Skip to main content
  • Study protocol
  • Open access
  • Published:

MetfOrmin BenefIts Lower Extremities with Intermittent Claudication (MOBILE IC): randomized clinical trial protocol

Abstract

Background

Peripheral artery disease (PAD) affects over 230 million people worldwide and is due to systemic atherosclerosis with etiology linked to chronic inflammation, hypertension, and smoking status. PAD is associated with walking impairment and mobility loss as well as a high prevalence of coronary and cerebrovascular disease. Intermittent claudication (IC) is the classic presenting symptom for PAD, although many patients are asymptomatic or have atypical presentations. Few effective medical therapies are available, while surgical and exercise therapies lack durability. Metformin, the most frequently prescribed oral medication for Type 2 diabetes, has salient anti-inflammatory and promitochondrial properties. We hypothesize that metformin will improve function, retard the progression of PAD, and improve systemic inflammation and mitochondrial function in non-diabetic patients with IC.

Methods

200 non-diabetic Veterans with IC will be randomized 1:1 to 180-day treatment with metformin extended release (1000 mg/day) or placebo to evaluate the effect of metformin on functional status, PAD progression, cardiovascular disease events, and systemic inflammation. The primary outcome is 180-day maximum walking distance on the 6-min walk test (6MWT). Secondary outcomes include additional assessments of functional status (cardiopulmonary exercise testing, grip strength, Walking Impairment Questionnaires), health related quality of life (SF-36, VascuQoL), macro- and micro-vascular assessment of lower extremity blood flow (ankle brachial indices, pulse volume recording, EndoPAT), cardiovascular events (amputations, interventions, major adverse cardiac events, all-cause mortality), and measures of systemic inflammation. All outcomes will be assessed at baseline, 90 and 180 days of study drug exposure, and 180 days following cessation of study drug. We will evaluate the primary outcome with linear mixed-effects model analysis with covariate adjustment for baseline 6MWT, age, baseline ankle brachial indices, and smoking status following an intention to treat protocol.

Discussion

MOBILE IC is uniquely suited to evaluate the use of metformin to improve both systematic inflammatory responses, cellular energetics, and functional outcomes in patients with PAD and IC.

Trial Registration: The prospective MOBILE IC trial was publicly registered (NCT05132439) November 24, 2021.

Peer Review reports

Background

Peripheral artery disease (PAD) results from systemic atherosclerosis and is associated with chronic systemic inflammation [1] and impaired mitochondrial function [2]. It affects over 230 million patients world-wide [3]. PAD is prevalent among United States Veterans with higher rates of revascularization within the Veterans Health Administration when compared to private sector hospitalizations overall and especially at an earlier age [4,5,6]. While individuals with PAD may remain asymptomatic, the classic presentation for PAD is intermittent claudication (IC) [7], defined as reproducible lower extremity muscular pain with ambulation due to arterial blood flow limitations. PAD and IC are associated with an increased risk of cardiovascular disease (CVD) morbidity and mortality in addition to a progressive decline in walking distance, functional independence, and quality of life [8,9,10].

Treatment of IC has two goals. First, reduce CVD morbidity and mortality. Second, improve walking ability and quality of life [11, 12]. The former is achieved with optimal medical therapy (OMT) including smoking cessation, blood pressure control, as well as lipid-lowering and antiplatelet therapy. The latter is managed with cilostazol, exercise programs, and surgical revascularization [11]. Unfortunately, pharmacologic treatment of IC with cilostazol is minimally effective and poorly tolerated [12]. Exercise and revascularization can improve symptoms [13] but the improvements are not consistently sustained, are highly dependent on patient compliance, and do not reduce associated CVD morbidity and mortality [14, 15]. Further, vascular interventions to alleviate IC may lead to accelerated PAD progression to critical limb threatening ischemia [16,17,18].

Metformin is the most frequently prescribed oral therapy for Type 2 diabetes and has an excellent safety profile [19]. The pleiotropic effects of metformin include reducing reactive oxygen species production [20], attenuating systemic inflammation [21], and inhibiting mitochondrial damage [22, 23], all of which can improve age-related organ dysfunction [24]. Mechanistically, metformin activates AMP-activated protein kinase (AMPK) [25], increases endothelial nitric oxide synthase (eNOS) activity [26], and promotes mitochondrial biogenesis, mitophagy, and autophagy [27]. In patients with diabetes, metformin is linked to improved cellular respiration and both decreased the incidence and progression of age-related comorbidity (i.e., cancer, CVD, kidney diseases, etc.), frailty, response to physiologic stress, and mortality [24, 28, 29]. These effects appear to be independent of glucose control [24].

In patients with PAD, preclinical studies have shown that metformin stimulates angiogenesis [30] and reduces inflammatory arterial calcification [31]. Over 30 years ago in Italy, Sirtori et al. hypothesized that metformin would improve symptomatic IC and generated supportive preliminary evidence with two small clinical trials [32, 33]. We will extend this work and hypothesize that metformin will improve the functional status of non-diabetic patients with IC by preventing PAD progression and age-related CVD comorbidities through the reduction of systemic inflammation and improvement in systemic cellular respiration as would be predicted by the known effects of metformin on AMPK, eNOS, angiogenesis and mitochondrial health. We will conduct a triple-blind, Phase III, single institution, randomized controlled trial allocating participants to metformin or placebo, MetfOrmin BenefIts Lower Extremities with Intermittent Claudication (MOBILE IC) Trial (NCT05132439), to address this hypothesis.

