Review Open Access
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Cardiol. May 26, 2024; 16(5): 240-259
Published online May 26, 2024. doi: 10.4330/wjc.v16.i5.240
Sodium glucose cotransporter-2 inhibitors and heart disease: Current perspectives
Sunetra Mondal, Vibhu Ranjan Khare, Department of Endocrinology, NRS Medical College, Kolkata 700020, West Bengal, India
Subhodip Pramanik, Department of Endocrinology, Neotia Getwel Multispecialty Hospitals, Siliguri 734010, West Bengal, India
Cornelius James Fernandez, Department of Endocrinology and Metabolism, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston PE21 9QS, United Kingdom
Joseph M Pappachan, Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston PR2 9HT, United Kingdom
Joseph M Pappachan, Faculty of Science, Manchester Metropolitan University, Manchester M15 6BH, United Kingdom
Joseph M Pappachan, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M13 9PL, United Kingdom
ORCID number: Sunetra Mondal (0000-0003-3064-466X); Subhodip Pramanik (0000-0002-3196-1192); Cornelius James Fernandez (0000-0002-1171-5525); Joseph M Pappachan (0000-0003-0886-5255).
Author contributions: Mondal S contributed to the initial drafting of the work by performing the literature search and interpretation of relevant literature with help from Pramanik S and Khare VR; Fernandez CJ prepared the figures for the manuscript and contributed additionally to the drafting process; Pappachan JM conceptualized the idea and provided overall supervision to the drafting process; and all authors contributed to the revision of the article for important intellectual content; All authors have read and approved the final version of the manuscript.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Joseph M Pappachan, MD, FRCP, Academic Editor, Professor, Department of Endocrinology and Metabolism, Lancashire Teaching Hospitals NHS Trust, Sharoe Green Lane, Preston PR2 9HT, United Kingdom. drpappachan@yahoo.co.in
Received: December 25, 2023
Revised: April 8, 2024
Accepted: April 28, 2024
Published online: May 26, 2024
Processing time: 149 Days and 20.9 Hours

Abstract

Sodium glucose cotransporter-2 inhibitors (SGLT-2i) are antidiabetic medications with remarkable cardiovascular (CV) benefits proven by multiple randomised controlled trials and real-world data. These drugs are also useful in the prevention of CV disease (CVD) in patients with diabetes mellitus (DM). Although DM as such is a huge risk factor for CVD, the CV benefits of SGLT-2i are not just because of antidiabetic effects. These molecules have proven beneficial roles in prevention and management of nondiabetic CVD and renal disease as well. There are various molecular mechanisms for the organ protective effects of SGLT-2i which are still being elucidated. Proper understanding of the role of SGLT-2i in prevention and management of CVD is important not only for the cardiologists but also for other specialists caring for various illnesses which can directly or indirectly impact care of heart diseases. This clinical review compiles the current evidence on the rational use of SGLT-2i in clinical practice.

Key Words: SGLT2 inhibitors, SGLT2i, Cardiovascular disease, Heart failure, Atherosclerotic cardiovascular disease, Diabetic kidney disease

Core Tip: The new antidiabetic medication class sodium glucose cotransporter-2 inhibitors (SGLT-2i) are found to have remarkable cardiovascular (CV) benefits proven by multiple randomised controlled trials and real-world observational studies. They are also useful in prevention of CV disease (CVD) in patients with diabetes mellitus. The CV benefits of SGLT-2i are not just because of antidiabetic effects. The preventive and management effects of SGLT2i molecules in diabetic and nondiabetic renal disease also translate into CV benefits. This clinical update review compiles the up-to-date evidence on the molecular mechanisms of SGLT-2i in prevention and management of CVD for empowering clinicians to rationalise the use of these molecules in day-to-day medical practice.



INTRODUCTION

Patients with type 2 diabetes mellitus (T2DM) are predisposed to develop atherosclerotic cardiovascular disease (ASCVD), heart failure (HF), increased cardiovascular (CV) mortality, and renal disease. The cardiac manifestations can include coronary artery disease (CAD), HF, atrial fibrillation (AF), as well as ischemic strokes as and peripheral arterial diseases. Also, diabetes increases the risk of developing albuminuria, and chronic kidney disease (CKD), both of which are independent risk factors for CVD. Thus, the combination of diabetes and cardio-renal comorbidities lead to a cumulative increase in the risk for CV events and mortality.

The first Sodium-linked glucose co-transporter-inhibitor (SGLTi) to be isolated was dihydrochalcone phlorizin, a nonselective SGLTi extracted from apple tree roots. Later, the aromatic O-glycoside sergliflozin and the aromatic C-glycoside dapagliflozin became the first selective SGLTi[1]. Currently several SGLT2i molecules like ipragliflozin, dapagliflozin, canagliflozin, empagliflozin, luseogliflozin, and tofogliflozin are available for treating T2DM, while ipragliflozin and dapagliflozin have also been approved for type 1 diabetes mellitus (T1DM) in some countries[1]. Though the cardio-vascular outcome trials (CVOTs) were conducted to demonstrate CV safety of the anti-diabetic agents, the remarkable results with Empagliflozin and Canagliflozin in the Empagliflozin CV Outcome Event Trial in T2DM Patients (EMPA-REG OUTCOME) and Canagliflozin Cardiovascular Assessment Study (CANVAS) trials demonstrating the beneficial effects of SGLT2i on CV events and HF as well as on renal outcomes revolutionised the management of heart disease in diabetes[2,3]. Soon, the CV and renal benefits of SGLT2i became apparent in patients without diabetes[4,5]. Initially marketed as an anti-diabetic agent, SGLT2i soon became a favourite medicine for cardiologists and nephrologists.

SGLT2I IN PATIENTS WITH ASCVD
Cardiovascular outcome trials of SGLT2i in T2DM

Since 2008 the United States Food and Drug Administration (FDA) required CVOTs to demonstrate CV safety for all new antidiabetic medications. These studies were primarily designed to assess whether new medications are non-inferior with respect to placebo for CV events. Generally, these trials do not assess efficacy for glycemic control, but enroll subjects with high CV risk to gather enough CV events in a short time. SGLT2 inhibitors have been evaluated in dedicated CVOTs and real-world studies for their CV safety and benefit. Given the huge benefits observed in different CV risk factors (hypertension, dyslipidemia, body weight, arterial stiffness, endothelial dysfunction), CVOTs of all 4 agents have been completed and the results are summarized in Table 1. A few meta-analyses and real-world data are also published and discussed below.

