Review Article

Cardiovascular Outcomes in Trials of New Antidiabetic Drug Classes

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Information image
Average (ratings)
No ratings
Your rating

Abstract

Type 2 diabetes is among the most prevalent chronic diseases worldwide and the prevention of associated cardiovascular complications is an important treatment goal. Sodium–glucose co-transporter 2 (SGLT2) inhibitors, glucagon-like peptide 1 (GLP-1) receptor agonists and dipeptidyl peptidase-4 (DPP-4) inhibitors are second-line options after metformin, while cardiovascular outcome trials have been conducted to establish the cardiovascular safety of these antidiabetic drug classes. SGLT2 inhibitors have been shown to have the best overall mortality, renal and cardiovascular outcomes. Reduction in hospitalisation for heart failure is particularly consistent. GLP-1 receptor agonists have also showed some benefits, especially in stroke prevention. DPP-4 inhibitors showed neutral effects on cardiovascular outcomes, but may increase the incidence of heart failure. Favourable outcomes observed in trials of SGLT2 inhibitors mean that these should be the preferred second-line option. DPP-4 inhibitors are useful for patients with diabetes at low cardiovascular risk.

Disclosure:The authors have no conflicts of interest to declare.

Received:

Accepted:

Published online:

Correspondence Details:Bernard Cheung, University Department of Medicine, Queen Mary Hospital, 102 Pokfulam Rd, Hong Kong, China. E: mycheung@hku.hk

Open Access:

This work is open access under the CC-BY-NC 4.0 License which allows users to copy, redistribute and make derivative works for non-commercial purposes, provided the original work is cited correctly.

Type 2 diabetes (T2D) is becoming increasingly prevalent across the world. Its cardiovascular complications are major causes of mortality and use of medical resources.1 Prevention of cardiovascular diseases is, therefore, an important goal of the treatment of T2D. Metformin is the first-line therapy, according to the European Society of Cardiology, the American Diabetes Association (ADA) and the International Diabetes Federation.2–4 After metformin, three new antidiabetic drug classes have emerged as second-line therapy options.

Sodium–glucose co-transporter 2 (SGLT2) inhibitors exert hypoglycaemic effects by inhibiting glucose reabsorption at the proximal convoluted tubules, causing glycosuria, natriuresis and volume contraction (Figure 1).5

Besides SGLT2 inhibition, incretin-based therapies have also been used in recent years for the treatment of T2D (Figure 2). Incretins are gut-derived hormones that send a signal to the pancreas after the ingestion of food.6,7 There are two main incretin hormones: glucagon-like peptide 1 (GLP-1) and gastric inhibitory polypeptide (GIP, also known as glucose-dependent insulinotropic polypeptide). Both are secreted by enteroendocrine cells in the intestines and stimulate pancreatic beta-cells to secrete insulin.7 GLP-1 additionally inhibits glucagon release by pancreatic alpha-cells and delays gastric emptying.6 GIP, on the other hand, also stimulates glucagon production, yet fails to stimulate insulin secretion in people with diabetes.7 Therefore, it has not been developed as a therapeutic agent. The plasma half-life of GLP-1 is short, so GLP-1 receptor agonists (RAs) have modifications in the peptide to prolong half-life.6 As these incretin hormones are degraded by dipeptidyl peptidase-4 (DPP-4), DPP-4 inhibitors can amplify their pharmacological actions.8 Both of these two incretin-based therapies improve postprandial glucose control.6,8

Among the three drug classes, the preferred second-line treatment remains unclear (Table 1). However, the thiazolidinediones are not favoured as second-line drugs. Indeed, pioglitazone-induced heart failure (HF) and the withdrawal of rosiglitazone because of cardiovascular concerns eventually led to a change in the policy of regulatory authorities.9,10

The US Food and Drug Administration and the European Medicines Agency now require all new antidiabetic drugs to undergo large cardiovascular outcome trials (CVOTs) to confirm cardiovascular safety and benefits. As a result of this requirement, multiple CVOTs have been published in recent years (Table 2). Some trials have shown cardiovascular benefits for GLP-1 RAs and SGLT2 inhibitors, which have been confirmed in meta-analyses.11-18 However, their effects on particular outcomes remain inconsistent in trials.15,19,20 This may be a result of limitations in statistical power and differences in patient characteristics and the drugs used. In the absence of adequately powered head-to-head trials, superiority amongst the three antidiabetic drug classes cannot be established.

