Journal of Gerontology & Geriatric Medicine Category: Medical Type: Review Article

Metformin Use beyond Diabetes: Reducing Cardiovascular Events in the Healthy Elderly

Dan Xu1*, Jacquita S Affandi2, Timothy Yap1 and Christopher M Reid2
1 Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth, Australia
2 School of Public Health, Curtin University, Perth, Australia

*Corresponding Author(s):
Dan Xu
Curtin Medical School, Faculty Of Health Sciences, Curtin University, Perth, Australia
Tel:+61 892661740,
Email:daniel.xu@curtin.edu.au

Received Date: Jun 18, 2020
Accepted Date: Jun 23, 2020
Published Date: Jun 30, 2020

Abstract

As the global ageing population rises, there is increasing interest and demand for research evaluating anti-ageing strategies. One such strategy involves investigating a drug that may have additional mechanisms and pathways of action to combat ageing - metformin. This common glucose-lowering agent for diabetes has been safe, effective and globally affordable for over 60 years. Research into the use of metformin and its beneficial influence on healthy ageing is currently emerging. Although metformin’s effect on clinical ageing outcomes may be speculative, findings from studies into cellular and animal models and from observational and pilot human studies support its potential beneficial effects on ageing. Ageing has a significant impact on the cardiovascular system and is the leading non-modifiable risk factor for Cardiovascular Disease (CVD). The incidence and prevalence of CVD increases with advancing age, and CVD is the leading cause of death for populations over 65 years of age. However, most CVD prevention research has focused on development of interventions that target “traditional” CV risk factors such as hypertension, hypercholesterolaemia and diabetes. Metformin has been proposed to be an “anti-ageing” drug, based on preclinical experiments with lower-order organisms and numerous retrospective data on beneficial health outcomes for patients with type 2 diabetes. At present, randomised clinical trials to evaluate metformin’s clinical impact on healthy ageing are limited. Here, we review the role of metformin and its potential to reduce cardiovascular events in the healthy elderly.

Keywords

Cardiovascular risk factors; Healthy ageing; metformin

INTRODUCTION

Australia’s ageing population continues to rise from 3.8 million aged over 65 years in 2017, to a projected 8.8 million by 2057 [1]. The combination of a growing ageing population, longer life expectancy and greater prevalence of multiple chronic diseases such as Cardiovascular Disease (CVD), is increasing and contributes significantly to the disease burden in Australia [2]. In 2017-2018, the prevalence of heart, stroke and vascular disease among adults increased with age, affecting more than 1 in 4 of those aged 75 years and over [3]. People with Type 2 Diabetes (T2DM) and pre-diabetes (impaired glucose tolerance and fasting glucose but insufficient for T2DM diagnosis) have also been shown to have an elevated risk of CVD [4]. CVD is the leading cause of death worldwide representing 31% of all deaths, of which 85% are due to heart attack and stroke [5]. In 2014, Americans between 60-79 years of age and over 80 years of age have over 70% prevalence and 80% prevalence rate of CVD respectively [6]. In Australia, CVD continues to be the most expensive disease burden, costing the government almost $8,8 billion in 2017-2018, with hospital admissions constituting more than half of that cost [7]. As the main risk factor for CVD is age, there is increasing interest and demand for research evaluating strategies that address the global burden of ageing. One such strategy involves investigating a drug that may have additional mechanisms and pathways of action to combat ageing - metformin.

The aim of this review is to outline the potential benefits of metformin, beyond glycaemic-control to reduce the burden of ageing and to promote healthy ageing in healthy people without diabetes. Current clinical trials involving metformin will be reviewed, and the feasibility of a large Randomized Clinical Trial (RCT) to examine the role of metformin in primary prevention to reduce cardiovascular events in the healthy elderly will be discussed.

METFORMIN USE AND ITS SAFETY

Metformin has been successfully used for long-term treatment in older adults as both the first-line therapy and prevention for T2DM [8,9]. Metformin can be safely started at 500 mg per day and titrated towards the target dose of 1000 mg twice daily with eGFR is above 45 ml/min/ 1.73m-2. Kidney function needs to be monitored in patients with cessation of metformin when Estimated Glomerular Filtration Rate (eGFR) is below 30 ml/min/ 1.73m-2 and lowering the dose to a maximum of 1000 mg per day if GFR stays above 30 but below 45 ml/min/ 1.73m-2 [10,11]. In terms of serious adverse events, there is ongoing literature debate whether or not metformin is associated with lactic acidosis [12,13]. Nonetheless, the current consensus in clinical practice preventive guideline is to temporarily withhold metformin in the setting of hospitalisation, acute kidney injury, and use of iodinated-contrast procedures and in the setting of acute severe illness with hypoxia following which metformin therapy is re-implemented [14].

Metformin and CVD

Recently, metformin has been proposed as an anti-ageing drug [15]. Research into metformin’s clinical benefits beyond diabetes originated from the observed cardiovascular risk reduction in individuals with diabetes treated with metformin [16]. Since then, there is growing interest and reports regarding its potential clinical benefits beyond glycaemic control. Metformin has a long history of successful use of as the primary drug for the prevention and treatment of T2DM in Australia and globally [17]. Beyond improving glycaemic control, metformin’s effects on clinical ageing outcomes may still be considered speculative, although findings from cellular and animal models, observational and pilot human studies support the existence of beneficial effects on ageing [18]. At present, progress for human research, using randomised clinical trials to evaluate metformin’s clinical impact are underway. Its use has been associated with a significantly decreased risk of Myocardial Infarction (MI), and CVD in populations with T2DM taking metformin, independent of its effects on blood glucose level, and all-cause mortality [19]. Compared with other medications prescribed for T2DM treatment, metformin does not cause weight gain, and is generally associated with a low risk of hypoglycaemia [20]. People living with diabetes and pre-diabetes have both been shown to have an elevated risk of CVD [4], but the effects of metformin on CVD risk in populations without diabetes are unclear.

Investigation of metformin’s mechanisms of action has gained traction due to the protective effects seen in both human and animal studies. As well as the anti-atherogenic effects of metformin [21,22], it has been shown to prevent diabetes-induced oxidative stress [23,24]. The role of metformin in preventing oxidative stress may account for the preventative CVD effects seen in populations with diabetes due to the role of diabetes-related oxidative stress in vascular tissue damage [25]. Metformin has been implicated in the inhibition of the Mechanistic Target of Rapamycin (mTOR) signaling in monocytes pathway [26], reducing cardiac remodeling [27]. Metformin reduces cardiovascular mortality, all-cause mortality and cardiovascular events in Coronary Arterial Disease (CAD) patients [28]. In MI and CAD patients without T2DM, metformin has no significant effect of reducing the incidence of cardiovascular events, although metformin was more effective than sulfonylureas. Clinical evidence shown in patients with diabetes clearly demonstrated the therapeutic benefit of metformin in reducing cardiovascular mortality and morbidity in T2DM patients. However, there is a paucity in research investigating metformin’s beneficial effects in healthy older individuals without diabetes. Except in the Diabetes Prevention Program, participants with higher baseline fasting glucose or glycosylated Haemoglobin (HbA1c) and women with a history of Gestational Diabetes Mellitus (GDM) benefitted the most from a 15-year metformin intervention [29].