Table 1 Major cardiovascular outcome trials of sodium glucose cotransporter 2 inhibitors.

EMPA-REG outcome
CANVAS
DECLARE-TIMI 58
VERTIS-CV
SCORED
InterventionEmpagliflozin 10 and 25 mg vs placeboCanagliflozin 100 and 300 mg vs placeboDapagliflozin 10 mg vs placeboErtugliflozin 5 and 15 mg vs placeboSotagliflozin vs placebo
Populationn = 7020, T2DM with established CV diseasen = 10142 patients, T2DM with established CV disease or ≥ 2 CV risk factorsn = 17160 patients, T2DM with established CV disease or risk factors for atherosclerotic CV diseasen = 8246, T2DM with established CV disease10584 patients with T2DM and established CV disease or risk factors for atherosclerotic CV disease
Established CV disease (%)9966419948.6
Follow up period (yr)3.13.64.23.51.3
HbA1c (%) at baseline7.0% to 10.0% (for those on a stable background therapy); 7.0%-9.0% (for medication-naive patients)7.0% to 10.5%6.5% to 12.0%7.0% to 10.5%> 7%
Estimated GFR≥ 30≥ 30≥ 60≥ 3025-60
Primary outcome, HR (95%CI)3P-MACE, 0.86 (0.74-0.99)3P-MACE, 0.86 (0.75-0.97)3P-MACE, 0.93 (0.84-1.03); CV death or hospitalization for HF, 0.83 (0.73-0.95)3P-MACE, 0.97 (0.85-1.11)Total no. of deaths from cardiovascular causes, hospitalizations for HF, and urgent visits for HF 0.74 (0.63-0.88)
Key secondary outcome (s), HR (95%CI)4P-MACE, 0.89 (0.78-1.01)All-cause mortality (as below); CV death (as below); progression of albuminuria, 0.73 (0.67-0.79); CV death or hospitalization for HF 0.78 (0.67-0.91)≥ 40% decline in eGFR to < 60 mL/min/1.73 m2 or new onset end-stage renal disease or renal/CV mortality, 0.76 (0.67-0.87); all-cause mortality (as below)CV death or hospitalization for HF, 0.88 (0.75-1.03); CV death (as below); renal death or dialysis/transplant or doubling of serum creatinine from baseline, 0.81 (0.63-1.04)Total No. or hospitalizations for HF and urgent visits for HF HR: 0.67 (0.55-0.82); deaths from cardiovascular causes (as below)
Other secondary outcomes
CV death, HR (95%CI)0.62 (0.49-0.77)0.87 (0.72-1.06)0.98 (0.82-1.17)0.92 (0.77-1.11)0.90 (0.73-1.12)
All-cause mortality, HR (95%CI)0.68 (0.57-0.82)0.87 (0.74-1.01)0.93 (0.82-1.04)0.93 (0.80-1.08)0.99 (0.83-1.18)
Fatal or non-fatal myocardial Infarction, HR (95%CI)0.87 (0.70-1.09)0.89 (0.73-1.09)0.89 (0.77 − 1.01)1.04 (0.86-1.26)0.68 (0.52-0.89)
Fatal or non-fatal stroke, HR (95%CI)1.18 (0.89-1.56)0.87 (0.69-1.09)1.01 (0.84-1.21)1.06 (0.82-1.37)0.66 (0.48-0.91)
Hospitalization for HF, HR (95%CI)0.65 (0.50-0.85)0.67 (0.52-0.87)0.73 (0.61-0.88)0.70 (0.54-0.90)0.67 (0.55-0.82)

The EMPA-REG OUTCOME was designed to test the CV safety of Empagliflozin[6]. In this study, 7020 patients with T2DM and established CVD were randomized to receive either 10 or 25 mg of Empagliflozin or placebo over a median observation period of around 3 years. All the participants received the existing standard of care in terms of CV protection and received antiplatelets, lipid-lowering medications, and blockers of the renin-angiotensin-aldosterone system (RAASi). This study showed a significantly lower percentage (10.5%) of the primary outcome, 3 point - major adverse CV event [(3P-MACE), which was a composite of death from CV causes, non-fatal myocardial infarction (MI) or non-fatal stroke], in the Empagliflozin group compared to the group receiving placebo (12.1%), with a hazard ratio (HR) in the empagliflozin group: 0.86, 95%CI: 0.74-0.99; P = 0.04 for superiority. Regarding the secondary outcomes, empagliflozin treatment resulted in a 32% reduction of mortality from any cause, a 38% reduction in death from CV causes, and 35% lesser rates of hospitalization for HF, although no significant effect was observed in nonfatal MI and stroke rates. Intriguingly, the CV event curves of the empagliflozin and placebo groups started to diverge early after trial initiation. The number-needed-to-treat for the empagliflozin group was only 39, indicative of the fact that 39 patients would need treatment with empagliflozin for 3 years to prevent one case of CV mortality.

The CANVAS Program was a composite of two sub-trials: The CANVAS, designed to assess CV safety of Canagliflozin, and the CANVAS-R study (CANVAS-Renal), designed to investigate the effect of canagliflozin on albuminuria[7]. The trial recruited a total of 10142 patients with T2DM, of whom 66% were having established CV disease while 34% had multiple CV risk factors. Patients were randomized to receive Canagliflozin 100 mg, 300 mg, or placebo and the mean follow-up was 3.6 years. Like EMPA-REG OUTCOME, participants were patients treated with routine CV protective regimens using statins, antiplatelets, and RAASi. The primary outcome was significantly lower in the canagliflozin group compared to placebo (26.9 vs 31.5 participants per 1000 patient-years; HR: 0.86; 95%CI: 0.75-0.97; P < 0.001 for noninferiority; P = 0.02 for superiority) Additionally, canagliflozin was found to reduce the rate of hospitalization due to HF by 33%. However, there was no significant effect was on all-cause mortality or CV mortality.

