Cardiovascular efficacy of liraglutide and semaglutide in individuals with diabetes and peripheral artery disease

Subodh Verma, Mohammed Al-Omran, Lawrence A Leiter, C David Mazer, Søren Rasmussen, Hans A Saevereid, Maria Sejersten Ripa, Marc P Bonaca, Subodh Verma, Mohammed Al-Omran, Lawrence A Leiter, C David Mazer, Søren Rasmussen, Hans A Saevereid, Maria Sejersten Ripa, Marc P Bonaca

Abstract

Aim: To evaluate the cardiovascular (CV) efficacy of liraglutide and semaglutide in patients with type 2 diabetes (T2D) and peripheral artery disease (PAD).

Materials and methods: LEADER and SUSTAIN 6 trials investigated subcutaneous liraglutide (≤1.8 mg/day) and semaglutide (0.5 or 1.0 mg/week), respectively, versus placebo in patients with T2D and high CV risk (median follow-up: 3.8 and 2.1 years, respectively). The primary outcome was a composite of CV death, non-fatal myocardial infarction or non-fatal stroke (major adverse CV event [MACE]) according to the presence of PAD at baseline.

Results: Overall, 1184/9340 (12.7%) patients in LEADER and 460/3297 (14.0%) in SUSTAIN 6 had PAD at baseline. Patients with PAD were at an ~35% increased risk of MACE versus those without (LEADER: hazard ratio [HR] 1.36, 95% confidence interval [CI] 1.17-1.58; SUSTAIN 6: HR 1.33, 95% CI 0.94-1.83). The effects of both therapies on MACE were consistently beneficial in patients with PAD (liraglutide: HR 0.77, 95% CI 0.58-1.01; semaglutide: 0.61, 0.33-1.13) and without (liraglutide: HR 0.89, 95% CI 0.79-1.00; semaglutide: HR 0.77, 95% CI 0.58-1.01; Pinteraction = .34 for liraglutide and .49 for semaglutide). Absolute risk reductions for MACE were higher in patients with PAD (liraglutide: 4.13%-point, 95% CI -0.15-8.42; semaglutide: 4.63%-point, 95% CI -0.58-9.84) versus without (liraglutide:1.42%-point, 95% CI -0.03-2.87; semaglutide: 1.90%-point, 95% CI 0.00-3.80).

Conclusion: Both liraglutide and semaglutide reduce MACE with consistent CV efficacy regardless of PAD status.

Trial registration: ClinicalTrials.gov NCT01179048 NCT01720446.

Keywords: GLP-1; cardiovascular disease; macrovascular disease; peripheral artery disease; receptor agonists; type 2 diabetes.

Conflict of interest statement

SV holds a Tier 1 Canada Research Chair in Cardiovascular Surgery; and reports receiving research grants and/or speaking honoraria from Amarin, Amgen, AstraZeneca, Bayer, Boehringer Ingelheim, Bristol‐Myers Squibb, Eli Lilly, EOCI Pharmacomm Ltd, HLS Therapeutics, Janssen, Merck, Novartis, Novo Nordisk, Pfizer, PhaseBio, Sanofi, Sun Pharma, and the Toronto Knowledge Translation Working Group. He is the President of the Canadian Medical and Surgical Knowledge Translation Research Group, a federally incorporated not‐for‐profit physician organization. MA‐O has received consulting fees from Amgen and Bayer. LAL has received research funding (to institution), provided continuing medical education on behalf of and/or acted as an adviser to AstraZeneca, Boehringer Ingelheim, Eli Lilly, Lexicon, Merck, Novo Nordisk, Pfizer, Sanofi and Servier. CDM has received consulting fees from Amgen, AstraZeneca, BioAge, Boehringer Ingelheim and PhaseBio. SR is an employee and shareholder of Novo Nordisk A/S. HAS is a former employee of Novo Nordisk A/S. MSR is an employee and shareholder of Novo Nordisk A/S. MPB is the Executive Director of CPC, a non‐profit academic research organization affiliated with the University of Colorado that receives research grant/consulting funding from Abbott, Agios, Alexion Pharma, Alnylam, Amgen, Angionetics, ARCA Biopharma, Array, AstraZeneca, Atentiv, Audentes, Bayer, Better Therapeutics, Brigham and Women's Hospital, Bristol‐Myers Squibb, Cardiol Therapeutics, CellResearch, Cook Medical, Cook, CSL Behring, Eidos Therapeutics, EP Trading Co, Esperion Therapeutics, EverlyWell, Faraday, Fortress. Biotech, HDL Therapeutics, Heartflow, Hummingbird Bioscience, Insmed, Janssen, Kowa Research, Lexicon, Merck, Medtronic, Moderna, Novate Medical, Novo Nordisk, Pfizer, PhaseBio, PPD Development, Prairie Education and Research, Prothena Ciosciences, Regeneron, Regio Biosciences, Sanifit Therapeutics, Sanofi, Smith and Nephew, Stealth, BioTherapeutics, University of Colorado, Worldwide Clinical Trials, Wraser and Yale Cardiovascular Research Group. MPB also reports being a scientific advisor for the STRIDE Trial (supported by Novo Nordisk).