Network meta-analyses (NMAs) can evaluate comparative risks or benefits using indirect evidence. Our 2019 NMA included 14 trials and a total of 121,047 patients.17 First, we found that both SGLT2 inhibitors and GLP-1 RAs significantly reduced major adverse cardiovascular events (MACE), hospitalisation for HF and renal composite outcome compared to placebo. Second, SGLT2 inhibitors were shown to have the greatest cardiovascular and all-cause mortality benefits amongst all three new antidiabetic drug classes. Third, the GLP-1 RA class was the only one that showed reductions in nonfatal stroke events. Finally, the risks of cardiovascular and renal outcomes in DPP-4 inhibitors were found to be neutral when compared to placebo and inferior to the other two drug classes.

Postulated Mechanisms of SGLT2 Inhibitors

Article image

SGLT2 Inhibitors

The EMPA-REG OUTCOME and CANVAS studies have shown favourable all-cause mortality and cardiovascular outcomes.11,12 Meta-analyses of CVOTs confirmed the cardiovascular benefits.17,18 Three studies from landmark CVOTs further stratified patients according to baseline characteristics.21,22,23 The benefits of SGLT2 inhibitors were more apparent in patients with a history of HF and reduced ejection fraction.22,23 However, baseline risk was higher in this group of patients, possibly accounting for higher absolute risk reductions in cardiovascular outcomes. The DECLARE-TIMI 58 trial enrolled a majority of patients who did not have established atherosclerotic cardiovascular disease (ASCVD), and did not show significant reduction in MACE.19 This may imply the cardioprotection offered by SGLT2 inhibitors is less evident in such a group of patients .18 However, analysis from the EMPA-REG OUTCOME trial reported consistent benefits regardless of baseline risks and prior history.21 Neither of these trials was designed to investigate HF outcomes, but they illustrated the value in identifying patient subgroups that benefit most from SGLT2 inhibitor treatment.

SGLT2 inhibitors are postulated to induce selective volume contraction.24 Thus, there is a selective reduction of interstitial fluid in contrast to traditional diuretics.24 Therefore, intravascular volume depletion and fluid retention by the renin–angiotensin–aldosterone system (RAAS) in the long run are limited.25 SGLT2 inhibitors also inhibit myocardial Na+/H+ exchanger (NHE) 1,5 which has been hypothesised to attenuate cardiac hypertrophy and HF development.26 Natriuresis is reported to improve left ventricular (LV) preload conditions, which could be potentiated by additional NHE-3 inhibition at the proximal tubules. 5,27 Furthermore, SGLT2 inhibitors reduce sympathetic tone and blood pressure (BP), which would improve afterload. 5,28,29 SGLT2 inhibitors are therefore particularly useful for diabetic patients with HF.3 Moreover, attenuation in endothelial dysfunction and arterial stiffness by reducing oxidative stress has also been reported, bringing potential benefits in vascular diseases.29,30 Dapagliflozin and empagliflozin have also shown anti-fibrotic effects in rats and human cardiac fibroblasts.31,32 Importantly, SGLT2 inhibitors induce weight loss and increase HDL levels in trials.11,12,26 They also increase lipolysis and reduce inflammation in adipose tissues.33 The switch from utilisation of glucose to ketones is also believed to be beneficial, especially for cardiac metabolism.5 All of these mechanisms may help to explain the cardiovascular benefits observed.11,12

Incretin Physiology and Mechanisms of Action of DPP-4 Inhibitors and GLP-1 Receptor Agonists

Article image

The DAPA-HF and the EMPEROR-Reduced (NCT03057977) trials showed statistically significant reductions in HF hospitalisation, regardless of history of diabetes.34 However, the change in mortality did not reach statistical significance in EMPEROR-Reduced.35 The cardioprotective mechanisms independent of glucose lowering are still not fully understood. The EMPA-HEART trial showed reduction in LV mass and systolic BP in HF with reduced ejection fraction (HFrEF) patients, although the effects on N-terminal pro-B-type natriuretic peptide (NT-proBNP) was insignificant.36 The EMPERIAL trials (NCT03448419 and NCT03448406) reported no significant improvements in exercise capacity measured by 6-minute walking distance. Improvements in quality of life scores were found in patients with HFrEF but not in those with HF with preserved ejection fraction. Results from on-going EMPA-KIDNEY (NCT03594110), DELIVER (NCT0319213), SMARTEST (NCT03982381), as well as the recently terminated SCORED (NCT03315143) and SOLOIST-WHF (NCT03521934) trials will further elucidate the cardioprotective effects and identify patients who may benefit most from treatment with SGLT2 inhibitors.