Methods/design

Over a 4-year study, non-diabetic Veterans with IC will be randomized 1:1 to 180-day treatment with either metformin or placebo to evaluate the effect of metformin on functional status, PAD progression, CVD events, and systemic inflammation at the Veterans Affairs Pittsburgh Healthcare System (VAPHS). The MOBILE IC trial protocol was reviewed by the Food and Drug Administration (FDA) and the VAPHS Institutional Review Board. The protocol follows Standard Protocol Items: Recommendations For Interventional Trials (SPIRIT) and meets all requirements for exception from investigational new drug application (IND145416), received regulatory approval (1622906), and has been publicly registered (ClinicalTrials.gov: NCT05132439). This clinical trial is funded by the VA Office of Research and Development’s Clinical Science Research and Development Merit Award (I01 CX002150).

Study aims and outcomes

The MOBILE IC trial aims to establish the effectiveness of metformin on improving overall functional status, PAD progression, and systemic inflammation and cellular respiration in Veterans with PAD and IC. The primary objective is to evaluate the effectiveness of metformin as a pharmacologic treatment for PAD and IC with a Phase III randomized controlled trial. Outcomes are validated and reproducible measures will be used in the evaluation of PAD and IC.

The primary outcome of interest is the maximum walking distance (MWD) on the 6-min walk test (6MWT) [34]. This is a validated measure of functional status in PAD and IC, is highly reproducible, and correlates best with real-life walking capacity [34,35,36]. Secondary assessments of functional status include distance to claudication pain and rest on the 6MWT, aerobic and anaerobic capacity during cardiopulmonary exercise testing (CPET), Walking Impairment Questionnaire [37], and grip strength. CPET indices include peak oxygen uptake (VO2), ratio of minute ventilation (VE) to exhaled carbon dioxide (VCO2) or breathing efficiency (VE/VCO2), respiratory exchange ratio (VCO2/VO2), heart rate, and blood pressure [38, 39]. Ventilatory anaerobic threshold (i.e. change from aerobic to anaerobic metabolism, and point of IC onset during CPET are also assessed [38]. CPET measures of symptom-limited (maximal) aerobic and anaerobic capacity in patients with PAD and IC correlate with systemic disease severity and outcomes [38, 39]. Functional outcomes will be supported by the general (SF-36) and disease specific (Vascular Quality of Life Questionnaire-6 [VascuQoL-6]) health related quality of life questionnaires.

The MOBILE IC trial will also assess subclinical and clinical PAD outcomes. Subclinical outcomes will be assessed with ankle-brachial index (ABI), pulse volume recording (PVR), and EndoPAT®. The ABI assesses regional lower extremity blood supply in large conduit arteries as well as the contribution of collateral blood vessels. Because of the frequent insensitivity of ABI due to arterial calcification, we will also assess PVR to capture changes in distal blood flow. Systemic endothelial cell and vasomotor function will be evaluated by EndoPAT which measures peripheral artery tonometry before and during reactive hyperemia induced by temporary brachial artery occlusion [40]. The EndoPAT software (Itamar Medical, Israel) calculates both reactive hyperemia index, a measure of endothelial function, as well as an augmentation index, a measure of arterial stiffness [41]. Clinical PAD outcomes include minor and major amputations, revascularization procedures, major adverse cardiac events (MACE; i.e., composite of CVD mortality, myocardial ischemia, coronary revascularization, arrhythmia, heart failure, non-fatal stroke, and transient ischemic attack), and all-cause mortality.

These outcomes will be supported by key evaluations to further understand the biologic mechanism of action for metformin in PAD through measurements of systemic inflammatory biomarkers and mitochondrial function. Inflammatory biomarkers of interest include IL-1β, IL-10, IL-2, IL-6, MCP-1, HMGB1, and VCAM-1. We will perform exploratory studies on the effect of metformin on neutrophil extracellular traps and plasma exosome/microRNA that have been linked to systemic inflammation and atherogenesis [42, add reference for exosomes]. Mitochondrial function including basal and maximal mitochondrial function, ATP production, and coupling efficiency will be assessed in peripheral blood mononuclear cells to examine the systemic effects of metformin on cellular energetics that parallel changes in skeletal muscle [43, 44]. Measures of systemic oxygen consumption with CPET will complement these analyses.

Study design

The MOBILE IC Trial is a single-center, triple blinded (i.e., Veteran, research staff, investigator), placebo-controlled trial testing the effectiveness of 180 days of two over-encapsulated metformin extended release (ER) tablets (500 mg each, 1000 mg/day) versus two placebo capsules daily for non-diabetic Veterans with PAD and IC. The research activities of the MOBILE IC trial, guided by a qualitative survey of Veteran interest in trial participation and visit frequency, are summarized in Fig. 1. In preparation for this trial, a checklist reviewing OMT among all patients evaluated in VAPHS vascular surgery clinic was implemented in September of 2019.

Fig. 1
figure 1

MOBILE IC trial protocol. MACE Major Adverse Cardiovascular Events

A centralized data monitoring committee (DMC) has been assigned by the VA Clinical Science Research and Development (CSRD) to ensure independent oversight of the safety and integrity of the trial without conflict of interest. The responsibilites of the DMC are documented in a Charter that has been approved by the PI, the DMC Chairperson, and the Director of CSRD. The DMC will review study enrollment, adverse events, unexpected problems, unblinded study data, and recruitment as well as retention every 12 months. Due to the excellent safety profile and long history of clinical Metformin use, no interim analysis will be completed.