The largest CVOT done with dapagliflozin was the ‘Dapagliflozin Effect on CV Events-Thrombolysis in MI 58’ (DECLARE-TIMI 58) study[8]. A total of 17160 patients with T2DM and established ASCVD (41%) or multiple risk factors for ASCVD (59%) were randomized to receive either dapagliflozin 10 mg or placebo for a median period of 4.2 years. Among two primary outcomes, dapagliflozin was seen to reduce the composite outcome of CV death or hospitalization for HF by 17%, but no beneficial effects were seen in terms of 3P-MACE. Hospitalization due to HF was reduced by 37% and that was the driving factor behind meeting the primary outcome, but no effect was seen in CV death or all-cause mortality. In terms of secondary endpoints, dapagliflozin reduced the composite renal outcome by 24% [≥ 40% decrease in estimated glomerular filtration rate (eGFR) to < 60 mL/min/1.73 m2, new end-stage renal disease, or death from renal or CV causes], but did not affect the all-cause mortality.

Ertugliflozin was studied in the VERTIS CV Trial, where a total of 8246 patients underwent randomization for ertugliflozin 5 mg, 15 mg, and placebo and were followed for a mean of 3.5 years[9]. Patients treated with Ertugliflozin showed noninferiority for 3P-MACE as compared with placebo (HR: 0.97; 95%CI: 0.85-1.11; P < 0.001 for noninferiority). Hospitalization due to heat failure was reduced by 12%, but no benefit was observed in the reduction of CV death, all-cause mortality, or renal outcomes. The basic characteristics of CVOTs are depicted in Table 1.

Sotagliflozin CV outcome trial, SCORED[10], was published recently. It included 10584 patients with established CVD (48.6%) and with multiple risk factors (51.4%) and was randomized between sotagliflozin and placebo and followed up for 1.3 years. Patients treated with sotagliflozin demonstrated a 26% reduction in primary outcome (total number of deaths from CV causes, hospitalizations for HF, and urgent visits for HF) HR: 0.74; 95%CI: 0.63-0.88; P < 0.001. There was a 33% reduction in HF, but no benefit was observed for the reduction of CV death. Genital mycotic infections, diarrhea, volume depletion, and diabetic ketoacidosis were more common with sotagliflozin than with a placebo.

SGLT2I AND HEART FAILURE

The use of SGLT2i in HF has drastically changed the therapeutic outcomes of these patients. Initially introduced as an agent for glycemic control, SGLT2i gained recognition in the management of HF after the EMPA-REG OUTCOME trial showed a significant reduction in hospitalisation due to HF (HHF) in the empagliflozin group compared to the placebo[2]. This trial and those that were performed in the later years suggested that independent of its glucose-lowering effect, SGLT2i must have a direct effect on HF. The current classification of HF is based on the ejection fraction (EF), with HF with reduced EF (HFrEF) defined as EF ≤ 40%, HF with preserved EF (HFpEF) defined as EF > 50%, and HF with mildly reduced EF (HFmrEF) defined as EF between 40% to 50%[11].

SGLT2i in HFrEF

The DAPA-HF trial recruited patients having HF with an EF ≤ 40% and an eGFR ≥ 30 mL/min per 1.73 m2. Dapagliflozin was found to reduce the primary composite CV outcomes (which included death from CV causes, hospitalization for HF, or an urgent hospital visit resulting in intravenous therapy for HF) by 24%[4]. The EMPEROR-Reduced trial included patients with HF with a mean EF of 27% and an eGFR ≥ 20 mL/min per 1.73 m2. There was 22% reduction in the primary composite outcome of CV death or hospitalization for HF in the group receiving empagliflozin[12].

SGLT2i in HFpEF or HFmrEF

In the earlier studies, the beneficial effect of the SGLT2i was demonstrated only in patients with HFrEF, but the EMPEROR-Preserved trial in 2022 showed that empagliflozin improved CV outcomes even in patients with HFpEF[13]. The EMPEROR-Preserved trial was the first trial of SGLT2i which included patients with HFmrEF and HFpEF regardless of whether they had diabetes or not. It was found that with the use of empagliflozin, there was a 19% reduction in the primary composite outcome of CV death and HHF[13]. The recently published DELIVER trial also enrolled patients with HFmrEF and HFpEF. There was an 18% reduction in the primary composite endpoint of worsening HF or CV death[14].

Although the newest addition to the HF therapies, SGLT2i helps in a significant reduction of morbidity and mortality in the entire range of EF. Thus, they form an important pillar in the management of HF. The details of the landmark trials of SGLT2i in HFrEF or HFpEF are outlined in Table 2.

Table 2 Major heart failure trials with sodium glucose cotransporter 2 inhibitors.
Trial and medication namePrimary endpointMedian follow-upOutcomes
HFrEF
DAPA-HF (dapagliflozin)Primary composite outcome: Worsening heart failure (hospitalization or an urgent visit resulting in intravenous therapy for HF) + cardiovascular death18 monthsReduction in the primary composite outcome by 24%
EMPEROR-reduced (empagliflozin)Primary composite outcome: Hospitalisation for heart failure + cardiovascular death16 monthsReduction in the primary composite outcome by 22%
HFpEF
EMPEROR-preserved (empagliflozin)Primary composite outcome: Hospitalisation for heart failure + cardiovascular death26 months19% reduction in the primary composite outcome
DELIVER (dapagliflozin)Primary composite outcome: Worsening heart failure (hospitalization or an urgent visit resulting in intravenous therapy for HF) + cardiovascular death28 months18% reduction in the primary composite endpoint
SGLT2i in acute heart failure

In a meta-analysis of RCTs involving 1831 patients with acute HF (AHF), SGLT2i improved the Kansas City Cardiomyopathy Questionnaire scores (mean difference: 4.12; 95%CI: 0.1.89-6.53) and reduced the risk of rehospitalization due to HF (OR: 0.52; 95%CI: 0.42-0.65). However, no significant effect on all-cause mortality was observed. Initiating SGLT2i in patients with AHF did not increase the risk of hypotension or acute kidney injury (AKI)[15].

While SGLT2i use reduces levels of plasma NT-proBNP and improves diastolic function of the heart, improvement in left-ventricular EF was observed only in patients having HFrEF who are in stage C HF. The benefits were not very prominent in patients with HFpEF with HF stages A or B.