SGLT2 inhibitors have shown clear-cut renal benefits in the CREDENCE and DAPA-CKD (NCT03036150) trials, which have also been confirmed in meta-analyses.17,37 The natriuresis induced by SGLT2 inhibitors stimulates tubuloglomerular feedback and vasoconstriction in afferent arterioles, thus reducing glomerular hyperfiltration and albuminuria.26,38,39 Concomitant use of RAAS inhibitors is reported to have synergistic effects on renal tubules, slowing down the progression of chronic kidney disease (CKD).40 The aforementioned shift in metabolic energetics is believed to inhibit oxidative stress and ischaemic injury, not only in myocardium but also in renal tubules.29,38 Reduction in BP and body weight as well as increase in uric acid excretion are all postulated as renoprotective mechanisms.39

The benefits of SGLT2 inhibitors are not always apparent. The effect of SGLT2 inhibitors on stroke events appears to be neutral or even slightly harmful. The EMPA-REG OUTCOME trial detected slight elevations in nonfatal stroke events.11 In a subgroup analysis of the CANVAS trial, the results showed protective effects against haemorrhagic stroke, yet neutral effects on ischaemic stroke (HR 0.95; 95% CI [0.74–1.22]).41 BP reduction may account for the prevention of haemorrhagic stroke.42 However, elevated haematocrit was also observed in the EMPA-REG OUTCOME trial, which might predispose to ischaemic strokes.11,42,43 Although haemoconcentration improves cardiovascular outcomes, the higher blood viscosity may trigger thrombus formation.27,42–44 Although weight loss should protect against ischaemic stroke, compensatory changes may limit weight loss in the long term.33 Although a harmful effect on stroke has not been confirmed, the protective effect on haemorrhagic stroke may mask the effect on ischaemia if both types of stroke are included as a composite endpoint.16–18 Population-wide observational studies may help to evaluate the long-term risk of ischaemic strokes during SGLT2 inhibitor therapy.

Current Recommendations on Antidiabetic Drugs

Article image

Overall, SGLT2 inhibitors have demonstrated the most favourable cardiovascular and all-cause mortality outcomes. Indirect evidence showed overall superiority over DPP-4 inhibitors, which appears to be a class effect.16,17 However, superiority between GLP-1 RAs and SGLT2 inhibitors varies across different outcomes. SGLT2 inhibitors are superior in terms of mortality outcomes, yet protection against MACE was similar among GLP-1 RAs and SGLT2 inhibitors.16,17 Also, mortality benefits did not reach statistical significance in the CREDENCE and DECLARE-TIMI 58 trials.19,37 The CREDENCE trial was prematurely terminated because of overwhelming renal and cardiovascular benefits. A shortened follow-up period limited the power of the study to detect changes in mortality, if present. Besides the CREDENCE trial, other non-empagliflozin trials also failed to show significant changes.12,19 Trials of non-empagliflozin SGLT2 inhibitors investigating mortality endpoints are needed to fill in the evidence gap. The combination of RAAS and SGLT2 inhibitors should be advocated as concomitant inhibition appears to achieve better cardiovascular and renal outcomes.40

GLP-1 Receptor Agonists

The LEADER, HARMONY OUTCOMES and EXSCEL trials demonstrated the cardiovascular safety of GLP-1 RAs.13–15 Meta-analyses have reported favourable cardiovascular safety profiles for GLP-1 RAs, especially in MACE and nonfatal stroke events.16,17,45 Improvements in the composite kidney outcome were also detected.45 However, the benefits were less clear-cut in comparison with SGLT2 inhibitors and somewhat conflicting across trials. Discrepancies in mortality-related outcomes could be due to differences in follow-up periods and study population. Intraclass differences and variations in pharmacokinetics are also possible explanations.