A recent cohort study of older veterans with T2DM in the US showed that metformin reduced CVD events among individuals with T2DM [30]. There were CVD risk reductions by 6% among otherwise healthy individuals, by 18% among those at risk of frailty and by 48% among those at high cardiovascular risk. Another recent large double-blind randomised, placebo-controlled trial evaluated the cardio-metabolic effects of metformin in adults with type 1 diabetes (for ≥ 5 years) and high CVD risk, showing 88% of overweight or obesity with an average age of 55.2 ± 8.5 years [31]. After 3 years, there was no difference in the primary outcome of carotid artery Intima-Media Thickness (cIMT), however there were reductions in body weight, LDL-cholesterol, and in atherosclerosis progression, based on maximal cIMT analysis. These findings highlight the potential of metformin for decreasing CVD risk. In a meta-analysis by Han et al., in MI patients and CAD patients without T2DM, metformin had no significant effect of reducing the incidence of cardiovascular events, however metformin was more effective in reducing the incidence of cardiovascular events than sulfonylureas [28,32,33].

Positive effects were seen in dyslipidemia and obese patients when prescribed atorvastatin combined with metformin compared with atorvastatin monotherapy in lowering the rate of obesity and subclinical inflammation [34]. Metformin use also showed positive cardiovascular effects in female patients with symptomatic myocardial ischemia [35], in women with polycystic ovary syndrome [36], and in patients with peripheral arterial disease [37]. Mohan et al., also showed that metformin reduced Left Ventricular (LV) mass indexed to height, improved Systolic Blood Pressure (SBP), reduced oxidative stress and reduced measures of obesity in patients with CAD [38]. Conversely, a more recent study showed no effects on several markers of CVD for patients without diabetes but with high cardiovascular risk [39]. Despite these findings, studies trialing metformin in populations with established CVD have little relevance to primary prevention of CVD due to the cumulative effect of lifestyle factors from a young age that impact on CVD risk in adulthood [40-42] and the comparatively short duration of the trial. The pilot placebo-controlled study of women without diabetes showed that, besides improving variables of vascular function, metformin also improved measures taken during an exercise tolerance test: maximal ST-segment depression, Duke Score and chest-pain incidence [35]. These findings stressed the importance of metformin use as an additional therapy to reduce cardiovascular risk factors [43]. Further research is required to establish the impact of metformin on CVD risk in those without diabetes [39].

DEFINITION OF AGEING IN GENERAL PRACTICE

There are different personal, cultural and societal perspectives on parameters constituting ‘ageing’ and elderly. Chronological age for older adults in the USA is age ≥ 65 years, whereas in Europe and other parts of the world, it is 60 years and older. Generally accepted biological definitions of ageing include ‘the reduced capacity to regenerate damaged tissue’ [44] and ‘a deficit in maintaining homeostatic processes over time, leading to functional decline and increased risk for disease and death’ [45]. Ageing, insulin resistance and inflammation are associated with the pathogenesis of non-communicable diseases including T2DM [46], CVD [47], cancer [48], depression [49], dementia [50] and frailty [51], a condition of increased vulnerability and adverse health outcomes. The prognosis of an elderly with multiple comorbidity is poor due to functional, psychological and social issues. As our society has a rising elderly population, there is growing interest for research addressing interventions beyond healthy lifestyle to extend the number of functional years. 

Age-dependent (physiological ageing) and age-related diseases (pathological ageing) in healthy ageing 

In order to better understand the healthy ageing process and how to invest in it, it is important to highlight the difference between age-dependent and age-related diseases. Age-dependent diseases include CAD, cerebrovascular disease, T2DM, osteoporosis and Alzheimer’s disease. Its pathogenesis appears to involve physiological ageing processes, chronic damage from inflammation [52] and metabolic syndrome [53]. Mortality and morbidity in these diseases increase exponentially with advanced age. In contrast, age-related diseases have a temporal relationship with age and are not necessarily related to the physiological ageing process. A recent review [54] raised the debate on whether ageing is a disease in itself, suggesting that physiological ageing is indistinguishable from pathology [55] whereas other studies argue that ageing differs from age-related diseases and other pathologies [56,57]. The answer to this question will have important theoretical and practical consequences with various interventions capable of decelerating the ageing process [58,59]. If ageing is treated as a disease, treatment and intervention including lifestyle modifications, drugs and medical treatments that counteract the mechanisms of ageing may delay frailty for decades, ideally until the apparent inevitable limit of human lifespan is reached [60]. The more radical approach is to rejuvenate human tissues, organs, and whole body in order to overtake the above-mentioned limits of human lifespan [60].

Mechanism of ageing

The process of ageing is complex and multifactorial. Indeed, physiological and evolutionary theories have been suggested in order to deduce the mechanisms of ageing [61]. DNA damage is one such mechanism that has received the most attention in order to identify pathways that can be contained or modified to halt or delay ageing itself. Endogenous sources for DNA damage include Reactive Oxygen Species (ROS), alkylation and hydrolysis, whereas exogenous sources include chemicals, Ultraviolet (UV) and other forms of radiation [62]. Oxidative stress, a process where free radicals cause DNA damage and impairment in proteins and lipids translation, provides another mechanism of ageing via genetic damage [63]. Most ageing research comes from animal experiments attempting to expand lifespan. Indeed, a novel CRISPR/Cas9 genome-editing therapy that can suppress the accelerated ageing observed in mice with Hutchison-Gilford progeria syndrome, a rare genetic disorder that also afflicts humans, has been put forward. This treatment provides important insight into new molecular pathways involved in accelerated ageing, as well as how to reduce toxins via gene therapy [64]. 

The evolutionary theory of ageing assumes a linear increase in mutations over time whilst ageing and death are initially circumvented by cellular redundancy mechanisms [65], as mutations overwhelm the system with ineffective protein translation resulting in ageing. From the cellular level, this leads to organ dysfunction, causing diabetes, cardiovascular diseases, neurodegenerative diseases, chronic inflammatory diseases leading to early frailty, delirium and falls, indicated that ageing and pathologies share the same common mechanism [54].