SGLT2I AND CV EFFECTS IN T2DM - REAL-WORLD DATA

Real-world data, which possibly better represent everyday clinical practice, do exist in favor of SGLT2i for CV outcome. An observational study from Denmark, Norway, and Sweden by Birkeland et al[16] included a total of 91320 patients, among which 22830 patients with T2DM were on SGLT2i (most commonly Dapagliflozin) and a total of 68490 patients were being treated with other anti-diabetic agents[15,16]. They were observed for a follow-up of around 0.9 years. One-fourth of them already had established CV disease. It was seen that SGLT2i use was associated with a significantly reduced risk of major adverse CV events, CV mortality, and HF-related hospitalisation in comparison to other glucose-lowering drugs. However, the difference in nonfatal MI or stroke was not significant.

SGLT2I IN THE PRIMARY PREVENTION OF CV DISEASES

Though the DECLARE-TIMI 58 and other trials suggested that SGLT2i can reduce the CV composite outcome in patients without established ASCVD, a meta-analysis including data from three major CVOTs with 34322 patients, 39.8% of whom did not have established ASCVD, SGLT2i was found to reduce MACE by 11% (HR: 0.89, 95%CI: 0.83-0.96, P = 0.001][17]. However, on subgroup analysis, the benefits were only seen in patients who had established ASCVD [0.86 (0.80-0.93)] but not in those without [1.00 (0.87-1.16), P for interaction = 0.05][16]. The reduction in the composite of CV death or HHF by 23% [0.77 (0.71-0.84), P < 0.0001] could, however, be in patients with and without ASCVD and with and without a history of HF. The renal benefits were also seen in both the groups with and without ASCVD. The magnitude of the benefit of SGLT2i differed according to the baseline renal function, with a greater decline in hospitalisations for HF (P = 0.007) and a lesser reduction in the progression of CKD (P = 0.03) seen in patients with more severe kidney disease at baseline.

CARDIOVASCULAR BENEFIT IN NON-DIABETIC INDIVIDUALS

Some of the SGLT2 trials evaluating kidney and HF outcomes have deliberately enrolled patients without T2DM, but none of these studies were powered to study their effects on atherosclerotic outcomes. Specially, MACE was not included as an outcome in the primary hierarchy of analyses in any of the HF trials like Empagliflozin Outcome Trial in Patients with Chronic HFrEF (EMPEROR Reduced)[17], Empagliflozin Outcome Trial in Patients with Chronic HFpEF (EMPEROR-Preserved)[18], and Dapagliflozin and Prevention of Adverse Outcomes in HF (DAPA HF)[19]. Although in the Dapagliflozin and Prevention of Adverse Outcomes in CKD (DAPA CKD). 3P-MACE (CV death, MI, and stroke) was included as a prespecified, exploratory outcome, but the comparative efficacy of dapagliflozin for this outcome were not presented by baseline T2DM status. Additionally, MACE was seen to occur in only 3% of the trial cohort, meaning that the power was likely insufficient for meaningful MACE analyses classified by T2DM status[20]. Further studies evaluating 3P-MACE in individuals without diabetes may answer this question in the future.

EFFECTS OF SGLT2I ON DIFFERENT CV RISK FACTORS

Trials with various SGLT2i have consistently shown marked benefits in various CV outcomes. This indicates that there might be a class effect of SGLT2i on CV parameters. This benefit cannot be solely attributed to their glucose-lowering effect as significant improvements in different CV outcomes have been found even in patients without diabetes. Instead, the CV benefit is because of the effect of SGLT2i on the various risk factors associated with heart disease most importantly blood pressure control, weight loss, and dyslipidemia. We will now briefly discuss the major CV risk factors and how they are ameliorated by SGLT2i.

Effects on glucose levels

The first indication of SGLT2i after they were designed was to control blood glucose. By inhibiting the SGLT2 co-transporters in the proximal convoluted tubules, they lower the blood glucose and have been found to reduce the HbA1c by around 0.5% in various trials[21]. This would reduce the glucotoxicity and oxidative stress on the cardiac tissues. However, the rapid efficacy observed with the SGLT2i on cardiac endpoints, starting days after the initiation of the drug, suggests other mechanisms playing a role in this cardio-protection.

Effects on body weight

With the use of SGLT2i, there is a loss of glucose in the urine leading to the loss of calories which, in turn, results in the mobilisation of the fatty acids from the adipose tissue stores resulting in weight loss. This has been consistently observed in various trials and metanalyses suggest that the weight reduction is around 2-3 kg[22]. In DAPA-HF, EMPEROR-Reduced, and EMPEROR-Preserved trials, weight reduction of 0.9 kg, 0.8 kg, and 1.3 kg were observed respectively as compared to the placebo[4,12,13]. Again, this modest weight reduction cannot entirely explain the CV benefit provided by the SGLT2i.

Effects on blood pressure

Hypertension is a very strong adverse risk factor for both HFrEF and HFpEF. Because of the osmotic and diuretic effects of the SGLT2i, there is a modest reduction in blood pressure. In the DAPA-HF trial, there was a mean difference of around -1.4 mmHg in the systolic BP after 8 months which was significant when compared to the placebo. In the EMPEROR-Reduced trial, compared to the placebo, empagliflozin showed a greater reduction (-2.4 vs -1.7 mmHg) but it was not significant[4,12]. These modest reductions, although not entirely, will affect the cardiac and vascular remodelling and afterload, leading to improvement in hemodynamics.

Effect on lipid parameters

Although there is a debate regarding the effect of SGLT2i on low-density lipoprotein (LDL) cholesterol, most of the studies showed that there is a minor increase in LDL cholesterol with its use. A study published in 2013 showed an increase in LDL cholesterol by 11.7% with the use of canagliflozin for 52 wk in patients with T2DM[23]. Still SGLT2i has a cardioprotective effect and this paradox can be explained by the fact that SGLT2i might decrease the small dense LDL and increase the large buoyant LDL as was seen with the use of dapagliflozin for 12 wk in T2DM[24]. SGLT2i also resulted in an increase of high-density lipoprotein cholesterol by around 10%-11% with the use of canagliflozin in one study[25]. Moreover, SGLT2i also decreases lipid deposition in the visceral fat, decreases lipid oxidation, and affect the transport of lipid into the cells[26]. All these taken together would provide a cardioprotective benefit with the use of SGLT2i.