Liraglutide, a long-acting GLP-1 RA, yielded favourable results in the LEADER trial.13 In addition to glycaemic control, slowing of atherosclerosis and anti-inflammatory actions also account for the benefits of GLP-1 agonism. The interaction between GLP-1 RAs and cardiac GLP-1 receptors has been suggested to improve myocardial ischaemia and protect cardiac progenitor cells.46 They also exert protective effects independent of GLP-1 receptors.6 Liraglutide has been reported to induce endothelial nitric oxide synthase via AMP-activated protein kinase signalling.6 Nitric oxide production improves coronary artery flow and endothelial dysfunction. Besides, GLP-1 RAs inhibit mitochondrial oxidative damage and attenuate reactive oxygen species production.47 Liraglutide has also been shown to suppress vascular cell adhesion molecule-1 expression in the endothelium.6 It also improves arterial stiffness and LV function, while reducing NT-proBNP levels, a biomarker for LV dysfunction.48 Vasodilatory and antioxidant actions could account for some of the antiatherogenic effects in GLP-1 RAs, therefore this drug class may be preferable in diabetic patients with predominant ASCVD risk.3

However, different results were found in studies of exenatide.15,20 Intravenous exenatide in patients after coronary artery bypass grafting surgery did not offer additional cardiovascular benefits compared to parenteral insulin.20 Differences have been suggested to be related to different immunogenicity profiles and signalling pathways in exendin-4 and GLP-1 based agonists.46 Exendin-4 based agonists are postulated to be more immunogenic and cause injection site reactions, leading to higher drug discontinuation rate and diminished benefits in the EXSCEL trial.15,46 Given the conflicting evidence, CVOTs were conducted to study GLP-1 RAs with different populations and formulations. The FREEDOM-CVO and REWIND trials included injection-free GLP-1 RA with osmotic mini-pump and patients without established cardiovascular background respectively, whereas the PIONEER 6 study investigated oral semaglutide.49–51 Mortality outcomes in some of these trials are encouraging, yet the results are inconsistent.49,51

GLP-1 RAs uniquely reduced nonfatal stroke events in the LEADER and SUSTAIN-6 trials.13,52 The effects were confirmed in our latest NMA (OR 0.88; 95% CI [0.77–0.99]).17 GLP-1 RAs are reported to be neuroprotective because of the ability to cross the blood-brain barrier and actions on neuroinflammation pathways.47 Besides atherosclerosis, oxidative stress is considered to be responsible for stroke development.53 GLP-1 RAs reduce oxidative stress and reactive oxygen species production via p-AKT/endothelial nitric oxide synthase and nuclear factor-κ B p65 pathways.47

Completed and On-going CVOTs of New Antidiabetic Drugs

Article image

Completed and On-going CVOTs of New Antidiabetic Drugs Cont.

Article image

CVOTs are being conducted in order to expand the indication of GLP-1 RAs from ASCVD prevention in diabetes to a broader patient population. Results from the FIGHT trial did not support the use of liraglutide in HF patients, which is not surprising given less clear-cut HF benefits in NMA compared to SGLT2 inhibitors.17,54 The on-going EGRABIS1 (NCT02829502) and Lirabolic (NCT04057261) trials also aim to explore the effects of GLP-1 RAs on cardiometabolic markers, such as mean cerebral flow velocity, as well as BP and lipid profiles. Together with the SLIM LIVER (NCT04216589) study in non-alcoholic fatty liver disease patients, these trials will clarify the neuroprotective and cardioprotective mechanisms of GLP-1 RAs independent of glucose levels. This might provide the scientific basis for the benefits of this drug class in the prevention of vascular diseases.

DPP-4 Inhibitors

DPP-4 inhibitors did not show cardiovascular benefits in meta-analyses by other researchers and ourselves.16,17,55 A previous meta-analysis concluded significant reductions in MACE, but the inclusion of trials with shorter follow-up periods may limit the robustness of the conclusions. Elevated hospitalisation for HF incidence remains a concern, as reported in the SAVOR-TIMI 53 trial.55–57 However, in the EXAMINE and the latest CARMELINA trials, HF incidences were neutral.58,59

DPP-4 inhibitors have been reported to inhibit degradation of endogenous peptides other than GLP-1, including stromal-cell-derived factor (SDF) 1, neuropeptide Y and substance P (Figure 2).60 Potentiation of these peptides results in sympathetic activation via cyclic AMP signalling and beta-receptor activation, which may contribute to HF development and cardiac fibrosis. Elevated SDF-1 and substance P levels are also suggested to induce apoptosis of the myocardium.60 In the SAVOR-TIMI 53 trial, attenuated HF elevation was detected when beta-blockers were used concomitantly.58,60 This might imply that chronic sympathetic activation might underlie the inferiority of DPP-4 inhibitors. Notably, most patients enrolled in trials with neutral HF outcomes have concomitant prescriptions for RAAS inhibitors, which could ameliorate the tendency of DPP-4 inhibitors to worsen HF.58,59 Combination therapy may counteract the detrimental effects of DPP-4 inhibitors, thus masking the harmful effects. In real life, patient with diabetes are often receiving a RAAS blocker. This makes the drug class useful even in dialysis patients.8 Nevertheless, DPP-4 inhibitors should be used with caution in patients with HF or at risk of HF.