Randomization, allocation concealment, and blinding

Study participants will be recruited from both in-person and virtual VAPHS vascular surgery clinics. Individuals meeting all inclusion and no exclusion criteria and agree to participate will be randomly assigned 1:1 to 180 days of metformin or placebo treatment (Table 1). The randomization table has been generated by the study statistician and provided to VAPHS Investigational Drug Service (IDS) for study drug allocation. The IDS will maintain the list of allocations for the duration of the study, but has no role in study recruitment, thereby preserving allocation concealment at the point of enrollment. The research staff enrolling participants have no knowledge of allocations. When an eligible participant is enrolled in the study, the VAPHS IDS is informed who then selects the next allocation from the correct sequence. Randomization will be stratified on baseline MWD on the 6MWT as guided by Fontaine Classification (Stage IIA: IC pain at ≥ 200 m; Stage IIB: < 200 m) [45] and smoking status. Within each stratum, variable block sizes (ranging from 2 to 6) are used to further protect allocation concealment. As a placebo-blinded trial, participants and research staff will not know their assignment, but variable block sizes within each stratum provides an additional layer of protection against research staff guessing future allocations. Stratified randomization is employed to maximize statistical power and allow for non-biased subgroup analysis [46].

Table 1 Inclusion and exclusion criteria

Follow up schedule and procedures

Veterans will return at 90 \(\pm \hspace{0.17em}\)21, 180\(\pm \hspace{0.17em}\)21, and 365\(\pm \hspace{0.17em}\)21 days for in-person evaluation. At the baseline, 180 day and 365 day in-person encounters, Veterans will undergo the same series of sequential testing: EndoPAT, venous blood sampling, 6MWT, ABI and PVR, health related quality of life testing and walking impairment questionnaire, grip strength, followed by CPET. The 90-day visit will be used to monitor medication compliance, dispense the next 90-day supply of medication, and collect blood with sample biobanking. Clinical PAD outcomes will be monitored throughout the 12-months following study drug initiation. The 365-day evaluation is intended to examine the durability of limited exposure to study drug and further understand the causality of treatments through temporal changes.

Study drug

Metformin is inexpensive, safe, and well tolerated in both diabetic and non-diabetic patients [47,48,49]. Metformin ER tablets will be over-encapsulated to match placebo capsules. In our experience and as previously published [47, 48, 50], approximately 25% of non-diabetic individuals may experience self-limited gastrointestinal symptoms (i.e., diarrhea, flatus, nausea, abdominal pain) when exposed to study drug. Veterans unable to tolerate these symptoms at any point during drug exposure will reduce dosage to 1 tablet daily. If symptoms persist and remain intolerable, study drug will be discontinued. If 1 tablet is tolerated for 7 days, the dose will be increased back to 2 capsules daily and maintained if tolerated [47]. Those who tolerate 1 tablet but not 2 will continue 1 tablet for the remainder of the planned exposure period.

The estimated glomerular filtration rate (eGFR) will be monitored throughout study drug exposure and as clinically indicated. Study drug will be stopped if eGFR drops to < 30. If organ dysfunction is independent of study drug, it will be re-initiated upon return of function (i.e., eGFR \(\ge \hspace{0.17em}\)45).

Medication tolerance and dose adjustment will be adjudicated with guidance from the DMC which will function as the Data Safety Monitoring Board. The study team will monitor study drug compliance through phone interview and with return of pill bottles at 90- and 180-day encounters. Unused study drug will be counted and then disposed. To encourage compliance and to respect the Veterans’ time commitment, monetary reimbursement will be provided for all in-person visits.

Statistical analysis plan

Power analysis

In accordance with trial design and reporting guidelines, sample size calculations focused on the primary outcome and efficacy of metformin in the treatment of IC [51,52,53]. The sample size was calculated to provide adequate power to assess clinically meaningful differences in the primary outcome between treatment and control groups. Based on the correlation with an associated decrease in mortality and improvement in health related quality of life, a large meaningful change of MWD is defined as 20–50 m which is the equivalent of a 5–100% change over baseline [54, 55]. Supervised exercise and endovascular intervention trials in PAD found a significant increase in MWD on 6MWT compared to control over 12 to 52 weeks with the effect size \(\pm \hspace{0.17em}\)standard deviation range of 22.2 \(\pm \hspace{0.17em}\)52.1 to 53.5 \(\pm \hspace{0.17em}\)105.0 m [56, 57]. The aforementioned initial Italian studies of non-diabetic patients with IC found a 53% increase in exercise capacity after 180 days of metformin exposure [33].

Assuming a baseline mean MWD of 300 \(\pm \hspace{0.17em}\)100 m in the overall trial population with a mean improvement of 30 \(\pm \hspace{0.17em}\)60 m in the metformin group versus 0 \(\pm \hspace{0.17em}\)60 m in the placebo group, a sample size of 80 patients per group will have 85% power to detect an improvement in MWD at 180 days in response to metformin versus placebo. Stratification of baseline MWD potentially reduces the variation between randomized groups and increases statistical power [46, 58]. The magnitude of power gained cannot be precisely quantified; thus, we did not adjust our sample size estimation for stratification. Therefore, allowing for a conservative 20% dropout rate, consistent with recently completed PAD and VA trials, we will use a total sample size of 200 with 100 per treatment group [14, 59]. Study advertisement and support will be provided to the primary care and cardiology departments within the VAPHS as needed to support enrollment.

Outcome evaluation

The primary analysis will be completed on an intention-to-treat plan with secondary analysis completed on per-protocol study drug allocation. All analysis will be subject to an \(\alpha\) level of 0.05 on two-sided testing, and findings will be reported using CONSORT guidelines [53]. Trial results will be disseminated through ClinicalTrials.gov and peer reviewed publication.