© 2022 The Authors. Diabetes, Obesity and Metabolism published by John Wiley & Sons Ltd.

Figures

FIGURE 1
FIGURE 1
Time to first MACE by history of PAD at baseline. A, LEADER; B, SUSTAIN 6. Data without adjustment for baseline variables are shown. HR, hazard ratio; CI, confidence interval; MACE, major adverse cardiovascular event; PAD, peripheral artery disease
FIGURE 2
FIGURE 2
Time to first MACE by history of PAD at baseline and randomized treatment group. A, LEADER; B, SUSTAIN 6. Data without adjustment for baseline variables are shown. ARR, absolute risk reduction at median follow‐up (3.8 years for LEADER; 2.1 years for SUSTAIN 6). CI, confidence interval; HR, hazard ratio; lira, liraglutide; MACE, major adverse cardiovascular event; N, number of patients; NNT, number needed to treat at median follow‐up (3.8 years for LEADER; 2.1 years for SUSTAIN 6); PAD, peripheral artery disease; sema, semaglutide
FIGURE 3
FIGURE 3
Risk of CV events and mortality by history of PAD at baseline and randomized treatment group. A, LEADER; B, SUSTAIN 6. Data without adjustment for baseline variables are shown. The primary outcome in both trials was a composite outcome including first occurrence of CV death, non‐fatal MI, or non‐fatal stroke., In SUSTAIN 6, expanded MACE included peripheral revascularization, which was site‐reported. CI, confidence interval; CV, cardiovascular; HF, heart failure; HR, hazard ratio; MACE, major adverse cardiovascular event; MI, myocardial infarction; n, number of patients with an event; PAD, peripheral artery disease; R, rate (events per 100 patient‐years of observation)
FIGURE 4
FIGURE 4
Time to first MACE by treatment arm and PAD only versus polyvascular disease (PAD and CAD and/or cerebrovascular disease) at baseline: A, LEADER; B, SUSTAIN 6. Kaplan–Meier plots have been truncated at A, 42 months for the LEADER trial and B, 80 weeks for the SUSTAIN 6 trial. CAD, coronary artery disease; MACE, major adverse cardiovascular event; PAD, peripheral artery disease