Clinical Implications

Observational studies have been conducted to provide insights into the clinical roles of the new antidiabetic drugs.61,62 A retrospective cohort study has demonstrated beneficial safety profiles over sulphonylureas, whereas O’Brien et al. reported similar improvements in cardiovascular outcomes across the three drug classes.61,62 However, it must be noted that selection bias could be prominent in observational studies, as the number of patients included in the study was significantly higher for those receiving DPP-4 inhibitors (28,898) than for SGLT2 inhibitors (5,677) or GLP-1 RAs (11,351).62 The lack of patients enrolled in specific groups also make the use of propensity score matching difficult. Nevertheless, these studies are still valuable because head-to-head randomised controlled trials are lacking. Population-wide observational studies provide a good and generalisable estimation of clinical effects with modest confidence, thus defining the clinical roles of each antidiabetic drug class more clearly.

Choosing an antidiabetic drug class with proven cardiovascular and mortality benefits is now advocated in guidelines.3,4,63 SGLT2 inhibitors and GLP-1 RAs should be recommended as second-line treatment options after metformin, echoing recommendations by the ADA and the American College of Cardiology.3,63 The choice between SGLT2 inhibitors and GLP-1 RAs should be tailor-made according to patient characteristics. SGLT2 inhibitors offered more overall mortality benefits. They should be considered superior in most patients, including those with HF and CKD.3 However, ischaemia prevention is superior for GLP-1 RAs, thus making them preferable in patients with predominant ASCVD risk.3 Differences in routes of administration and adverse effect profiles may also play a role in prescription decisions. Most GLP-1 RAs are available as subcutaneous injections, which are inconvenient. Oral semaglutide might make this class attractive to use, but it is expensive. On-going trials, including head-to-head trials, are required to address comparative safety and efficacy, as well as possible intraclass differences.

Conclusion

Current evidence confirmed the cardiovascular safety of the three new antidiabetic drug classes, but it is important to appreciate the differences among them. SGLT2 inhibitors show superiority in mortality and cardiovascular events, mainly driven by hospitalisation for HF, and renal events. GLP-1 RAs can reduce nonfatal stroke and MACE, yet inconsistent evidence suggests possible intraclass differences. Both drug classes should now be considered as the preferred second-line treatment in T2D patients after metformin according to patient characteristics. In terms of cardiovascular and renal outcomes, DPP-4 inhibitors have not demonstrated benefits in comparison with placebo and have been proven to be inferior to GLP-1 RAs and SGLT2 inhibitors. In this new era, antidiabetic drugs no longer just control blood glucose, but should also address the cardiovascular risks of T2D patients.