Effects on albuminuria and progression of CKD

CV events are the chief cause of mortality in patients with CKD and the risk progressively increases with a decline in eGFR or increasing degrees of albuminuria, making the latter an independent predictor of CV risk[27]. In addition to their glycemic lowering properties and effects on body weight and systemic blood pressure, SGLT2i can reduce intraglomerular pressure, and therefore albuminuria and also slow down GFR decline[28]. Recent data also suggest that SGLT2i can directly reduce oxidative stress, and angiotensinogen levels as well as reduce NLRP3 inflammasome activity in the kidney[29]. The promising results of Canagliflozin in the CREDENCE trial led to it being stopped early and showed a significant reduction in the primary composite end point of ESRD, doubling of serum creatinine, or renal or CV death with up to 32% risk reduction for development of ESRD[28]. It also demonstrated clear benefits on CV outcomes in the advanced CKD group[3]. The DAPA-CKD study with Dapagliflozin was similar except that one-third of the population had other one-third had CKD without T2DM and the endpoints were slightly different. The HR for the renal composite outcome of a sustained decline in eGFR of > 50%, ESRD, or death from renal causes was 0.56 (95%CI: 0.45-0.68; P < 0.001)[5].

Effects on uric acid levels in serum

Several studies and meta-analyses have verified the effectiveness of SGLT2i in improving hyperuricemia. The uricosuric effects of dapagliflozin, empagliflozin, and canagliflozin have been seen in patients with or without diabetes[30,31]. The likely mechanism involves the GLUT9 isoform 2. By preventing reabsorption, SGLT2i can increase the concentration of glucose reaching the collecting ducts, which in turn compete with urate for reabsorption via the GLUT9 isoform 2 leading to the excretion of more uric acid. Other mechanisms involving activation of the xanthine oxidase by sirtuin activation and alteration of URAT1 transporter have been also proposed[32]. In a nationwide study from Taiwan, that investigated the association, the use of SGLT2i was associated with a lower incidence of gout (HR: 0.89; 95%CI: 0.82-0.96) than DPP4 inhibitors, and this was particularly seen in patients receiving dapagliflozin[33]. Up to 15% reduction in the risk of gout was observed with SGLT2i. Another meta-analysis of 62 studies, including 34941 patients, however, reported that although all the SGLT2 inhibitors (empagliflozin, canagliflozin, dapagliflozin, tofogliflozin, luseogliflozin, or ipragliflozin) significantly decreased uric acid levels, empagliflozin resulted in the most superior reduction[30]. No consistent dose-related effects were observed.

Effects on haematocrit

All SGLT2i has been associated with a modest increase in haematocrit between 2%-4%. This effect was seen with empagliflozin in patients with T2DM and stage 2 or 3 CKD, but not stage 4 CKD. Though initially thought to be due to its diuretic effects, the urine volume returns to baseline within 1 wk of SGLT2i, whereas the increase in haematocrit continues beyond 2 months. In patients with diabetes mellitus, increased glucose uptake by SGLT2 in the proximal tubular epithelial cells results in increased ATP consumption by the Na+/K+ pump. There is increased oxygen consumption by the proximal tubular mitochondria to meet the high demand for ATP, resulting in a relative hypoxia within the renal cortical cells[34]. Selective damage to the proximal tubular epithelial cells as seen in diabetic kidney disease induces trans-differentiation of the erythropoietin-producing fibroblasts into myofibroblasts[35], which loses the capacity to produce erythropoietin and starts producing fibrogenic molecules. Low erythropoietin levels have been demonstrated even in patients with T2DM with normal kidney function[36]. This might be due to metabolic stress associated with excessive glucose resorption by the tubular epithelial cells causing a hypoxic microenvironment. SGLT2i can reduce this metabolic stress and reduce ATP consumption by the Na+/K+ pump, causing a possible reversion of myofibroblasts to erythropoietin-producing fibroblasts and elevation of the haematocrit. Additionally, the nephroprotective effect of SGLT2i which prevents progression of CKD can also improve erythropoietin levels.

Effects on inflammatory markers

Inflammation is a key component in the development of atherosclerosis and plaque destabilisation/rupture. Indeed, SGLT2 inhibitors have all been shown to reduce inflammation in Apo E -/- mice[37]. Reduced IL-1β, IL-6 and IL-10 levels have been seen with empagliflozin, while dapagliflozin has demonstrated reduced NLRP3, IL-1β and IL-18 levels[38]. Canagliflozin has demonstrated significant reduction in the adhesion molecules, VCAM-1 and MCP-1, while increasing the TIMP-1 inhibitor[37,38]. These anti-inflammatory and vaso-protective effects might explain some of the major mechanisms involved in the CV and nephroprotective benefits of sodium glucose cotransporter-2 inhibitors (SGLT-2i) molecules.

Effects on metabolic syndrome-associated fatty liver disease

The effects of SGLT2i on body weight and their antioxidant and anti-inflammatory effects make them promising candidates for the management of MAFLD. In addition to decreases in in insulin and glucose levels in T2DM, SGLT2i can lead to reduction in the de-novo lipid synthesis in the liver[39]. Also, the glucagon-secreting alpha cells express SGLT2, and inhibition of this reduces intracellular glucose concentration in them thus increasing the secretion of glucagon[40]. The high glucagon levels and elevated glucagon/insulin ratio can stimulate β-oxidation leading to a shift from carbohydrate to fatty acid metabolism and reduction in the hepatic triglyceride content[41]. Thus, SGLT2i can play an important role in reducing hepatic lipid content by reduction in de novo lipid synthesis due to reduced blood glucose and insulin levels along with increased beta-oxidation of fatty acids.

A study comparing ipragliflozin to Pioglitazone found that while pioglitazone demonstrated benefit in terms of reduction in serum ALT and HbA1c; reductions in body weight and visceral fat were seen in those with Ipragliflozin[42]. Canagliflozin has been found to significantly reduce FIB-4 index and ferritin levels in T2DM patients with MAFLD, suggesting improvement in hepatic fibrosis[43]. Dapagliflozin has also been shown to reduce Fibroblast Growth Factor 21 levels and indices of hepatocyte injury[44]. A study using serial liver biopsies showed Canagliflozin use for 24 wk showed remarkable histologic improvement of metabolic-associated steatohepatitis (MASH); with even demonstration from MASH to borderline or non-MASH status[45].