The impact of metformin on mechanisms related to the ageing process 

The mechanisms by which metformin impact on glucose homeostasis includes the non-competitive inhibition of the mitochondrial glycerophosphate dehydrogenase enzyme, which alters hepatocellular redox state, thus reducing the conversion of lactate and glycerol to glucose, decreasing hepatic gluconeogenesis [66], altering mechanisms related to ageing. Furthermore, it has been well documented that metformin mildly reduces levels of High-Sensitivity C-Reactive Protein (hsCRP) [67], and improves endothelial function [68,69], enhancing the benefits against the ageing process. Anti-diabetic activity of metformin may be potentiated via action on the enterocytes and enteroendocrine cells [70]. Metformin has been implicated as the antagonist of the gut hormone Glucagon-Like with Peptide-1 (GLP-1) in patients with T2DM [71]. In addition to GLP-1, two other hormones including Peptide YY (PYY) and Growth/Differentiation Factor 15 (GDF15) are increased significantly on metformin therapy, GDF15 being produced mainly in the intestine via the “integrated stress response” [72]. GDF15 was positively associated with the incidence of diabetes mellitus in the longitudinal Malmo Diet and Cancer-Cardiovascular cohort [73]. Metformin exerts its anti-diabetic effects by increasing the activity of Adenosine Monophosphate-Activated Protein Kinase (AMPK) and mediating the mitochondrial complex I receptor for the anti-diabetic action of metformin [74].

Psychological health including depression and anxiety

A placebo-controlled Chinese study by Guo et al., with participants with T2DM and mild to moderate depression, showed metformin improved depressive symptoms, possibly related to better glycemic control [75]. The outcome of this study is clinically relevant for clinician to be proactive in view of high prevalence of depression in the elderly with or without diabetes. Another case-control study in China examining elderly patients with diabetes using geriatric depression scale concluded that overweight status, poor physical capabilities and low activity level, and the presence of more than two additional illnesses were risk factors for depression, and metformin was a protective factor against depression in elderly patients with diabetes [76]. In light of the anti-depressive effects of metformin in the Chinese population, it will be prudent to examine whether metformin will have similar effects in the western population or different cultural groups. 

There is no direct evidence in human regarding the effects of metformin on anxiety. An interesting mice study suggest that metformin may act by decreasing circulating Branched-Chain Amino Acids (BCAAs) levels to favour serotonergic neurotransmission in the hippocampus and promote anti-anxiety effects in mice fed a High-Fat Diet (HFD) [77]. These findings can potentially translate into clinical practice that a diet poor in BCAAs with metformin use or as add-on therapy to conventional anti-anxiety drugs could help to relieve anxiety symptoms in patients with metabolic comorbidities. It will be interesting to explore whether there is any role of metformin in relieving anxiety symptoms in the elderly as part of healthy ageing coping strategies.

Mild cognitive impairment and dementia

Metformin has been shown in many observational studies to lead to reductions in Mild Cognitive Impairment (MCI) [78] and dementia [79,80] among participants with diabetes taking metformin when compared with no medication or other glucose-lowering agents. A study in Taiwanese individuals aged ≥ 50 years showed that metformin use significantly decreased the risk of dementia compared with no medication (Hazard Ratio [HR] 0.76 [95% Confidence Interval {CI} 0.58-0.98]) [75]. Another evaluation study examined data from 365 individuals from the Singapore Longitudinal Ageing Study (aged ≥ 55 years) to demonstrate that metformin use was associated with lower risk of MCI (Odds Ratio [OR] 0.49 [CI 0.25, 0.95]) [79]. Another study from Taiwan used an insurance database from 67,731 individuals and found that dementia risk was lower in those taking metformin compared with other glucose-lowering medications [80]. It is essential to be aware of the limitations of confounding factors influencing the outcome interpretation of these studies. Despite of the limitations in these studies, a recent clinical trial substantiated the above evidence by showing that metformin improved cognition in individuals without Diabetes [81]. Eighty amnestic MCI participants (aged 55 to 90 years) without treated diabetes were randomly assigned to metformin or placebo for 12 months’ follow-up. Participants on the metformin arm showed improvements in the selective reminding test, even after adjusting for baseline values for the Alzheimer’s Disease Assessment Scale-cognitive subscale (ADAS-cog) score. A recent retrospective cohort study suggested that metformin use in T2DM patients was associated with a significantly lower risk of dementia, especially after use for more than two years [82]. The risk reduction showed a dose-response pattern, was consistent in sensitivity analyses and was not affected by the year of enrollment.

In contrast, an Australian clinical study showed that participants with diabetes (n=126, including 35 metformin users and 91 non-users) displayed worse cognitive performance in the metformin group (OR 2.23, 95% CI 1.05-4.75) [83]. The finding was in consistent with later evidence that metformin may be associated with deleterious effects on cognitive function in the elderly [84]. Researchers in mouse models of ageing reported worsening tau aggregation (causing neurofibrillary degeneration and neurotoxicity associated with the development of Alzheimer’s disease) and abnormal behavior [85] or impaired spatial memory and visual acuity [86]. Furthermore, a recent analysis of patients’ cognitive function 8-10 years after metformin use did not support any benefit of metformin use [87]. The most recent study showed that metformin is associated with a higher risk of vitamin B12 and vitamin B6 deficiency, leading to an increased risk of cognitive dysfunction [88]. Thus, vitamin supplementation is strongly recommended to metformin users.

In summary, although there have been extensive research and clinical trials involving metformin in patients with diabetes, inconsistent dementia risk and cognitive function results warrant more in-depth research. Metformin is relatively safe and would not cause hypoglycemia when used as monotherapy. However, its clinical impact on the prevention of dementia in individuals with and without diabetes warrants a large RCT as part of the healthy ageing strategies.

FUTURE DEVELOPMENTS IN METFORMIN RESEARCH

One of the priorities to reduce healthcare costs in an ageing society is to delay the onset of frailty by identifying risk factors such as cardiovascular and musculoskeletal diseases in the pre-frailty stage. It is well documented that lifestyle modifications including diverse exercise interventions have several advantages with the challenge of adherence over time [89]. Metformin offers a cost-effective alternative besides controlling diabetes or reducing its risk, improving mood and cognitive and physical function. Metformin’s “off-label” use alone or in combination with other anti-diabetics gains popularity for effective prevention of diabetes [90] as well as for management of metabolic syndrome, obesity and polycystic ovarian disease [91,92]. The TAME trial primary endpoints comprise the incidence of MI, congestive heart failure, stroke, most cancers, dementia, and death, but not diabetes and frailty. A clinical trial by Espinoza et al. provides arguments that frailty may be an important endpoint [93]. TAME plans to randomize 3,000 older persons aged between 65-79 years without diabetes including persons with chronic diseases [94] and most likely only persons with impaired glucose tolerance/fasting hyperglycemia. In addition, the TAME biomarker sub-study includes blood-based biomarkers including interleukin- 6, C-reactive protein, insulin-like growth factor 1, insulin, cystatin C, N-terminal prohormone of brain natriuretic peptide, HbA1c, and GDF15, but not GLP-1 [95]. The biomarker workgroup raised important mechanism questions from basic science points of view for TAME and other trials including cancer or neurological diseases impacted by metformin use, in which metformin increases the activity of AMPK and may stimulate autophagy [96-98] or modulate it in leukocytes of T2DM patients [99]. Similar to the pharmaceutical system in the United States, metformin can be listed on our Australian Pharmaceutical Benefits Scheme (PBS) system for the novel indications if the research proves its efficacy. There is also plan from the TAME researchers to “repurpose” metformin for increasing lists of disease indications in mice and humans with advancing age [100,101]. The TAME trial has been listed in ClinicalTrials.gov as of November 2019 and the study protocol is now in the public domain.