References

  1. Morrish NJ, Wang SL, Stevens LK, et al. Mortality and causes of death in the WHO Multinational Study of Vascular Disease in Diabetes. Diabetologia 2001;44:S14–21.
    Crossref | PubMed
  2. 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.
    Crossref | PubMed
  3. American Diabetes Association. 9. Pharmacologic approaches to glycemic treatment: standards of medical care in diabetes – 2020. Diabetes Care 2020;43:S98–S110.
    Crossref | PubMed
  4. International Diabetes Federation. Recommendations For Managing Type 2 Diabetes In Primary Care. IDF, 2017. https://www.idf.org/e-library/guidelines/128-idf-clinical-practice-recommendations-for-managing-type-2-diabetes-in-primary-care.html (accessed 11 December 2020).
  5. Verma S, McMurray JJV. SGLT2 inhibitors and mechanisms of cardiovascular benefit: a state-of-the-art review. Diabetologia 2018;61:2108–17.
    Crossref | PubMed
  6. Sposito AC, Berwanger O, de Carvalho LSF, et al. GLP-1RAs in type 2 diabetes: mechanisms that underlie cardiovascular effects and overview of cardiovascular outcome data. Cardiovasc Diabetol 2018;17:157.
    Crossref | PubMed
  7. Campbell JE, Drucker DJ. Pharmacology, physiology, and mechanisms of incretin hormone action. Cell Metab 2013;17:819–37.
    Crossref | PubMed
  8. Deacon CF. Dipeptidyl peptidase-4 inhibitors in the treatment of type 2 diabetes: a comparative review. Diabetes Obes Metab 2011;13:7–18.
    Crossref | PubMed
  9. Lincoff AM, Wolski K, Nicholls SJ, et al. Pioglitazone and risk of cardiovascular events in patients with type 2 diabetes mellitus: a meta-analysis of randomized trials. JAMA 2007;298:1180–8.
    Crossref | PubMed
  10. Cheung BM. Behind the rosiglitazone controversy. Expert Rev Clin Pharmacol 2010;3:723–5.
    Crossref | PubMed
  11. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 2015;373:2117–28.
    Crossref | PubMed
  12. Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med 2017;377:644–57.
    Crossref | PubMed
  13. Marso SP, Daniels GH, Brown-Frandsen K, et al. Liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2016;375:311–22.
    Crossref | PubMed
  14. 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;392:1519–29.
    Crossref | PubMed
  15. Mentz RJ, Thompson VP, Aguilar D, et al. Effects of once-weekly exenatide on clinical outcomes in patients with preexisting cardiovascular disease. Circulation 2018;138:2576–8.
    Crossref | PubMed
  16. Fei Y, Tsoi MF, Kumana CR, et al. Network meta-analysis of cardiovascular outcomes in randomized controlled trials of new antidiabetic drugs. Int J Cardiol 2018;254:291–6.
    Crossref | PubMed
  17. Fei Y, Tsoi MF, Cheung BMY. Cardiovascular outcomes in trials of new antidiabetic drug classes: a network meta-analysis. Cardiovasc Diabetol 2019;18:112.
    Crossref | PubMed
  18. Zelniker TA, Wiviott SD, Raz I, et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet 2019;393:31–9.
    Crossref | PubMed
  19. Wiviott SD, Raz I, Bonaca MP, et al. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med 2019;380:347–57.
    Crossref | PubMed
  20. Besch G, Perrotti A, Salomon du Mont L, et al. Impact of intravenous exenatide infusion for perioperative blood glucose control on myocardial ischemia-reperfusion injuries after coronary artery bypass graft surgery: sub study of the phase II/III ExSTRESS randomized trial. Cardiovasc Diabetol 2018;17:140.
    Crossref | PubMed
  21. Fitchett D, Butler J, van de Borne P, et al. Effects of empagliflozin on risk for cardiovascular death and heart failure hospitalization across the spectrum of heart failure risk in the EMPA-REG OUTCOME® trial. Eur Heart J 2018;39:363–70.
    Crossref | PubMed
  22. Kato ET, Silverman MG, Mosenzon O, et al. Effect of dapagliflozin on heart failure and mortality in type 2 diabetes mellitus. Circulation 2019;139:2528–36.
    Crossref | PubMed
  23. Rådholm K, Figtree G, Perkovic V, et al. Canagliflozin and heart failure in type 2 diabetes mellitus: Results from the CANVAS program. Circulation 2018;138:458–68.
    Crossref | PubMed
  24. Hallow KM, Helmlinger G, Greasley PJ, et al. Why do SGLT2 inhibitors reduce heart failure hospitalization? A differential volume regulation hypothesis. Diabetes Obes Metab 2018;20:479–87.
    Crossref | PubMed
  25. Schork A, Saynisch J, Vosseler A, et al. Effect of SGLT2 inhibitors on body composition, fluid status and renin-angiotensin-aldosterone system in type 2 diabetes: a prospective study using bioimpedance spectroscopy. Cardiovasc Diabetol 2019;18:46.