References

    1. American Diabetes Association . Peripheral arterial disease in people with diabetes. Diabetes Care. 2003;26:3333‐3341.
    1. Shah AD, Langenberg C, Rapsomaniki E, et al. Type 2 diabetes and incidence of cardiovascular diseases: a cohort study in 1.9 million people. Lancet Diabetes Endocrinol. 2015;3:105‐113.
    1. Silbernagel G, Rein P, Saely CH, et al. Prevalence of type 2 diabetes is higher in peripheral artery disease than in coronary artery disease patients. Diab Vasc Dis Res. 2015;12:146‐149.
    1. Khoury H, Lavoie L, Welner S, Folkerts K. The burden of major adverse cardiac events and antiplatelet prevention in patients with coronary or peripheral arterial disease. Cardiovasc Ther. 2016;34:115‐124.
    1. Verma S, Mazer CD, Al‐Omran M, et al. Cardiovascular outcomes and safety of empagliflozin in patients with type 2 diabetes mellitus and peripheral artery disease: a subanalysis of EMPA‐REG OUTCOME. Circulation. 2018;137:405‐407.
    1. Low Wang CC, Blomster JI, Heizer G, et al. Cardiovascular and limb outcomes in patients with diabetes and peripheral artery disease: the EUCLID trial. J Am Coll Cardiol. 2018;72:3274‐3284.
    1. Bonaca MP, Nault P, Giugliano RP, et al. Low‐density lipoprotein cholesterol lowering with evolocumab and outcomes in patients with peripheral artery disease: insights from the FOURIER trial (further cardiovascular outcomes research with PCSK9 inhibition in subjects with elevated risk). Circulation. 2018;137:338‐350.
    1. Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med. 2017;377:644‐657.
    1. Marso SP, Bain SC, Consoli A, et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2016;375:1834‐1844.
    1. Marso SP, Daniels GH, Brown‐Frandsen K, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375:311‐322.
    1. Gerstein HC, Colhoun HM, Dagenais GR, et al. Dulaglutide and cardiovascular outcomes in type 2 diabetes (REWIND): a double‐blind, randomised placebo‐controlled trial. Lancet. 2019;394:121‐130.
    1. Hernandez AF, Green JB, Janmohamed S, et al. Albiglutide and cardiovascular outcomes in patients with type 2 diabetes and cardiovascular disease (harmony outcomes): a double‐blind, randomised placebo‐controlled trial. Lancet. 2018;392:1519‐1529.
    1. Gerstein HC, Sattar N, Rosenstock J, et al. Cardiovascular and renal outcomes with efpeglenatide in type 2 diabetes. N Engl J Med. 2021;385:896‐907.
    1. Sattar N, Lee MMY, Kristensen SL, et al. Cardiovascular, mortality, and kidney outcomes with GLP‐1 receptor agonists in patients with type 2 diabetes: a systematic review and meta‐analysis of randomised trials. Lancet Diabetes Endocrinol. 2021;9:653‐662.
    1. American Diabetes Association . 10. Cardiovascular disease and risk management: standards of medical care in diabetes‐2021. Diabetes Care. 2021;44:S125‐S150.
    1. Diabetes Canada Clinical Practice Guidelines Expert Committee , Stone JA, Houlden RL, Lin P, Udell JA, Verma S. Cardiovascular protection in people with diabetes. Can J Diabetes. 2018;42(Suppl 1):S162‐S169.
    1. Cosentino F, Grant PJ, Aboyans V, et al. 2019 ESC guidelines on diabetes, pre‐diabetes, and cardiovascular diseases developed in collaboration with the EASD. Eur Heart J. 2020;41:255–323.
    1. Arnett DK, Blumenthal RS, Albert MA, et al. 2019 ACC/AHA guideline on the primary prevention of cardiovascular disease: a report of the American College of Cardiology/American Heart Association task force on clinical practice guidelines. Circulation. 2019;140:e596‐e646.
    1. Verma S, Bain SC, Buse JB, et al. Occurence of first and recurrent major adverse cardiovascular events with Liraglutide treatment among patients with type 2 diabetes and high risk of cardiovascular events: a post hoc analysis of a randomized clinical trial. JAMA Cardiol. 2019;4:1214‐1220.
    1. Altman DG, Andersen PK. Calculating the number needed to treat for trials where the outcome is time to an event. BMJ. 1999;319:1492‐1495.
    1. Verma S, Bhatt DL, Bain SC, et al. Effect of liraglutide on cardiovascular events in patients with type 2 diabetes mellitus and polyvascular disease: results of the LEADER trial. Circulation. 2018;137:2179‐2183.
    1. Bonaca MP, Gutierrez JA, Cannon C, et al. Polyvascular disease, type 2 diabetes, and long‐term vascular risk: a secondary analysis of the IMPROVE‐IT trial. Lancet Diabetes Endocrinol. 2018;6:934‐943.
    1. Cainzos‐Achirica M, Patel KV, Quispe R, et al. Coronary artery calcium for the allocation of GLP‐1RA for primary prevention of atherosclerotic cardiovascular disease. JACC Cardiovasc Imaging. 2021;14:1470‐1472.