Another large clinical trial sponsored by the Veterans Administration (NCT02915198; VA-IMPACT) started on February 19, 2019. This trial plans to study 7,868 subjects with prediabetes and established atherosclerotic disease for 4.5 years in a double-blind placebo control trial with metformin extended release versus placebo for a combined primary endpoints. The primary endpoints include the time to death from any cause, MI, stroke, hospitalization for unstable angina, or symptom-driven coronary revascularization, while time-to-events for oncology-related diseases and diabetes are secondary endpoints.

A recent double blind, randomized Glucose Lowering in Non-Diabetic Hyperglycaemia Trial (GLINT) testing the feasibility of metformin reducing the risk of cancer in elderly obese patients with a high risk of CVD and non-diabetic hyperglycemia concluded that 20,000 subjects are required to obtain significant results for only CVD [102]. Metformin in Longevity Study (MILES) was a small, double-blind, placebo-controlled crossover trial in which they investigated differentially expressed genes in muscular and adipose tissue biopsies after 6-week administration of placebo or 1.5 g per day of metformin [103]. The study reported biomarkers (such as interleukin-6, C-reactive protein, tumour necrosis factor α, insulin-like growth factor 1, cystatin C, N-terminal B-type natriuretic peptides and haemoglobin A1c) selected by the TAME working group except GDF15 [95]. Early Prevention of Diabetes Complications in Europe (ePREDICE) is another large multicenter RCT and currently recruiting participants mostly in Europe, evaluating the impact of metformin (compared with a dipeptidyl peptidase-4 inhibitor) on microvascular complications and cognitive function in individuals with non-diabetic intermediate hyperglycaemia [104].

Taken together, metformin has been identified as a potential agent for primary prevention of cardiovascular diseases and reduction in Major Adverse Cardiovascular Events (MACE) and mortality in low cardiovascular risk and healthy individual (Table 1).

Author and Year

Design

Target population and size

Outcomes measured

Follow up time

Summary of findings

Orio et al., [36]

Prospective, baseline-controlled trial

N=30

Young women with PCOS without additional metabolic or CV disease

Complete hormone profile, serum insulin, glucose, lipid and endothelin-1. Brachial artery diameter and cIMT

6 months

6-month course of metformin improves endothelial structure and function in young, normal-weight women with PCOS.

Khan et al., [37]

Retrospective cohort study

N=1204

Patients who underwent revascularization for chronic limb ischemia

Primary patency, secondary patency, limb salvage, major adverse limb events, major adverse cardiac events and survival rates

Mean of 48 months

Metformin is associated with improved survival and decreased incidence of adverse cardiac events in PAD patients. It did not have an impact on patency or limb salvage rates.

Preiss et al., [39]

Double-blind, placebo-controlled trial

N=173

Patients taking statins, without T2DM with coronary heart disease and large WC

Mean distal cIMT, carotid plaque score, glucose, lipids, C-reactive protein and tissue plasminogen activator

18 months

Metformin had no effect on cIMT and little or no effect on several surrogate markers of cardiovascular disease in non-diabetic patients with high cardiovascular risk, taking statins.

Mohan et al., [38]

Double-blind, placebo-controlled trial

N=63

Patients without diabetes who have CAD with insulin resistance and/or prediabetes

LV mass indexed to height, LV ejection fraction, mass, and volume; abdominal obesity, glycaemic parameters, endothelial function, and blood biomarkers

12 months

Metformin treatment significantly reduced LV mass indexed to height and LV mass compared with placebo in patients with CAD without T2DM. It also improved SBP, reduced oxidative stress and reduced measures of obesity.

Lexis et al., [32]

Double-blind, placebo-controlled trial

N=379

Patients who underwent primary percutaneous coronary intervention for STEMI

LV ejection fraction, NT-proBNP, major adverse cardiac events

4 months

Metformin compared with placebo did not result in improved LV ejection fraction or NT-proBNP levels. As LV function is an important predictor of morbidity and mortality after STEMI, it is unlikely that metformin will have a significant effect on long-term outcomes.

Hartman et al., (2 year follow up of Lexis et al.,) [33]

Double-blind, placebo-controlled trial

N=379

Patients who underwent primary percutaneous coronary intervention for STEMI

Major adverse cardiac events, NT-proBNP, death, reinfarction, recurrent coronary intervention, stroke, heart failure, ICD implantation, and new-onset diabetes mellitus

24 months

Four months metformin treatment initiated at the time of hospitalization in STEMI patients without diabetes did not exert beneficial long-term effects

Knowler et al., [9]

Randomized clinical trial

N=3234

Non-diabetic persons with elevated fasting and post-load plasma glucose concentrations

Diabetes (glucose), adverse effects

Mean of 34 months

Lifestyle changes and treatment with metformin both reduced the incidence of diabetes in persons at high risk. The lifestyle intervention was more effective than metformin.

Kulkarni et al., [103]

Double-blind, placebo-controlled, Crossover trial

N=14

Older patients with impaired glucose tolerance but no diabetes.

Skeletal muscle and subcutaneous adipose tissue for number of expressed genes. Serum glucose, insulin,

3 months

6 weeks of metformin can improve age-associated metabolic derangements in glucose intolerant older adults. Metformin has metabolic and nonmetabolic effects linked to aging

Luchsinger et al., [81]

Double-blind placebo-controlled randomized pilot trial

N=80

Patients with aMCI, overweight or obese, without treated diabetes

Bushcke SRT, ADAS-cog, plasma Aβ42, relative glucose uptake in the posterior cingulate-precuneus measured by brain FDG PET and MRI

12 months

Preliminary evidence of efficacy to improve SRT but not ADASCog. A larger trial seems warranted to evaluate the efficacy and cognitive safety of metformin in prodromal Alzheimer's disease

Table 1: Summary of clinical trials with metformin in non-diabetic population.