    Crossref | PubMed
  26. Packer M, Anker SD, Butler J, et al. Effects of sodium-glucose cotransporter 2 inhibitors for the treatment of patients with heart failure: proposal of a novel mechanism of action. JAMA Cardiol 2017;2:1025–9.
    Crossref | PubMed
  27. Santos-Ferreira D, Gonçalves-Teixeira P, Fontes-Carvalho R. SGLT-2 inhibitors in heart failure and type-2 diabetes: hitting two birds with one stone? Cardiology 2020;145:311–20.
    Crossref | PubMed
  28. Briasoulis A, Al Dhaybi O, Bakris GL. SGLT2 inhibitors and mechanisms of hypertension. Curr Cardiol Rep 2018;20:1.
    Crossref | PubMed
  29. Kaplan A, Abidi E, El-Yazbi A, et al. Direct cardiovascular impact of SGLT2 inhibitors: mechanisms and effects. Heart Fail Rev 2018;23:419–37.
    Crossref | PubMed
  30. Pulakazhi Venu VK, El-Daly M, Saifeddine M, et al. Minimizing hyperglycemia-induced vascular endothelial dysfunction by inhibiting endothelial sodium-glucose cotransporter 2 and attenuating oxidative stress: implications for treating individuals with type 2 diabetes. Can J Diabetes 2019;43:510–4.
    Crossref | PubMed
  31. Lee TM, Chang NC, Lin SZ. Dapagliflozin, a selective SGLT2 Inhibitor, attenuated cardiac fibrosis by regulating the macrophage polarization via STAT3 signaling in infarcted rat hearts. Free Radic Biol Med 2017;104:298–310.
    Crossref | PubMed
  32. Kang S, Verma S, Hassanabad AF, et al. Direct effects of empagliflozin on extracellular matrix remodelling in human cardiac myofibroblasts: novel translational clues to explain EMPA-REG OUTCOME results. Can J Cardiol 2020;36:543–53.
    Crossref | PubMed
  33. Pereira MJ, Eriksson JW. Emerging role of SGLT-2 inhibitors for the treatment of obesity. Drugs 2019;79:219–30.
    Crossref | PubMed
  34. McMurray JJV, Solomon SD, Inzucchi SE, et al. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med 2019;381:1995–2008.
    Crossref | PubMed
  35. Packer M, Anker SD, Butler J, et al. Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med 2020 Oct 8;383(15):1413-1424.
    PubMed
  36. Verma S, Mazer CD, Yan AT, et al. Effect of empagliflozin on left ventricular mass in patients with type 2 diabetes mellitus and coronary artery disease: the EMPA-HEART CardioLink-6 randomized clinical trial. Circulation 2019;140:1693–702.
    Crossref | PubMed
  37. Perkovic V, Jardine MJ, Neal B, et al. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med 2019;380:2295–306.
    Crossref | PubMed
  38. de Albuquerque Rocha N, Neeland IJ, McCullough PA, et al. Effects of sodium glucose co-transporter 2 inhibitors on the kidney. Diab Vasc Dis Res 2018;15:375–86.
    Crossref | PubMed
  39. Georgianos PI, Divani M, Eleftheriadis T, et al. SGLT-2 inhibitors in diabetic kidney disease: what lies behind their renoprotective properties? Curr Med Chem 2019;26:5564–78.
    Crossref | PubMed
  40. Zou H, Zhou B, Xu G. SGLT2 inhibitors: a novel choice for the combination therapy in diabetic kidney disease. Cardiovasc Diabetol 2017;16:65.
    Crossref | PubMed
  41. Zhou Z, Lindley RI, Rådholm K, et al. Canagliflozin and stroke in type 2 diabetes mellitus. Stroke 2019;50:396–404.
    Crossref | PubMed
  42. Imprialos KP, Boutari C, Stavropoulos K, et al. Stroke paradox with SGLT-2 inhibitors: a play of chance or a viscosity-mediated reality? J Neurol Neurosurg Psychiatry 2017;88:249–53.
    Crossref | PubMed
  43. Jin YZ, Zheng DH, Duan ZY, et al. Relationship between hematocrit level and cardiovascular risk factors in a community-based population. J Clin Lab Anal 2015;29:289–93.
    Crossref | PubMed
  44. Inzucchi SE, Zinman B, Fitchett D, et al. How does empagliflozin reduce cardiovascular mortality? Insights from a mediation analysis of the EMPA-REG OUTCOME trial. Diabetes Care 2018;41:356–63.
    Crossref | PubMed
  45. Kristensen SL, Rørth R, Jhund PS, 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 cardiovascular outcome trials. Lancet Diabetes Endocrinol 2019;7:776–85.
    Crossref | PubMed
  46. Caruso I, Cignarelli A, Giorgino F. Heterogeneity and similarities in GLP-1 receptor agonist cardiovascular outcomes trials. Trends Endocrinol Metab 2019;30:578–89.
    Crossref | PubMed
  47. Oh YS, Jun HS. Effects of glucagon-like peptide-1 on oxidative stress and Nrf2 signaling. Int J Mol Sci 2017;19:26.
    Crossref | PubMed