Effects on obstructive sleep apnea

Obstructive sleep apnea (OSA) is related to CVD development and has been identified as a modifiable CV risk factor[46]. SGLT2i have been found to reduce apnea-hypopnea index in patients with T2DM with OSAS in small studies, though their beneficial effects on OSA or sleep-disordered breathing have not been substantiated by other studies or meta-analyses[47-49]. Apart from weight reduction, other postulated beneficial mechanisms could include rostral nasal fluid shift due to diuresis and reduction of circadian sympathetic nerve activity, nocturnal hypertension, and oxidative stress by which SGLT2i might reduce the incidence or the CV effects of OSA[50,51]. Further studies are required to elucidate the benefits of SGLT2i in this regard. The CV benefits of SGLT2i are depicted in Figure 1.

Figure 1
Figure 1 Cardiovascular benefits of sodium glucose cotransporter 2 inhibitors - the evidence from trials. CV: Cardiovascular; HHF: Hospitalisation due to heart failure; 3P-MACE: 3 point - major adverse cardiovascular event; HFrEF: Heart failure with reduced ejection fraction; HFpEF: Heart failure with preserved ejection fraction; HFmrEF: Heart failure with mildly reduced ejection fraction; AHF: Acute heart failure; OSA: Obstructive sleep apnea; NAFLD: Non-alcoholic fatty liver disease; eGFR: Estimated glomerular filtration rate; BP: Blood pressure.
CARDIOVASCULAR BENEFITS WITH SGLT2I - POSSIBLE MECHANISMS

The mechanisms that drive the CV benefits of SGLT2i can be grossly categorised into hemodynamic alterations, metabolic changes, and direct effects on the cardiomyocytes. The CV benefits with Empagliflozin were seen as early as 12 wk after randomization, when the patients treated with empagliflozin were found to have lower rates of HHF (0% vs 2.9%), of the composite of HHF/CV deaths (0.2% vs 4.1%), and of the composite of HHF or all-cause mortality (0.2% vs 4.1%)[52]. A posthoc analysis of the EMPAREG-OUTCOME trial showed that the reduction in risk for empagliflozin vs placebo reached a significance at day 17 for HHF, day 27 for the composite of CV death/HHF, and day 59 for CV deaths[53]. The direct effects on the cardiomyocytes have been seen in several in-vitro studies and animal models, but the time taken for these effects to manifest as beneficial effects on the functioning of the human heart is not clear.

Given that the cardioprotective effects of glycemic or weight reduction and other metabolic effects would take more time to manifest, the hemodynamic alterations and effects on the cardiomyocyte actions with SGLT2i may have a more important role behind the early benefits. However, in the long run, the metabolic effects become equally important as the benefits are sustained throughout use[53]. The effects of SGLT2i on different hemodynamic and metabolic risk factors that drive ASCVD or HF are discussed in an earlier section. In the ensuing part, we have discussed the direct effects of SGLT2i on the structure and functioning of the myocardium and blood vessels.

SGLT2I - DIRECT EFFECTS ON MYOCARDIUM AND BLOOD VESSELS

Studies with SGLT2i suggest that the effects of SGLT-2i may not cause significant changes in cardiac stroke volume or output in patients with or without established CV disease. However, diastolic function appears to be directly influenced by SGLT-2i and significant decreases in left ventricular mass have been documented following treatment with SGLT2i[54]. Both of these are associated with endothelial dysfunction, and it is possible that SGLT-2i, by improving endothelial dysfunction, inhibits negative cardiac remodelling and improves diastolic function. The receptors and mediators through which SGLT2i exert their direct effects on the cardiomyocytes remain to be fully elucidated. Cardiomyocytes have been found to express SGLT1, which may be one of the targets for SGLT2i[54]. However, the effects of SGLT2i on HF may not only be mediated by their target receptors. SGLT2i have been found to directly inhibit Na+/H+ exchanger-1 (NHE1) in cardiomyocytes. Also, dapagliflozin has been found to directly activate AMP-activated protein kinase (AMPK) leading to reduced lipopolysaccharide-induced myocardial fibrosis[55]. Other cardiac sodium channels like Nav1.5 have also been found to be the targets for SGLT2i and inhibiting these sodium channels can ameliorate dysfunctional calcium overload[56].

Effects on myocardial fuel energetics

Under physiologic circumstances, cardiomyocytes prefer fatty acids as the predominant metabolic fuel for energy generation which account for 70%-90% of ATP synthesis. Although fatty acid metabolism produces more ATP than glucose, complete oxidation of the former also requires more oxygen. In those with diabetes mellitus, due to lesser glucose uptake in cardiomyocytes, they utilise more fatty acids and less glucose as the preferred substrate for oxidative metabolism, leading to greater oxygen consumption and decreased pumping efficiency of the heart[57,58]. SGLT2i has been found to benefit myocardial energy metabolism by increased GLUT1 expression and therefore increased glucose uptake in the human and murine myocardium. Enhanced rates of glycolysis and glucose oxidation have also been demonstrated in the myocardium of db/db mice[59,60]. In mouse models with diabetes, it has been demonstrated that there is increased expression of the O-palmitoyl transferase (CPT) isoform CPT1b on the outer mitochondrial membrane, which facilitates mitochondrial transport and β-oxidation of fatty acids in cardiomyocytes. Empagliflozin was found to reduce mRNA and protein expression of CPT1b[61]. Additionally, empagliflozin has been found to inhibit the mRNA and protein expression of CD36, which serves a downstream mediator of PPAR-γ in cardiomyocytes[62,63]. Following activation of PPAR-α, fatty acid uptake is enhanced compared to glucose. Thus, this effect of SGLT2i might reduce the uptake and accumulation of fatty acids within the myocardium.

One meta-analysis of RCTs showed that SGLT2i can increase adiponectin levels in T2DM[61]. Adiponectin has a negative correlation with serum triglycerides and higher adiponectin levels lead to enhanced utilization of glucose and fatty acids by muscle tissue. SGLT2i increases myocardial utilization of ketone bodies to increase ATP production. Although empagliflozin has not been seen to directly improve the efficiency of myocardial ketone body utilization, empagliflozin can increase levels of ketone bodies in serum, predominantly by promoting expression of the enzyme HMG CoA Synthase which is necessary for ketone body production[64]. The “thrifty” or frugal fuel hypothesis suggests that ketonemia and ketone body utilisation by cardiomyocytes can increase the efficiency of cardiac mitochondrial oxidation[65].