PCOS, Polycystic Ovary Syndrome; CV, Cardiovascular; cIMT, Carotid Intima-Media Thickness; PAD, Peripheral Arterial Disease; T2DM, Type 2 Diabetes Mellitus; WC, Waist Circumference; CAD, Coronary Arterial Disease; LV, Left Ventricular; SBP, Systolic Blood Pressure; STEMI, ST-Elevation Myocardial Infarction; NT-proBNP, N-Terminal Pro-Brain Natriuretic Peptide; ICD, Implantable Cardioverter-Defibrillator; aMCI, Amnestic Mild Cognitive Impairment; SRT, Selective Reminding Test; ADAS-cog, Alzheimer's Disease Assessment Scale Cognitive Subscale; Aβ42, Amyloid Beta 42; FDG, F-labelled 2-Deoxy-2-fluoro-D-Glucose; PET, Positron Emission Tomography; MRI, Magnetic Resonance Imaging.

CONCLUSION AND CLINICAL PERSPECTIVES

The rationale for the ongoing or planned metformin trials is almost exclusively based on observations of potential benefits in a population with diabetes (or prediabetes). Beyond its impact on diabetic control, metformin has diverse ranges of effects targeting multiple age-related mechanisms. Cellular and animal studies have found that metformin reduces insulin resistance and decreases inflammatory markers, NF-κB, ROS and mTOR pathways, thus decreasing DNA damage. Human observational studies have shown that metformin decreases the risk of CVD, cancer, depression and other age-related conditions. A few studies found that metformin may reduce MCI and dementia. Ongoing randomized clinical trials will evaluate whether metformin can decrease death from any cause, CVD, stroke, prevent or delay the development of age-dependent diseases, and improve physical and cognitive function. Given its known safety and long-term use in humans, metformin could become a pharmacological intervention against multi-morbidity, frailty and ageing in individuals with or without diabetes, especially in people with low cardiovascular risks or healthy elderly in the primary prevention trial. In terms of primary prevention trial, the Aspirin in Reducing Events in the Elderly (ASPREE) was a primary prevention trial that was established to investigate whether the daily use of 100 mg of enteric-coated aspirin would prolong the healthy life span of older adults conducted in Australia and the United States, recruiting 19,114 relatively healthy older persons from community settings. The primary end point was disability-free survival, which was defined as survival free from dementia or persistent physical disability. The primary composite end point was derived from the first end-point events of death, dementia and persistent physical disability. The use of low-dose aspirin did not differ significantly from placebo in influencing the rate of the primary end point after a median of 4.7 years of follow-up [105]. Taking together, metformin will have great potential to play an important healthy ageing role in combating cardiovascular risks and prolonging disability-free survival. Hence, a properly designed clinical trial similar to the ASPREE trial involving metformin use in the healthy population will most likely answer the challenges of healthy ageing in our modern society.

FUNDING

No external funding.

ACKNOWLEDGEMENTS

School of Public Health provided Ms Jacinta Chapman-Williams vocational scholarship to help collect the references of the literature review for the manuscript. Dr Natalie Ward provided some helpful comments in constructing the initial manuscript.

CONFLICTS OF INTEREST

The authors declare no conflict of interest.