  48. Lambadiari V, Pavlidis G, Kousathana F, et al. Effects of 6-month treatment with the glucagon like peptide-1 analogue liraglutide on arterial stiffness, left ventricular myocardial deformation and oxidative stress in subjects with newly diagnosed type 2 diabetes. Cardiovasc Diabetol 2018;17:8.
    Crossref | PubMed
  49. Intarcia Therapeutics. Intarcia announces successful cardiovascular safety results in phase 3 FREEDOM-CVO trial for ITCA 650, an investigational therapy for type 2 diabetes. PR Newswire 6 May 2016. https://www.prnewswire.com/news-releases/intarcia-announces-successful-cardiovascular-safety-results-in-phase-3-freedom-cvo-trial-for-itca-650-an-investigational-therapy-for-type-2-diabetes-300264245.html (accessed 20 January 2021).
  50. 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;394:121–30.
    Crossref | PubMed
  51. Husain M, Birkenfeld AL, Donsmark M, et al. Oral semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 2019;381:841–51.
    Crossref | PubMed
  52. Marso SP, Bain SC, Consoli A, et al. Semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med 2016;375:1834–44.
    Crossref | PubMed
  53. Allen CL, Bayraktutan U. Oxidative stress and its role in the pathogenesis of ischaemic stroke. Int J Stroke 2009;4:461–70.
    Crossref | PubMed
  54. Margulies KB, Hernandez AF, Redfield MM, et al. Effects of liraglutide on clinical stability among patients with advanced heart failure and reduced ejection fraction: A randomized clinical trial. JAMA 2016;316:500–8.
    Crossref | PubMed
  55. Rehman MB, Tudrej BV, Soustre J, et al. Efficacy and safety of DPP-4 inhibitors in patients with type 2 diabetes: meta-analysis of placebo-controlled randomized clinical trials. Diabetes Metab 2017;43:48–58.
    Crossref | PubMed
  56. Monami M, Ahrén B, Dicembrini I, et al. Dipeptidyl peptidase-4 inhibitors and cardiovascular risk: a meta-analysis of randomized clinical trials. Diabetes Obes Metab 2013;15:112–20.
    Crossref | PubMed
  57. Scirica BM, Bhatt DL, Braunwald E, et al. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med 2013;369:1317–26.
    Crossref | PubMed
  58. Zannad F, Cannon CP, Cushman WC, et al. Heart failure and mortality outcomes in patients with type 2 diabetes taking alogliptin versus placebo in EXAMINE: a multicentre, randomised, double-blind trial. Lancet 2015;385:2067–76.
    Crossref | PubMed
  59. Rosenstock J, Perkovic V, Johansen OE, et al. Effect of linagliptin vs placebo on major cardiovascular events in adults with type 2 diabetes and high cardiovascular and renal risk: the CARMELINA randomized clinical trial. JAMA 2019;321:69–79.
    Crossref | PubMed
  60. Packer M. Do DPP-4 inhibitors cause heart failure events by promoting adrenergically mediated cardiotoxicity? Clues from laboratory models and clinical trials. Circ Res 2018;122:928–32.
    Crossref | PubMed
  61. Elharram M, Moura CS, Abrahamowicz M, et al. Novel glucose lowering agents are associated with a lower risk of cardiovascular and adverse events in type 2 diabetes: a population based analysis. Int J Cardiol 2020;310:147–54.
    Crossref | PubMed
  62. O’Brien MJ, Karam SL, Wallia A, et al. Association of second-line antidiabetic medications with cardiovascular events among insured adults with type 2 diabetes. JAMA Netw Open 2018;1:e186125.
    Crossref | PubMed
  63. Das SR, Everett BM, Birtcher KK, et al. 2018 ACC expert consensus decision pathway on novel therapies for cardiovascular risk reduction in patients with type 2 diabetes and atherosclerotic cardiovascular disease: a report of the American College of Cardiology Task Force on Expert Consensus Decision Pathways. J Am Coll Cardiol 2018;72:3200–23.
    Crossref | PubMed
  64. National Institute for Health and Care Excellence. Type 2 diabetes in adults: management. London: NICE, 2015. https://www.nice.org.uk/guidance/ng28 (accessed 11 December 2020).
  65. Cannon CP, McGuire DK, Pratley R, et al. Design and baseline characteristics of the evaluation of ertugliflozin efficacy and safety cardiovascular outcomes trial (VERTIS-CV). Am Heart J 2018;206:11–23.
    Crossref | PubMed
  66. Pfeffer MA, Claggett B, Diaz R, et al. Lixisenatide in patients with type 2 diabetes and acute coronary syndrome. N Engl J Med 2015;373:2247–57.
    Crossref | PubMed
  67. Green JB, Bethel MA, Armstrong PW, et al. Effect of sitagliptin on cardiovascular outcomes in type 2 diabetes. N Engl J Med 2015;373:232–42.
    Crossref | PubMed