metformin retards aging in C. elegans by it for everyone? Trends Endocrinol Metab 2019. pii: S1043-
altering microbial folate and methionine metabolism. Cell 2760(19)30147-X.
2013;153:228–39. [83] Kalender A, Selvaraj A, Kim SY, Gulati P, Brûlé S, Viollet B,
[77] Hand TW, Vujkovic-Cvijin I, Ridaura VK, Belkaid Y. Linking et al. Metformin, independent of AMPK, inhibits mTORC1 in a
the microbiota, chronic disease, and the immune system. rag GTPase-dependent manner. Cell Metab 2010;11:390–401.
Trends Endocrinol Metab 2016;27:831–43. [84] Howell JJ, Hellberg K, Turner M, Talbott G, Kolar MJ, Ross DS, et al.
[78] Xie Z, Lau K, Eby B, Lozano P, He C, Pennington B, et al. Metformin inhibits hepatic mtorc1 signaling via dose-dependent
Improvement of cardiac functions by chronic metformin mechanisms involving ampk and the tsc complex. Cell Metab
treatment is associated with enhanced cardiac autophagy in 2017;25:463–71.
diabetic OVE26 mice. Diabetes 2011;60:1770–8. [85] Wu L, Zhou B, Oshiro-Rapley N, Li M, Paulo JA, Webster CM, et al.
[79] Batandier C, Guigas B, Detaille D, El-Mir MY, Fontaine E, An ancient, unified mechanism for metformin growth inhibition
Rigoulet M, et al. The ROS production induced by a reverse- in C elegans and cancer. Cell 2016;167(7). pp. 1705–1718 e13.
electron flux at respiratory-chain complex 1 is hampered by [86] D’Angelo MA, Raices M, Panowski SH, Hetzer MW. Age-
metformin. J Bioenerg Biomembr 2006;38:33–4. dependent deterioration of nuclear pore complexes causes a
[80] Cuyàs E, Verdura S, Llorach-Parés L, Fernández-Arroyo S, loss of nuclear integrity in postmitotic cells. Cell 2009;136
Joven J, Martin-Castillo B, et al. Metformin is a direct SIRT1- (2):284–95.
activating compound: computational modeling and [87] Chen J, Chen J, Ou Y, Li Y, Hu S, Shao LW, et al. Metformin
experimental validation. Front Endocrinol (Lausanne) 2018;6 extends C. elegans lifespan through lysosomal pathway. Elife
(9):657. 2017;6(e3126825):71.

You might also like