REFERENCES

  1. Australian Institute of Health and Welfare (2018) Older Australia at a glance. Australian Institute of Health and Welfare, Canberra, Australia.
  2. Australian Institute of Health and Welfare (2019) Australian Burden of Disease Study: impact and causes of illness and death in Australia 2015. Australia Institute of Health and Welfare, Canberra, Australia.
  3. Australian Institute of Health and Welfare (2019) Cardiovascular disease. Australian Institute of Health and Welfare, Canberra, Australia.
  4. Australian Institute of Health and Welfare (2015) Cardiovascular disease, diabetes and chronic kidney disease-Australian facts: risk factors. Australian Institute of Health and Welfare, Canberra, Australia.
  5. World Health Organization (2011) Global Atlas on Cardiovascular Disease Prevention and Control. WHO, Geneva, Switzerland.
  6. Go AS, Mozaffarian D, Roger VL, Benjamin EJ, Berry JD, et al. (2014) Heart Disease and Stroke Statistics—2014 Update: A Report From the American Heart Association. Circulation 129: 28-292.
  7. Australian Institute of Health and Welfare (2011) Cardiovascular disease: Australian facts 2011. Australian Institute of Health and Welfare, Canberra, Australia.
  8. Kirkman MS, Briscoe VJ, Clark N, Florez H, Haas LB, et al. (2012) Diabetes in older adults. Diabetes Care 35: 2650-2664.
  9. Knowler WC, Barrett-Connor E, Fowler SE, Hamman RF, Lachin JM, et al. (2002) Reduction in the Incidence of Type 2 Diabetes With Lifestyle Intervention or Metformin. N Engl J Med 346: 393-403.
  10. FDA (2017) FDA Drug Safety Communication: FDA revises warnings regarding use of the diabetes medicine metformin in certain patients with reduced kidney function. FDA, USA.
  11. European Medicines Agency (2017) Metformin and metformin-containing medicines. European Medicines Agency, Amsterdam, Netherlands.
  12. Misbin RI (2004) The Phantom of Lactic Acidosis Due to Metformin in Patients With Diabetes. Diabetes Care 27: 1791-1793.
  13. DeFronzo R, Fleming GA, Chen K, Bicsak TA (2016) Metformin-associated Lactic Acidosis: Current Perspectives on Causes and Risk. Metabolism 65: 20-29.
  14. Valencia W, Florez H (2014) Pharmacological Treatment of Diabetes in Older People. Diabetes Obes Metab 16: 1192-1203.
  15. Glossmann HH, Lutz OMD (2019) Metformin and Aging: A Review. Gerontology 65: 581-590.
  16. Johnson JA, Simpson SH, Toth EL, Majumdar SR (2005) Reduced Cardiovascular Morbidity and Mortality Associated With Metformin Use in Subjects With Type 2 Diabetes. Diabet Med 22: 497-502.
  17. Inzucchi SE, Bergenstal RM, Buse JB, Diamant M, Ferrannini E, et al. (2015) Management of Hyperglycemia in Type 2 Diabetes, 2015: A Patient-Centered Approach: Update to a Position Statement of the American Diabetes Association and the European Association for the Study of Diabetes. Diabetes Care 38: 140-149.
  18. Barzilai N, Crandall JP, Kritchevsky SB, Espeland MA (2016) Metformin as a Tool to Target Aging. Cell Metab 23: 1060-1065.
  19. UK Prospective Diabetes Study (UKPDS) Group (1998) Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34). Lancet 352: 854-865.
  20. Inzucchi SE, Bergenstal RM, Buse JB, Diamant M, Ferrannini E, et al. (2012) Management of Hyperglycemia in Type 2 Diabetes: A Patient-Centered Approach. Position Statement of the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care 55: 1577-1596.
  21. Mamputu JC, Wiernsperger NF, Renier G (2003) Antiatherogenic Properties of Metformin: The Experimental Evidence. Diabetes Metab 29: 6S71-6S76.
  22. Libby P (2003) Metformin and Vascular Protection: A Cardiologist's View. Diabetes Metab 29: 6S117-6S120.
  23. Batchuluun B, Inoguchi T, Sonoda N, Sasaki S, Inoue T, et al. (2014) Metformin and Liraglutide Ameliorate High Glucose-Induced Oxidative Stress via Inhibition of PKC-NAD(P)H Oxidase Pathway in Human Aortic Endothelial Cells. Atherosclerosis 232: 156-164.
  24. Batchuluun B, Sonoda N, Takayanagi R, Inoguchi T (2014) The Cardiovascular Effects of Metformin: Conventional and New Insights. J Endocrinol Diabetes Obes 2: 1035.
  25. Nesti L, Natali A (2017) Metformin Effects on the Heart and the Cardiovascular System: A Review of Experimental and Clinical Data. Nutr Metab Cardiovasc Dis 27: 657-669.
  26. Vazirpanah N, Ottria A, van der Linden M, Wichers CGK, Schuiveling M, et al. (2019) mTOR Inhibition by Metformin Impacts Monosodium Urate Crystal-Induced Inflammation and Cell Death in Gout: A Prelude to a New Add-On Therapy? Ann Rheum Dis 78: 663-671.
  27. Sciarretta S, Volpe M, Sadoshima J (2014) Mammalian Target of Rapamycin Signaling in Cardiac Physiology and Disease. Circ Res 114: 549-564.
  28. Han Y, Xie H, Liu Y, Gao P, Yang X, et al. (2019) Effect of Metformin on All-Cause and Cardiovascular Mortality in Patients With Coronary Artery Diseases: A Systematic Review and an Updated Meta-Analysis. Cardiovasc Diabetol 18: 96.
  29. Diabetes Prevention Program Research G (2019) Long-term Effects of Metformin on Diabetes Prevention: Identification of Subgroups That Benefited Most in the Diabetes Prevention Program and Diabetes Prevention Program Outcomes Study. Diabetes Care 42: 601-608.
  30. Wang CP, Lorenzo C, Habib SL, Jo B, Espinoza SE (2017) Differential Effects of Metformin on Age Related Comorbidities in Older Men With Type 2 Diabetes. J Diabetes Complications 31: 679-686.
  31. Petrie JR, Chaturvedi N, Ford I, Brouwers MCGJ, Greenlaw N, et al. (2017) Cardiovascular and Metabolic Effects of Metformin in Patients With Type 1 Diabetes (REMOVAL): A Double-Blind, Randomised, Placebo-Controlled Trial. Lancet Diabetes Endocrinol 5: 597-609.
  32. Lexis CPH, van Der Horst ICC, Lipsic E, Wieringa WG, de Boer RA, et al. (2014) Effect of Metformin on Left Ventricular Function After Acute Myocardial Infarction in Patients Without Diabetes: The GIPS-III Randomized Clinical Trial. JAMA 311: 1526-1535.
  33. Hartman MHT, Prins JKB, Schurer RAJ, Lipsic E, Lexis CPH, et al. (2017) Two-year Follow-Up of 4 Months Metformin Treatment vs. Placebo in ST-elevation Myocardial Infarction: Data From the GIPS-III RCT. Clin Res Cardiol 106: 939-946.
  34. Maruthur NM, Tseng E, Hutfless S, Wilson LM, Suarez-Cuervo C, et al. (2016) Diabetes Medications as Monotherapy or Metformin-Based Combination Therapy for Type 2 Diabetes: A Systematic Review and Meta-analysis. Ann Intern Med 164: 740-751.
  35. Jadhav S, Ferrell W, Greer IA, Petrie JR, Cobbe SM, et al. (2006) Effects of Metformin on Microvascular Function and Exercise Tolerance in Women With Angina and Normal Coronary Arteries: A Randomized, Double-Blind, Placebo-Controlled Study. J Am Coll Cardiol 48: 956-963.