The primary outcome will be analyzed using linear mixed-effects model analysis with MWD at 180 days as the outcome variable, a fixed effect for treatment assignment, and covariate adjustment for patient baseline MWD [60, 61]. This approach addresses the study question systematically: For 2 patients with the same pre-trial MWD, one given metformin and one placebo, what is the estimated difference in MWD after 180 days of study drug exposure? The approach, with a covariate adjustment for the baseline measure, is preferred over change scores due to favorable estimation properties and increased statistical power [61]. The model will adjust for baseline covariates with known strong associations to PAD outcomes (age, smoking status, and ABI [8]) to improve precision and increase statistical power [60, 61]. As randomization will be stratified by baseline MWD (MWD < 200 or  \(\ge\) 200 m) and smoking status, we will test the interaction between these factors and treatment assignment to determine if the treatment effect is different for patients across the baseline MWD stratum.

Secondary outcome analysis will mirror that of the primary outcome for continuous variables. Each of the exploratory secondary outcomes, best summarized as time-to-event (amputations, vascular interventions, MACE, and all-cause mortality), will be reported using Kaplan–Meier analysis, log-rank tests, Cox proportional-hazards, and Fine-Gray models to estimate the difference between treatment groups, controlling for the competing risk of mortality, as appropriate. The MOBILE IC trial is not powered to test for differences in these outcomes but they are included to assess safety, mechanisms of action, allow for future secondary analysis with related studies, as described below, and inform future clinical trials of metformin in PAD and other diseases.

Secondary analysis of outcomes will include per protocol analysis as well as a Bayesian statistical analysis plan.

Missing data

All clinical outcomes will be de-identified, collected and managed using REDCap (Research Electronic Data Capture) tools hosted at the Veterans Affairs Information Resource Center (ViREC) [62, 63]. REDCap is a secure, web-based software platform designed to support data capture for research studies, providing (1) an intuitive interface for validated data capture; (2) audit trails for tracking data manipulation and export procedures; (3) automated export procedures for seamless data downloads to common statistical packages; and (4) procedures for data integration and interoperability with external sources.

We will report all adverse events and reasons for study drop-out using a withdrawal/termination form to assess the missing data mechanism: missing completely at random, missing at random, or non-ignorable missingness. We will conduct sensitivity analyses for primary and secondary outcomes using several validated methods: (1) complete case analyses which assumes missing completely at random; (2) multiple imputation which assumes missing at random; (3) assigning poor scores for missing values differentially by treatment group which aligns with non-ignorable missingness, and (4) the composite approach proposed by Colantuoni et al. [64]. Sensitivity analyses are recommended for trials with missing data, and we will use similar methods to those used successfully in a recent VA trial [59].

Trial harmonization and secondary analysis

The study drug exposure duration, evaluation of the primary outcome, and quality of life testing in MOBILE IC were synchronized with the Improved PAD PERformance with METformin (PERMET) trial (NCT03054519) [34,35,36]. The actively enrolling PERMET trial will evaluate if up to 2000 mg metformin daily for 180 days will improve 6MWT performance among individuals with PAD compared to placebo. Data from both trials will be synchronized (expected n = 412) and will allow for secondary analyses including the (1) dose–response relationship, (2) effects of metformin among pre-specified subgroups, and (3) effects of metformin on CVD events. Pre-specified subgroups include stratification by baseline age, MWD on 6MWT, ABI, and smoking status.

Discussion

PAD is a disease of systemic inflammation and atherosclerosis often presenting as IC. Patients with PAD and IC reduce their physical activity to limit the pain associated with ambulation. The systemic nature of atherosclerosis in PAD increases their overall risk of CVD. Currently, treatments are limited and, in the United States, the FDA has not approved an effective medication for IC in over 2 decades [65]. Existing therapies include preoperative medical optimization that target systemic CVD risk factors. Lower extremity revascularization, especially endovascular techniques, is increasingly prescribed and provides some symptomatic relief. However, it does not address the underlying systemic atherosclerosis or the risk of associated CVD. Lower extremity revascularization may also accelerate the progression to critical limb threatening ischemia [16,17,18, 66]. Metformin has salient properties beyond that of glucose control with mounting evidence for its pro-mitochondrial and anti-inflammatory properties, potential to promote arteriogenesis, reduction in the incidence of diseases of aging including CVDs, and improved longevity [21, 24, 30]. Therefore, the evidence supports investigating the ability of metformin to improve cellular energetics, reduce systemic inflammation, and improve functional status in patients with PAD and IC. The MOBILE IC Trial will provide empiric evidence and mechanistic data for the potential effectiveness of metformin as an innovative therapy in Veterans with PAD. The a priori planned synchronization of the MOBILE IC and PERMET trials will allow for further investigation of the effects of metformin on PAD among pre-specified subgroups and in the analysis of outcomes that each trial alone is under-powered to evaluate.

Availability of data and materials

Data access will be limited to investigators and study personnel. The data and study materials will not be made available to other researchers because they contain subject identifiers.