Effects on myocardial mitochondria

Cardiac mitochondrial dysfunction is a factor behind diabetic cardiomyopathy. Under hyperglycemic conditions, mitochondria within cardiomyocytes undergo dynamin-related protein 1 (Drp1)-mediated fission, ultimately leading to fragmentation, ROS production, and increased oxidative stress. Dapagliflozin significantly reduced myocardial mitochondrial Drp1 level thereby reversing this impairment[66]. Similarly, SGLT2i have been found to normalise the alteration in proteins like MFN1, MFN2 and OPA1 which are responsible for mitochondrial fusion[66]. Additionally, empagliflozin has been seen to reverse the downregulation of PGC-1α, NRF-1, and mtTFA in rat models of T2DM[67]. These allow increased transcription and replication of mitochondrial DNA and activation of mitochondrial electron transport chain (ETC). Hyperglycemia increases O-GlcNAcylation, which in turn leads to decreased activity of ETC complexes I, III, and IV. Dapagliflozin and other SGLT2i can directly reduce O-GlcNAc transferase activity, leading to improvement in the functioning of the mitochondrial respiratory chain[68].

Effects on endothelial cells

Empagliflozin leads to activation of AMPK and inhibition of Drp1 by serine phosphorylation, leading to anti-inflammatory effects on arterial endothelial cells[69]. Dapagliflozin activates voltage-dependent K+ channels, also known as the Kv channels, which are responsible for maintaining the membrane resting potential and vascular tone. Opening of these channels will lead to hyperpolarization and endothelial-independent vascular smooth muscle relaxation and vasodilation. SGLT2i have been found to inhibit TNFα-induced ROS generation and therefore reduced NO consumption in coronary arterial endothelial cells. Reduced serum uric acid concentrations with SGLT2i can also lead to increased NO synthase activity and improved NO synthesis[55]. Improved flow-mediated dilation has been seen with dapagliflozin, which might be dependent on COX-2 inhibition and reduction in ROS production[70]. SGLT2i have been found to inhibit COX-2 mRNA expression[71].

Effects on ventricular compliance, myocardial fibrosis, and infarct size

Chronic hyperglycaemia increases formation of AGEs by nonenzymatic glycation of proteins. AGEs activate the receptor for AGE (RAGE) leading to proliferation, function, and migration of cardiac fibroblasts, ultimately ending in myocardial fibrosis and cardiac aging. Empagliflozin has been shown to inhibit the AGE/RAGE axis in the kidney, though this action has still now not been demonstrated in the heart[72]. SGLT2i acts directly on cardiomyocyte NHE1 to reduce cytosolic Na+ and thereafter Ca2+ accumulation within the cardiomyocyte[69].

People living with diabetes have larger size of myocardial infarcts than non-diabetics. SGLT2i can promote angiogenesis by reducing the loss of CD31+ micro-vessels, leading to reduction in the size of perfusion defects in diabetes model mice[73]. SGLT2i improves ventricular remodelling and reduces myocardial fibrosis by modulating macrophage polarization in the cardiomyocytes and reducing myocardial expression of collagen I and collagen III proteins as also pro-fibrotic molecules like TGF-β1, p-Smad2, and p-Smad3[71].

Empagliflozin has been found to improve ventricular remodelling in diabetic patients. Empagliflozin increases NO and cGMP concentrations as well as sGC and PKGIα activity leading to a decrease in cardiomyocyte stiffness[74]. Empagliflozin has been shown to reduce left ventricular mass in patients with T2DM and improve left ventricular hypertrophy[75].

Effects on oxidative stress

Canagliflozin and empagliflozin have been found to activate and restore eNOS activity in the myocardium, while the former also reduced iNOS levels, which in turn decreased superoxide and nitrate[76]. In animal T2DM models, SGLT2i has been shown to increase lipid hydroperoxide and MDA levels significantly compared to the control group, while reducing levels of glutathione peroxidase (GSH-Px) and superoxide dismutase (SOD). Empagliflozin has the potential to reverse the imbalance between pro-oxidant molecules like lipid hydroperoxide and MDA levels and anti-oxidant molecules like GSH-Px and SOD in T2DM models, possibly by activation of the Nrf2/HO-1 pathway[77]. SGLT2i also activates Silencing information regulator 2 related enzyme 1 (SIRT1) and corresponding downstream pathways, which might explain how SGLT2i can decrease oxidative stress in diabetic cardiomyopathy via the SIRT1/Nrf2 signalling pathway in T1DM or the Sirt1/fork head box[69].

Protective effects against myocardial apoptosis

Diabetes is known to promote programmed cell death of cardiomyocytes. The SGLT2i inhibits caspase-3 activity in the myocardium, as also inhibits the ERK1/2 pathway and promotes the STAT3 pathway, ultimately leading to decreased cardiomyocyte apoptosis[78]. Dapagliflozin has been seen to reverse the increased NLRP3, ASC, IL-1β, or caspase-1 in mice models with T2DM, which reflects increased NLRP3-inflammasome complex formation and risk for pyroptosis (a highly inflammatory type of programmed cell death)[79].

Empagliflozin has been reported to inhibit enhanced crease autophagy of cardiomyocytes by downregulation of NHE1 and NHE1-related genes like Beclin 1 which induce autophagy[80,81]. ER stress leads to the accumulation of misfolded or unfolded proteins thus initiating an unfolded protein response (UPR) that leads to apoptosis of cells. In ER stress models induced by pressure overload or ischemic injury, SGLT2i, via SIRT1 activation and GRP78 reduction can inhibit the increase in p-PERK and its downstream molecules which are associated with ER stress[82,83].

OTHER MECHANISMS OF CARDIOVASCULAR BENEFIT WITH SGLT2I
Effects on the intestinal microbiota

Dapagliflozin has shown favourable alteration in the gut microbiota, including an increased abundance of Akkermansia muciniphila[84]. This has been associated with improved glycemic profile and improved generalized vascular functioning in mice with T2DM. Induction of the expression of tight junctions in the gut also reduces endotoxemia-related inflammation and prevents atherosclerosis. Luseogliflozin was found to increase the abundance of other bacteria like Syntrophothermus lipocalidus, Parabacteroidesdistasonis distasonis, and Anaerotignum sp, which produce short-chain fatty acids (SCFAs) which leads to improvement in diabetes and CV function[85]. However, these changes in gut microbiota have not been confirmed in patients with T2DM. The possible mechanisms of CV benefits from SGLT2i are summarised in Figure 2.