  36. Orio F Jr, Palomba S, Cascella T, De Simone B, Manguso F, et al. (2005) Improvement in Endothelial Structure and Function After Metformin Treatment in Young Normal-Weight Women With Polycystic Ovary Syndrome: Results of a 6-month Study. J Clin Endocrinol Metab 90: 6072-6076.
  37. Khan SZ, Rivero M, Nader ND, Cherr GS, Harris LM, et al. (2019) Metformin Is Associated With Improved Survival and Decreased Cardiac Events With No Impact on Patency and Limb Salvage After Revascularization for Peripheral Arterial Disease. Ann Vasc Surg 55: 63-77.
  38. Mohan M, Al-Talabany S, McKinnie A, Mordi IR, Singh JSS, et al. (2019) A Randomized Controlled Trial of Metformin on Left Ventricular Hypertrophy in Patients With Coronary Artery Disease Without Diabetes: The MET-REMODEL Trial. Eur Heart J 40: 3409-3417.
  39. Preiss D, Lloyd SM, Ford I, McMurray JJ, Holman RR, et al. (2014) Metformin for Non-Diabetic Patients With Coronary Heart Disease (The CAMERA Study): A Randomised Controlled Trial. Lancet Diabetes Endocrinol 2: 116-124.
  40. Huang RC, Burke V, Newnham JP, Stanley FJ, Kendall GE, et al. (2007) Perinatal and Childhood Origins of Cardiovascular Disease. Int J Obes (Lond) 31: 236-244.
  41. Lai CC, Sun D, Cen R, Wang J, Li S, et al. (2014) Impact of Long-term Burden of Excessive Adiposity and Elevated Blood Pressure from Childhood on Adult Left Ventricular Remodeling Patterns: The Bogalusa Heart Study. J Am Coll Cardiol 64: 1580-1587.
  42. Litwin SE (2014) Childhood Obesity and Adulthood Cardiovascular Disease: Quantifying the Lifetime Cumulative Burden of Cardiovascular Risk Factors. J Am Coll Cardiol 64: 1588-1590.
  43. Whittington HJ, Hall AR, McLaughlin CP, Hausenloy DJ, Yellon DM, et al. (2013) Chronic Metformin Associated Cardioprotection Against Infarction: Not Just a Glucose Lowering Phenomenon. Cardiovasc Drugs Ther 27: 5-16.
  44. Losordo DW, Henry TD (2016) New Definition of Aging? Measuring Regenerative Capacity in Patients. Circ Res 119: 774-775.
  45. Barzilai N, Huffman DM, Muzumdar RH, Bartke A (2012) The Critical Role of Metabolic Pathways in Aging. Diabetes 61: 1315-1322.
  46. Ford ES, Giles WH, Dietz WH (2002) Prevalence of the Metabolic Syndrome Among US Adults: Findings From the Third National Health and Nutrition Examination Survey. JAMA 287: 356-359.
  47. Bonora E, Kiechl S, Willeit J, Oberhollenzer F, Egger G, et al. (2007) Insulin Resistance as Estimated by Homeostasis Model Assessment Predicts Incident Symptomatic Cardiovascular Disease in Caucasian Subjects From the General Population: The Bruneck Study. Diabetes Care 30: 318-324.
  48. Djiogue S, Kamdje AHN, Vecchio L, Kipanyula MJ, Farahna M, et al. (2013) Insulin Resistance and Cancer: The Role of Insulin and IGFs. Endocr Relat Cancer 20: 1-17.
  49. Stuart MJ, Baune BT (2012) Depression and type 2 diabetes: inflammatory mechanisms of a psychoneuroendocrine co-morbidity. Neurosci Biobehav Rev 36: 658-676.
  50. De Felice FG, Ferreira ST (2014) Inflammation, defective insulin signaling, and mitochondrial dysfunction as common molecular denominators connecting type 2 diabetes to Alzheimer disease. Diabetes 63: 2262-2272.
  51. Espinoza SE, Jung I, Hazuda H (2012) Frailty transitions in the San Antonio Longitudinal Study of Aging. J Am Geriatr Soc 60: 652-660.
  52. Goldberg EL, Dixit VD (2015) Drivers of age-related inflammation and strategies for healthspan extension. Immunol Rev 265: 63-74.
  53. Harris RA, Tindale L, Cumming RC (2014) Age-dependent metabolic dysregulation in cancer and Alzheimer's disease. Biogerontology 15: 559-577.
  54. Franceschi C, Garagnani P, Morsiani C, Conte M, Santoro A, et al. (2018) The Continuum of Aging and Age-Related Diseases: Common Mechanisms but Different Rates. Front Med (Lausanne) 5: 61.
  55. Gems D (2015) The aging-disease false dichotomy: understanding senescence as pathology. Front Genet 6: 212.
  56. Rattan SI (2014) Aging is not a disease: implications for intervention. Aging Dis 5: 196-202.
  57. Gladyshev TV, Gladyshev VN (2016) A Disease or Not a Disease? Aging As a Pathology. Trends Mol Med 22: 995-996.
  58. Baar MP, Van Willigenburg H, de Keizer PLJ (2017) Maintenance and repair of an aging life cycle. Oncotarget 8: 86985-86986.
  59. Longo VD, Antebi A, Bartke A, Barzilai N, Brown-Borg HM, et al. (2015) Interventions to Slow Aging in Humans: Are We Ready? Aging Cell 14: 497-510.
  60. Vijg J, Dong X, Milholland B, Zhang L (2017) Genome instability: a conserved mechanism of ageing? Essays Biochem 61: 305-315.
  61. Fedarko NS (2011) The biology of aging and frailty. Clin Geriatr Med 27: 27-37.
  62. Hoeijmakers JH (2009) DNA damage, aging, and cancer. N Engl J Med 361(15): 1475-1485.
  63. Tamariz L, Hare JM (2015) Xanthine oxidase inhibitors in heart failure: where do we go from here? Circulation 131: 1741-1744.
  64. Beyret E, Liao HK, Yamamoto M, Hernandez-Benitez R, Fu Y, et al. (2019) Single-dose CRISPR-Cas9 therapy extends lifespan of mice with Hutchinson-Gilford progeria syndrome. Nat Med 25: 419-422.
  65. Milholland B, Suh Y, Vijg J (2017) Mutation and catastrophe in the aging genome. Exp Gerontol 94: 34-40.
  66. Madiraju AK, Erion DM, Rahimi Y, Zhang XM, Braddock DT, et al. (2014) Metformin suppresses gluconeogenesis by inhibiting mitochondrial glycerophosphate dehydrogenase. Nature 510: 542-546.
  67. Haffner S, Temprosa M, Crandall J, Fowler S, Goldberg R, et al. (2005) Intensive lifestyle intervention or metformin on inflammation and coagulation in participants with impaired glucose tolerance. Diabetes 54: 1566-1572.
  68. Vitale C, Mercuro G, Cornoldi A, Fini M, Volterrani M, et al. (2005) Metformin improves endothelial function in patients with metabolic syndrome. J Intern Med 258: 250-256.
  69. de Aguiar LG, Bahia LR, Villela N, Laflor C, Sicuro F, et al. (2006) Metformin improves endothelial vascular reactivity in first-degree relatives of type 2 diabetic patients with metabolic syndrome and normal glucose tolerance. Diabetes Care 29: 1083-1089.
  70. Wu T, Horowitz M, Rayner CK (2017) New insights into the anti-diabetic actions of metformin: From the liver to the gut. Expert Rev Gastroenterol Hepatol 11: 157-166.
  71. Bahne E, Sun EWL, Young RL, Hansen M, Sonne DP, et al. (2018) Metformin-induced glucagon-like peptide-1 secretion contributes to the actions of metformin in type 2 diabetes. JCI Insight 3.
  72. Pakos-Zebrucka K, Koryga I, Mnich K, Ljujic M, Samali A, et al. (2016) The integrated stress response. EMBO Rep 17: 1374-1395.