Abbreviations

PAD:

Peripheral artery disease

IC:

Intermittent claudication

CVD:

Cardiovascular disease

OMT:

Optimal medical therapy

AMPK:

AMP-activated protein kinase

eNOS:

Endothelial nitric oxide synthase

MOBILE IC:

MetfOrmin BenefIts Lower Extremities with Intermittent Claudication

VAPHS:

Veterans Affairs Pittsburgh Healthcare System

FDA:

Food and Drug Administration

SPIRIT:

Standard Protocol Items: Recommendations for Interventional Trials

IND:

Investigational new drug application

MWD:

Maximum walking distance

6MWT:

6-Minute walk test

CPET:

Cardiopulmonary exercise testing

VO2 :

Peak oxygen uptake

VE:

Ratio of minute ventilation

VCO2 :

Carbon dioxide

VE/VCO2 :

Breathing efficiency

VCO2/VO2 :

Respiratory exchange ratio

VascuQoL-6:

Vascular Quality of Life Questionnaire-6

ABI:

Ankle-brachial index

PVR:

Pulse volume recording

ER:

Extended release

IDS:

Investigational Drug Service

eGFR:

Estimated glomerular filtration rate

MACE:

Major adverse cardiac events

REDCap:

Research Electronic Data Capture

ViREC:

Veterans Affairs Information Resource Center

PERMET:

PAD PERformance with METformin

CSRD:

Clinical Science Research and Development

DMC:

Data Monitoring Committee

References

  1. Brevetti G, Giugliano G, Brevetti L, Hiatt WR. Inflammation in periapheral artery disease. Circulation. 2010;122:1862–75.

    Article  Google Scholar 

  2. Park SY, Pekas EJ, Headid RJ, Son WM, Wooden TK, Song J, et al. Acute mitochondrial antioxidant intake improves endothelial function, antioxidant enzyme activity, and exercise tolerance in patients with peripheral artery disease. Am J Physiol Hear Circ Physiol. 2020;319:H456–67.

    Article  CAS  Google Scholar 

  3. Polonsky TS, McDermott MM. Lower extremity peripheral artery disease without chronic limb-threatening ischemia: a review. JAMA J Am Med Assoc. 2021;325:2188–98.

    Article  Google Scholar 

  4. Mayfield JA, Caps MT, Reiber GE, Maynard C, Czerniecki JM, Sangeorzan BJ. Trends in peripheral vascular procedures in the Veterans Health Administration. J Rehabil Res Dev. 2001;38:1989–98.

    Google Scholar 

  5. Brown DW. Smoking prevalence among US veterans. J Gen Intern Med. 2010;25:147–9.

    Article  Google Scholar 

  6. Willey J, Mentias A, Vaughan-Sarrazin M, McCoy K, Rosenthal G, Girotra S. Epidemiology of lower extremity peripheral artery disease in veterans. J Vasc Surg. 2018;68:527-535.e5. https://doi.org/10.1016/j.jvs.2017.11.083.

    Article  Google Scholar 

  7. Sigvant B, Lundin F, Wahlberg E. The risk of disease progression in peripheral arterial disease is higher than expected: a meta-analysis of mortality and disease progression in peripheral arterial disease. Eur J Vasc Endovasc Surg. 2016;51:395–403. https://doi.org/10.1016/j.ejvs.2015.10.022.

    Article  CAS  Google Scholar 

  8. McDermott M, Liu K, Greenland P, Guralnik J, Criqui M, Chan C, et al. Functional decline in peripheral arterial disease: associations with the ankle brachial index and leg symptoms. J Am Med Assoc. 2004;292:453–61. https://doi.org/10.1001/jama.292.4.453.

    Article  CAS  Google Scholar 

  9. McDermott MM, Guralnik JM, Tian L, Liu K, Ferrucci L, Liao Y, et al. Associations of borderline and low normal ankle-brachial index values with functional decline at 5-year follow-up. The WALCS (Walking and Leg Circulation Study). J Am Coll Cardiol. 2009;53:1056–62.

    Article  Google Scholar 

  10. McDermott MMG, Greenland P, Liu K, Guralnik JM, Celic L, Criqui MH, et al. The ankle brachial index is associated with leg function and physical activity: the walking and leg circulation study. Ann Intern Med. 2002;136:873–83.

    Article  Google Scholar 

  11. Barrett C, Barshes NR, Corriere MA, Drachman DE, Fleisher LA, Gerry Fowkes FR, et al. AHA/ACC guideline on the management of patients with lower extremity peripheral artery disease: executive summary. Circulation. 2016;2017:135. https://doi.org/10.1161/CIR.0000000000000470.

    Article  Google Scholar 

  12. Bedenis R, Stewart M, Cleanthis M, Robless P, Mikhailidis DP, Stansby G. Cilostazol for intermittent claudication. Cochrane Database Syst Rev. 2014. https://doi.org/10.1002/14651858.CD003748.pub4.

    Article  Google Scholar 

  13. Hiatt WR, Rogers RK, Brass EP. The treadmill is a better functional test than the 6-minute walk test in therapeutic trials of patients with peripheral artery disease. Circulation. 2014;130:69–78.

    Article  Google Scholar 

  14. McDermott MM, Kibbe MR, Guralnik JM, Ferrucci L, Criqui MH, Domanchuk K, et al. Durability of benefits from supervised treadmill exercise in people with peripheral artery disease. J Am Heart Assoc. 2019. https://doi.org/10.1161/JAHA.118.009380.

    Article  Google Scholar 

  15. Murphy TP, Cutlip DE, Regensteiner JG, Mohler ER, Cohen DJ, Reynolds MR, et al. Supervised exercise, stent revascularization, or medical therapy for claudication due to aortoiliac peripheral artery disease: the CLEVER study. J Am Coll Cardiol. 2015;65:999–1009.

    Article  Google Scholar 

  16. Madabhushi V, Davenport D, Jones S, Khoudoud SA, Orr N, Minion D, et al. Revascularization of intermittent claudicants leads to more chronic limb threatening ischemia and higher amputation rates. J Vasc Surg. 2021. https://doi.org/10.1016/j.jvs.2021.02.045.

    Article  Google Scholar 

  17. Fakhry F, Fokkenrood HJP, Spronk S, Teijink JAW, Rouwet EV, Hunink MGM. Endovascular revascularisation versus conservative management for intermittent claudication. Cochrane Database Syst Rev. 2018;2018.