    1. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373:2117‐2128.
    1. Hess DA, Terenzi DC, Trac JZ, et al. SGLT2 inhibition with empagliflozin increases circulating provascular progenitor cells in people with type 2 diabetes mellitus. Cell Metab. 2019;30:609‐613.
    1. Badjatiya A, Merrill P, Buse JB, et al. Clinical outcomes in patients with type 2 diabetes mellitus and peripheral artery disease: results from the EXSCEL trial. Circ Cardiovasc Interv. 2019;12:e008018.
    1. Lin DS, Lee JK, Chen WJ. Major adverse cardiovascular and limb events in patients with diabetes treated with GLP‐1 receptor agonists vs DPP‐4 inhibitors. Diabetologia. 2021;64:1949‐1962.
    1. Anand SS, Bosch J, Eikelboom JW, et al. Rivaroxaban with or without aspirin in patients with stable peripheral or carotid artery disease: an international, randomised, double‐blind, placebo‐controlled trial. Lancet. 2018;391:219‐229.
    1. Drucker DJ. The cardiovascular biology of glucagon‐like peptide‐1. Cell Metab. 2016;24:15‐30.
    1. Terenzi DC, Trac JZ, Teoh H, et al. Vascular regenerative cell exhaustion in diabetes: translational opportunities to mitigate cardiometabolic risk. Trends Mol Med. 2019;25:640‐655.
    1. Giblett JP, Clarke SJ, Dutka DP, Hoole SP. Glucagon‐like peptide‐1: a promising agent for cardioprotection during myocardial ischemia. JACC Basic Transl Sci. 2016;1:267‐276.
    1. Giblett JP, Axell RG, White PA, et al. Glucagon‐like peptide‐1‐mediated cardioprotection does not reduce right ventricular stunning and cumulative ischemic dysfunction after coronary balloon occlusion. JACC Basic Transl Sci. 2019;4:222‐233.
    1. Marx N, Libby P. Cardiovascular benefits of GLP‐1 receptor agonism: Is inflammation a key? JACC Basic Transl Sci. 2018;3:858‐860.
    1. Garg V, Verma S, Connelly K. Mechanistic insights regarding the role of SGLT2 inhibitors and GLP1 agonist drugs on cardiovascular disease in diabetes. Prog Cardiovasc Dis. 2019;62:349‐357.
    1. Sharma A, Verma S. Mechanisms by which glucagon‐like‐peptide‐1 receptor agonists and sodium‐glucose Cotransporter‐2 inhibitors reduce cardiovascular risk in adults with type 2 diabetes mellitus. Can J Diabetes. 2020;44:93‐102.
    1. Rakipovski G, Rolin B, Nohr J, et al. The GLP‐1 analogs liraglutide and semaglutide reduce atherosclerosis in ApoE(‐/‐) and LDLr(‐/‐) mice by a mechanism that includes inflammatory pathways. JACC Basic Transl Sci. 2018;3:844‐857.
    1. Verma S, Leiter LA, Mazer CD, et al. Liraglutide reduces cardiovascular events and mortality in type 2 diabetes mellitus independently of baseline Low‐density lipoprotein cholesterol levels and statin use. Circulation. 2018;138:1605‐1607.
    1. Verma S, McGuire DK, Bain SC, et al. Effects of glucagon‐like peptide‐1 receptor agonists liraglutide and semaglutide on cardiovascular and renal outcomes across body mass index categories in type 2 diabetes: results of the LEADER and SUSTAIN 6 trials. Diabetes Obes Metab. 2020;22:2487‐2492.
    1. Leiter LA, Bain SC, Bhatt DL, et al. The effect of glucagon‐like peptide‐1 receptor agonists liraglutide and semaglutide on cardiovascular and renal outcomes across baseline blood pressure categories: analysis of the LEADER and SUSTAIN 6 trials. Diabetes Obes Metab. 2020;22:1690‐1695.
    1. A/S NN A research study to compare a medicine called semaglutide against placebo in people with peripheral arterial disease and type 2 diabetes (STRIDE). 2020. . Accessed 04 October 2021.
    1. A/S NN Semaglutide effects on heart disease and stroke in patients with overweight or obesity (SELECT). 2019. . Accessed 04 October 2021.
    1. A/S NN A heart disease study of semaglutide in patients with type 2 diabetes (SOUL). 2019. . Accessed 04 October 2021.
    1. A/S NN A research study of how semaglutide works in people with disease affecting the heart and/or blood vessels and type 2 diabetes. 2019. . Accessed 04 October 2021.
    1. Anand SS, Caron F, Eikelboom JW, et al. Major adverse limb events and mortality in patients with peripheral artery disease: the COMPASS trial. J Am Coll Cardiol. 2018;71:2306‐2315.
    1. Dhatariya K, Bain SC, Buse JB, et al. The impact of liraglutide on diabetes‐related foot ulceration and associated complications in patients with type 2 diabetes at high risk for cardiovascular events: results from the LEADER trial. Diabetes Care. 2018;41:2229‐2235.
    1. Husain M, Birkenfeld AL, Donsmark M, et al. Oral semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2019;381:841‐851.

Source: PubMed

3
Subscribe