  73. Bao X, Borne Y, Muhammad IF, Nilsson J, Lind L, et al. (2019) Growth differentiation factor 15 is positively associated with incidence of diabetes mellitus: The malmo diet and cancer-cardiovascular cohort. Diabetologia 62: 78-86.
  74. Bridges HR, Sirviö VA, Agip AN, Hirst J (2016) Molecular features of biguanides required for targeting of mitochondrial respiratory complex I and activation of AMP-kinase. BMC Biol 14: 65.
  75. Guo M, Mi J, Jiang QM, Xu JM, Tang YY, et al. (2014) Metformin may produce antidepressant effects through improvement of cognitive function among depressed patients with diabetes mellitus. Clinical and Experimental Pharmacology and Physiology 41: 650-656.
  76. Chen F, Wei G, Wang Y, Liu T, Huang T, et al. (2019) Risk factors for depression in elderly diabetic patients and the effect of metformin on the condition. BMC Public Health 19.
  77. Zemdegs J, Martin H, Pintana H, Bullich S, Manta S, et al. (2019) Metformin promotes anxiolytic and antidepressant-like responses in insulin-resistant mice by decreasing circulating branched-chain amino acids. J Neurosci 39: 5935-5948.
  78. Ng TP, Feng L, Yap KB, Lee TS, Tan CH, et al. (2014) Long-term metformin usage and cognitive function among older adults with diabetes. J Alzheimers Dis 41: 61-68.
  79. Hsu C-C, Wahlqvist ML, Lee M-S, Tsai H-N (2011) Incidence of dementia is increased in type 2 diabetes and reduced by the use of sulfonylureas and metformin. J Alzheimers Dis 24: 485-493.
  80. Cheng C, Lin C-H, Tsai Y-W, Tsai C-J, Chou P-H, et al. (2014) Type 2 diabetes and antidiabetic medications in relation to dementia diagnosis. J Gerontol A Biol Sci Med Sci 69: 1299-1305.
  81. Luchsinger JA, Perez T, Chang H, Mehta P, Steffener J, et al. (2016) Metformin in amnestic mild cognitive impairment: results of a pilot randomized placebo controlled clinical trial. Journal of Alzheimer's Disease 51: 501-514.
  82. Chin-Hsiao T (2019) Metformin and the risk of dementia in type 2 diabetes patients. Aging Dis 10: 37.
  83. Moore EM, Mander AG, Ames D, Kotowicz MA, Carne RP, et al. (2013) Increased risk of cognitive impairment in patients with diabetes is associated with metformin. Diabetes Care 36: 2981-2987.
  84. Moreira PI (2014) Metformin in the diabetic brain: Friend or foe? Ann Transl Med 2: 54.
  85. Barini E, Antico O, Zhao Y, Asta F, Tucci V, et al. (2016) Metformin promotes tau aggregation and exacerbates abnormal behavior in a mouse model of tauopathy. Molecular neurodegeneration 11.
  86. Thangthaeng N, Rutledge M, Wong JM, Vann PH, Forster MJ, et al. (2017) Metformin impairs spatial memory and visual acuity in old male mice. Aging and disease 8: 17.
  87. Luchsinger JA, Ma Y, Christophi CA, Florez H, Golden SH, et al. (2017) Metformin, lifestyle intervention, and cognition in the diabetes prevention program outcomes study. Diabetes care 40: 958-965.
  88. Porter KM, Ward M, Hughes CF, O’Kane M, Hoey L, et al. (2019) Hyperglycemia and metformin use are associated with B vitamin deficiency and cognitive dysfunction in older adults. J Clin Endocrinol Metab 104: 4837-4847.
  89. Valencia WM, Stoutenberg M, Florez H (2014) Weight loss and physical activity for disease prevention in obese older adults: An important role for lifestyle management. Curr Diab Rep 14: 539.
  90. Armato JP, DeFronzo RA, Abdul-Ghani M, Ruby RJ (2018) Successful treatment of prediabetes in clinical practice using physiological assessment (STOP DIABETES). The Lancet Diabetes & Endocrinology 6: 781-789.
  91. Salpeter S, Buckley N, Kahn J, Salpeter E (2008) Meta-analysis: Metformin treatment in persons at risk for diabetes mellitus. Am J Med 121: 149-157.
  92. Glossmann H, Reider N (2013) A marriage of two “Methusalem” drugs for the treatment of psoriasis?Arguments for a pilot trial with metformin as add-on for methotrexate. Dermato-endocrinology 5: 252-263.
  93. Espinoza SE, Musi N, Wang C-p, Michalek J, Orsak B, et al. (2019) Rationale and Study Design of a Randomized Clinical Trial of Metformin to Prevent Frailty in Older Adults With Prediabetes. J Gerontol A Biol Sci Med Sci 75: 102-109.
  94. Konopka A, Miller B (2019) Taming expectations of metformin as a treatment to extend healthspan. GeroScience 41: 1-8.
  95. Justice JN, Ferrucci L, Newman AB, Aroda VR, Bahnson JL, et al. (2018) A framework for selection of blood-based biomarkers for geroscience-guided clinical trials: report from the TAME Biomarkers Workgroup. Geroscience 40: 419-436.
  96. Leidal AM, Levine B, Debnath J (2018) Autophagy and the cell biology of age-related disease. Nature cell biology 20: 1338-1348.
  97. Ren J, Zhang Y (2018) Targeting autophagy in aging and aging-related cardiovascular diseases. Trends Pharmacol Sci 39: 1064-1076.
  98. Kanamori H, Naruse G, Yoshida A, Minatoguchi S, Watanabe T, et al. (2019) Metformin enhances autophagy and provides cardioprotection in δ-sarcoglycan deficiency-induced dilated cardiomyopathy. Circ Heart Fail 12: 005418.
  99. Diaz-Morales N, Iannantuoni F, Escribano-Lopez I, Banuls C, Rovira-Llopis S, et al. (2018) Does metformin modulate endoplasmic reticulum stress and autophagy in type 2 diabetic peripheral blood mononuclear cells? Antioxid Redox Signal 28: 1562-1569.
  100. Espeland MA, Crimmins EM, Grossardt BR, Crandall JP, Gelfond JA, et al. (2017) Clinical trials targeting aging and age-related multimorbidity. The Journals of Gerontology: Series A 72: 355-361.
  101. Barzilai N, Cuervo A, Austad S (2018) Aging as a biological target for prevention and therapy. JAMA 320: 1321-1322.
  102. Griffin SJ, Bethel MA, Holman RR, Khunti K, Wareham N, et al. (2018) Metformin in non-diabetic hyperglycaemia: The GLINT feasibility RCT. Health Technol Assess 22: 1-64.
  103. Kulkarni AS, Brutsaert EF, Anghel V, Zhang K, Bloomgarden N, et al. (2018) Metformin regulates metabolic and nonmetabolic pathways in skeletal muscle and subcutaneous adipose tissues of older adults. Aging Cell 17: 12723.
  104. Consortium Te (2016) Early Prevention of Diabetes Complications in Europe 2016.
  105. McNeil JJ, Wolfe R, Woods RL, Tonkin AM, Donnan GA, et al. (2018) Effect of aspirin on cardiovascular events and bleeding in the healthy elderly. N Engl J Med 379: 1509-1518.

Citation: Xu D, Yap T, Affandi JS, Reid C (2020) Metformin Use beyond Diabetes: Reducing Cardiovascular Events in the Healthy Elderly. J Gerontol Geriatr Med 6: 058.

Copyright: © 2020  Dan Xu, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.


Herald Scholarly Open Access is a leading, internationally publishing house in the fields of Sciences. Our mission is to provide an access to knowledge globally.



© 2024, Copyrights Herald Scholarly Open Access. All Rights Reserved!