  18. Levin SR, Farber A, Cheng TW, Arinze N, Jones DW, Rybin D, et al. Patients undergoing interventions for claudication experience low perioperative morbidity but are at risk for worsening functional status and limb loss. J Vasc Surg. 2019;72:241–9.

    Article  Google Scholar 

  19. American Diabetes Association. American Diabetes Association standards of medical care in diabetes. J Clin Appl Res Educ. 2018;41. www.copyright.com. Accessed 16 May 2019.

  20. Cabreiro F, Au C, Leung KY, Vergara-Irigaray N, Cochemé HM, Noori T, et al. Metformin retards aging in C. elegans by altering microbial folate and methionine metabolism. Cell. 2013;153:228–39. https://doi.org/10.1016/j.cell.2013.02.035.

    Article  CAS  Google Scholar 

  21. Cameron A, Morrison V, Levin D, Mohan M, Fortheath C, Beall C, et al. Anti-inflammatory effects of metfromin irrespective of diabetes status. Circ Res. 2016;119:652–65.

    Article  CAS  Google Scholar 

  22. Algire C, Moiseeva O, Deschênes-Simard X, Amrein L, Petruccelli L, Birman E, et al. Metformin reduces endogenous reactive oxygen species and associated DNA damage. Cancer Prev Res. 2012;5:536–43.

    Article  CAS  Google Scholar 

  23. Bhatti JS, Bhatti GK, Reddy PH. Mitochondrial dysfunction and oxidative stress in metabolic disorders—a step towards mitochondria based therapeutic strategies. Biochim Biophys Acta Mol Basis Dis. 2017;1863:1066–77. https://doi.org/10.1016/j.bbadis.2016.11.010.

    Article  CAS  Google Scholar 

  24. Campbell JM, Bellman SM, Stephenson MD, Lisy K. Metformin reduces all-cause mortality and diseases of ageing independent of its effect on diabetes control: a systematic review and meta-analysis. Ageing Res Rev. 2017;40:31–44. https://doi.org/10.1016/j.arr.2017.08.003.

    Article  CAS  Google Scholar 

  25. Musi N, Hirshman MF, Nygren J, Svanfeldt M, Bavenholm P, Rooyackers O, et al. Metformin increases AMP-activated protein kinase activity in skeletal muscle of subjects with type 2 diabetes. Diabetes. 2002;51:2074–81.

    Article  CAS  Google Scholar 

  26. Yu J-WW, Deng Y-PP, Han X, Ren G-FF, Cai J, Jiang G-JJ. Metformin improves the angiogenic functions of endothelial progenitor cells via activating AMPK/eNOS pathway in diabetic mice. Cardiovasc Diabetol. 2016;15:88. https://doi.org/10.1186/s12933-016-0408-3.

    Article  CAS  Google Scholar 

  27. Bharath LP, Agrawal M, McCambridge G, Nicholas DA, Hasturk H, Liu J, et al. Metformin enhances autophagy and normalizes mitochondrial function to alleviate aging-associated inflammation. Cell Metab. 2020;32:44-55.e6. https://doi.org/10.1016/j.cmet.2020.04.015.

    Article  CAS  Google Scholar 

  28. Barzilai N, Crandall JP, Kritchevsky SB, Espeland MA. Metformin as a tool to target aging. Cell Metab. 2016;23:1060–5.

    Article  CAS  Google Scholar 

  29. Reitz KM, Marroquin OC, Zenati MS, Kennedy J, Korytkowski M, Tzeng E, et al. Association between metformin exposure and postoperative outcomes in diabetic adults. JAMA Surg. 2020;15213:1–10.

    Google Scholar 

  30. Takahashi N, Shibata R, Ouchi N, Sugimoto M, Murohara T, Komori K. Metformin stimulates ischemia-induced revascularization through an eNOS dependent pathway in the ischemic hindlimb mice model. J Vasc Surg. 2015;61:489–96.

    Article  Google Scholar 

  31. Forouzandeh F, Salazar G, Patrushev N, Xiong S, Hilenski L, Fei B, et al. Metformin beyond diabetes: pleiotropic benefits of metformin in attenuation of atherosclerosis. J Am Heart Assoc. 2014;3.

  32. Sirtori CR, Franceschini G, Gianfranceschi G, Sirtori M, Montanari G, Bosisio E, et al. Metformin improves peripheral vascular flow in nonhyperlipidemic patients with arterial disease. J Cardiovasc Pharmacol. 1984;6:914–23.

    Article  CAS  Google Scholar 

  33. Montanari G, Bondioli A, Rizzato G, Puttini M, Tremoli E, Mussoni L, et al. Treatment with low dose metformin in patients with peripheral vascular disease. Pharmacol Res. 1992;25:63–73.

    Article  CAS  Google Scholar 

  34. McDermott M, Guralnik J, Criqui M, Liu K, Kibbe M, Ferrucci L. The six-minute walk is a better outcome measure than treadmill walking tests in therapeutic trials of patients with peripheral artery disease. Circulation. 2014;130:61–8.

    Article  Google Scholar 

  35. McDermott M, Ades P, Dyer A, Ferrucci L, Guralnick J, Pearce W, et al. Corridor-based functional performance measures correlate better with physical activity during daily life than treadmill measures in persons with peripheral arterial disease. J Vasc Surg. 2010;46:220–31.

    Google Scholar 

  36. Montgomery PS, Gardner AW. The clinical utility of a six-minute walk test in peripheral arterial occlusive disease patients. J Am Geriatr Soc. 2015;46:706–11. https://doi.org/10.1111/j.1532-5415.1998.tb03804.x.

    Article  Google Scholar 

  37. Jain A, Liu K, Ferrucci L, Criqui MH, Tian L, Guralnik JM, et al. Declining walking impairment questionnaire scores are associated with subsequent increased mortality in peripheral artery disease. J Am Coll Cardiol. 2013;61:1820–9. https://doi.org/10.1016/j.jacc.2013.01.060.

    Article  Google Scholar 

  38. Forman DE, Arena R, Boxer R, Dolansky MA, Eng JJ, Fleg JL, et al. Prioritizing functional capacity as a principal end point for therapies oriented to older adults with cardiovascular disease: a scientific statement for healthcare professionals from the American Heart Association. Circulation. 2017;135:e894-918.

    Article  Google Scholar 

  39. Chehuen MDR, Cucato GG, Saes GF, Costa LAR, Leicht AS, Ritti-Dias RM, et al. Reproducibility of anaerobic and pain thresholds in male patients with intermittent claudication. J Cardiopulm Rehabil Prev. 2016;36:358–67.

    Article  Google Scholar 

  40. Onkelinx S, Cornelissen V, Goetschalckx K, Thomaes T, Verhamme P, Vanhees L. Reproducibility of different methods to measure the endothelial function. Vasc Med (United Kingdom). 2012;17:79–84.

    Article  Google Scholar 

  41. Moerland M, Kales AJ, Schrier L, Van Dongen MGJ, Bradnock D, Burggraaf J. Evaluation of the endoPAT as a tool to assess endothelial function. Int J Vasc Med. 2012;2012.

  42. Döring Y, Soehnlein O, Weber C. Neutrophil extracellular traps in atherosclerosis and atherothrombosis. Circ Res. 2017;120:736–43.

    Article  Google Scholar 

  43. Chacko BK, Kramer PA, Ravi S, Benavides GA, Mitchell T, Dranka BP, et al. The Bioenergetic Health Index: a new concept in mitochondrial translational research. Clin Sci. 2014;127:3 7-373.

    Article  Google Scholar 

  44. Altintas MM, DiBartolo S, Tadros L, Samelko B, Wasse H. Metabolic changes in peripheral blood mononuclear cells isolated from patients with end stage renal disease. Front Endocrinol (Lausanne). 2021;12:629239.

    Article  Google Scholar 

  45. Hardman RL, Jazaeri O, Yi J, Smith M, Gupta R. Overview of classification systems in peripheral artery disease. Semin Intervent Radiol. 2014;31:378–88. https://doi.org/10.1055/s-0034-1393976.

    Article  Google Scholar 

  46. Kernan W, Viscoli C, Makuch R, Brass L, Horwitz R. Stratified randomization for clinical trials. J Clin Epidemiol. 1999;52:19–26. https://doi.org/10.1016/S0895-4356(98)00138-3.

    Article  CAS  Google Scholar 

  47. Bonnet F, Scheen A. Understanding and overcoming metformin gastrointestinal intolerance. Diabetes Obes Metab. 2017;19:473–81.

    Article  CAS  Google Scholar 

  48. Sardu C, Paolisso P, Sacra C, Mauro C, Minicucci F, Portoghese M, et al. Effects of metformin therapy on coronary endothelial dysfunction in prediabetic patients with stable angina and non obstructive coronary artery stenosis: the CODYCE multicenter prospective study. Diabetes Care. 2019. https://doi.org/10.2337/dc18-2356.

    Article  Google Scholar 

  49. Salpeter S, Greyber E, Pasternak G, Salpeter E. Risk of fatal and nonfatal lactic acidosis with metformin use in type 2 diabetes mellitus. Cochrane Database Syst Rev. 2010.

  50. Reitz KM, Seymour CW, Vates J, Quintana M, Viele K, Detry M, et al. Strategies to Promote ResiliencY (SPRY): a randomised embedded multifactorial adaptative platform (REMAP) clinical trial protocol to study interventions to improve recovery after surgery in high-risk patients. BMJ Open. 2020;10:e037690.

    Article  Google Scholar 

  51. Mullins CD, Vandigo J, Zheng Z, Wicks P. Patient-centeredness in the design of clinical trials. Value Health. 2014;17:471–5. https://doi.org/10.1016/j.jval.2014.02.012.

    Article  Google Scholar 

  52. Sox H. The patient-centered outcomes research institute should focus on high-impact problems that can be solved quickly. Health Aff. 2012;31:2176–82.

    Article  Google Scholar 

  53. Schulz KF, Altman DG, Moher D, Barbour V, Berlin JA, Boutron I, et al. CONSORT 2010 statement: updated guidelines for reporting parallel group randomised trials. Consort. 2010;8:604–12. https://doi.org/10.3736/jcim20100702.

    Article  Google Scholar 

  54. Gardner AW, Montgomery PS, Wang M. Minimal clinically important differences in treadmill, 6-minute walk, and patient-based outcomes following supervised and home-based exercise in peripheral artery disease. Vasc Med (United Kingdom). 2018;23:349–57.

    Article  Google Scholar 

  55. McDermott MM, Tian L, Criqui MH, Ferrucci L, Conte MS, Zhao L, et al. Meaningful change in 6-minute walk in people with peripheral artery disease. J Vasc Surg. 2021;73:267-276.e1.

    Article  Google Scholar 

  56. McDermott MM, Ferrucci L, Tian L, Guralnik JM, Lloyd-Jones D, Kibbe MR, et al. Effect of granulocyte-macrophage colony-stimulating factor with or without supervised exercise on walking performance in patients with peripheral artery disease: the PROPEL randomized clinical trial. JAMA. 2017;318:2089–98.

    Article  CAS  Google Scholar 

  57. McDermott MM, Liu K, Guralnik JM, Criqui MH, Spring B, Tian L, et al. Home-based walking exercise intervention in peripheral artery disease: a randomized clinical trial. JAMA. 2013;310:57–65. https://doi.org/10.1001/jama.2013.7231.

    Article  CAS  Google Scholar 

  58. Grizzle J. Covariate adjustment increases statistical power in randomized controlled trials. J Clin Epidemiol. 1982;3:1391.

    Google Scholar 

  59. Nidich S, Mills P, Rainforth M, Heppner P, Schneider R, Rosenthal N, et al. Non-trauma-focused mediation versus exposure therapy in veterans with post-traumatic stress disorder: a randomied controlled trial. Lancet. 2018;5:975–86.

    Google Scholar 

  60. Kent DM, Trikalinos TA, Hill MD. Are unadjusted analyses of clinical trials inappropriately biased toward the null? Stroke. 2009;40:672–3.

    Article  Google Scholar 

  61. Vickers AJ, Altman DG. Statistics notes: analysing controlled trials with baseline and follow up measurements. BMJ. 2001;323:1123–4. https://doi.org/10.1136/bmj.323.7321.1123.

    Article  CAS  Google Scholar 

  62. Harris PA, Taylor R, Thielke R, Payne J, Gonzalez N, Conde JG. Research electronic data capture (REDCap)—a metadata-driven methodology and workflow process for providing translational research informatics support. J Biomed Inform. 2009;42:377–81. https://doi.org/10.1016/j.jbi.2008.08.010.

    Article  Google Scholar 

  63. Harris PA, Taylor R, Minor BL, Elliott V, Fernandez M, O’Neal L, et al. The REDCap consortium: building an international community of software platform partners. J Biomed Inform. 2019;95:103208. https://doi.org/10.1016/j.jbi.2019.103208.

    Article  Google Scholar 

  64. Colantuoni E, Scharfstein DO, Wang C, Hashem MD, Leroux A, Needham DM, et al. Statistical methods to compare functional outcomes in randomized controlled trials with high mortality. BMJ. 2018;360: j5748.

    Article  Google Scholar 

  65. Food and Drug Administration. Center for Drug Evaluation and Research—Pletal. 1999.

  66. Reitz KM, Althouse AD, Meyer J, Arya S, Goodney PP, Shireman PK, et al. Association of smoking with postprocedural complications following open and endovascular interventions for intermittent claudication. JAMA Cardiol. 2021;15213:1–10.

    Google Scholar 

Download references

Acknowledgements

We acknowledge the Veterans Research Foundation’s Clinical Trials Center for regulatory support, the VAPHS IDS pharmacy for patient randomization and for study drug management, and the Surgery and Medicine Service Lines at VAPHS for institutional support for the conduct of the trial. Further, we acknowledge Dr. Mary McDermott, Professor of Medicine and Preventative Medicine at Northwestern University for her support and feedback on the MOBILE IC trial protocol.

Funding

The MOBILE IC Trial is funded by the Veterans Affairs Office of Research and Development’s CSRD Merit Award I01 CX002150 (Tzeng). This research was supported in part by Grant 5T32HL0098036 from the National Heart, Lung, and Blood Institute (Reitz) and L30 AG064730 National Institute on Aging (Reitz). These funding sources had no role in the design and conduct of the study; collection, management, analysis, and interpretation of the data; preparation, review, or approval of the manuscript; and decision to submit for publication. Dr. Tzeng is the Sponsor-investigator who obtained the funding for the trial and designed the study. She is responsible for the oversight of the data collection, management, analysis, interpretation of the data, and writing of the report. She will have the ultimate authority over these activities.

Author information

Authors and Affiliations

Authors

Contributions

The trial protocol was developed by KR (MD MSc), AA (PhD—Statistician), DH (MD, MSc), DF (MD), and ET (MD); statistical analysis plan by KR and AA. The subclinical outcomes including but not limited to the mitochondrial, NET, exosomes/microRNA, and biomarker evaluation were developed and supported by KR, DF, BZ (MD), YV (PhD), RZ (PhD), RR (PhD) and ET. The manuscript was drafted by KR and ET. The trial protocol and this manuscript was critically reviewed by all authors. KR and ET hold responsibility for the entirety of the trial development and manuscript. All authors have reviewed and approved the manuscript.

Corresponding author

Correspondence to Edith Tzeng.

Ethics declarations

Ethics approval and consent to participate

The MOBILE IC Trial was reviewed by the FDA and was identified as exempt from Investigational New Drug review (IND145416). The VAPHS Investigational Review Board approved the procedures specified by the MOBILE IC study protocol which includes written informed consent for each participant completed by the investigators in combination with study personnel (1622906; Supplement). Any future protocol modifications will be reviewed by the institutional review board, discussed with the DMC, and any other relevant parties prior to implementation.

Consent for publication

Not applicable.

Competing interests

No author reports any disclosures, conflict of interest or relevant financial interests related to the content of the manuscript. The opinions expressed here are those of the authors and do not necessarily reflect the position of the Department of Veterans Affairs or the US government.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Reitz, K.M., Althouse, A.D., Forman, D.E. et al. MetfOrmin BenefIts Lower Extremities with Intermittent Claudication (MOBILE IC): randomized clinical trial protocol. BMC Cardiovasc Disord 23, 38 (2023). https://doi.org/10.1186/s12872-023-03047-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12872-023-03047-8

Keywords