Figure 2
Figure 2 Mechanisms of cardiovascular benefits of sodium glucose cotransporter 2 inhibitors. CV: Cardiovascular; SGLT2i: Sodium glucose cotransporter-2 inhibitors.
Effects on sympathetic nervous system activity

Since sympathetic nervous system hyperactivity is intricately linked with the initiation, progression, and deterioration (poor prognosis) of chronic human HFrEF, in a mechanism akin to β-blockers, SGLT2i, by suppressing sympathetic neural activity can protect the failing myocardium against adrenergic overstimulation. In the EMPA-REG OUTCOME Trial, empagliflozin was found to reduce heart rate somewhat unexpectedly. Consistent with this, Luseogliflozin was also found to cause bradycardia in patients with baseline elevated heart rate. Studies have shown that the FFAR3 (GPR41) receptors are abundant on sympathetic ganglia and nerve endings; and while SCFAs, via their stimulation of FFAR3 leads to activation of sympathetic neuronal firing, the ketone body 3-hydroxybutyrate can block it leading to reduced norepinephrine release from sympathetic nerve terminals. The SGLT2i, by their potential to increase production of ketone bodies in humans, could potentially exert sympatholysis by this mechanism[86].

Additionally, treatment of HFD-fed mice with dapagliflozin has demonstrated diminished tyrosine hydroxylase activity in the medulla, primarily by inhibition of G-protein coupled receptor kinase 2 (GRK2) leading to reduced noradrenaline levels. GRK2 has inhibitory effects on the α2-adrenergic receptor (α2-AR) which mediates the feedback mediated reduction in catecholamine release from sympathetic nerve terminals. Downregulated GRK2 by Dapagliflozin can thus lead to increased α2-AR mediated feed-back and an overall reduced catecholamine release from the nerve terminals[87].

Notably, while SGLT2 inhibition can affect sympathoinhibition in some critical target organs, such as the heart and the kidneys, dapagliflozin has been shown to promote sympatho-excitation in white adipose tissue. Increased mRNA levels of the brown adipose tissue-selective gene Ucp1 and its upstream mediator, Pgc-1 has been demonstrated suggestive of “beiging” effect of dapagliflozin[87]. Thus, available evidence suggests SGLT2i can potentially reduce secretion of catecholamines and their effects on the myocardium but promotes sympathetic overactivity of white adipose tissue.

ROLE OF SGLT2I IN ATRIAL FLUTTER/FIBRILLATION

The presence of diabetes mellitus independently predicts the risk for AF[2], and diabetes is part of the CHA2DS2-VASc score used to predict stroke risk in patients with AF. HF increases the risk of arrhythmias including atrial flutter and/or fibrillation and the presence of AF is associated with adverse outcomes in patients with HFrEF and HFpEF. In the DECLARE-TIMI 58 trial, dapagliflozin was found to reduce the relative risk of AF by 19% (HR: 0.81, 95%CI: 0.68-0.95)[8]. Also, empagliflozin has demonstrated a greater absolute benefit on renal and HF-related events in individuals with a history of AF (HR: 0.58, 95%CI: 0.36-0.92) and without AF (HR: 0.67, 95%CI: 0.55-0.82, Pinteraction = 0.56)[88]. The reduction in AF events was seen regardless of the presence of HF, and ASCVD. Other CVOTs of patients with diabetes mellitus also reported lower rates of AF with SGLT2i inhibitor, though the absolute reduction was small ranging between 0.1% to 0.2% per year. However, no consistent reduction in stroke was found. Factors that may contribute to a reduction in atrial tachyarrhythmias could be reduced rates of HF and atrial stretch, reduction in blood pressure and improvements in cardiomyocyte energetics, and arterial compliance. There needs to be additional studies to confirm the reliability and clinical importance of this finding.

ROLE OF SGLT2I IN CARDIAC AUTONOMIC NEUROPATHY

Patients with long-standing diabetes can have cardiac autonomic neuropathy (CAN), in which sympathetic tone predominates over parasympathetic activity. This significantly increases CV morbidity and mortality with a high risk for sudden cardiac death. Unfortunately, to date, there is no definitive treatment for CAN. SGLT2i, by its property to reduce sympathetic nervous system activity, offers hope in the management of CAN. Small studies have demonstrated that SGLT2i can reduce the risk of recurrence of vasovagal syncope, which is related to altered autonomic system function, as evaluated by heart rate variability (HRV), and by 123I-metaiodobenzylguanidine myocardial scintigraphy indexes and also improve HRV and heart rate turbulence parameters while decreasing the frequency of ventricular premature beats[89]. However, in a preliminary analysis of data from the EMPA-HEART CardioLink-6 trial, Holter monitoring analyses and automated algorithms to determine HRV domain measures over 6 months found that the observed cardiac benefits of empagliflozin were not likely associated with modulation of autonomic tone in patients with T2DM and stable CAD[90].

CAUTION AND CONTRAINDICATIONS TO THE USE OF SGLT2I IN HEART DISEASE

SGLT2i have been associated with different adverse effects. The glucosuric effect of SGLT2i leads to higher urine glucose levels and thus predisposes patients to urinary tract and genital infections. The data regarding urinary tract infection is conflicting among different studies, but most studies report neutral findings. In the large three CVOT outcome trials, the rates of UTI were not significantly increased when compared with placebo[6-8]. In 2013, one meta-analysis reported a higher incidence of UTI between SGLT2i and either placebo or active competitors[91]. But two subsequent metanalysis refuted this finding[92,93]. Available real-world data highlight the fact that SGLT2i do not increase the frequency of UTIs compared to either DPP 4 inhibitors or GLP1 receptor agonists[94,95]. However, genital mycotic infections have been consistently found to be more frequent in patients on SGLT2i in all three major CVOT outcome trials[6-8] as well as in different meta-analyses[92,93]. Risk factors for genital mycotic infection include female gender and previous history of genital infection. However, the genital infection tends to be non-severe and manageable with systemic or topical antifungal agents without the need for treatment discontinuation[96].

SGLT2i have an osmotic diuretic effect, thus a mild volume depletion state can be observed with them. A slight reduction in blood pressure, orthostatic hypotension, and dizziness can occur with these agents especially when combined with diuretics. A meta-analysis did not find a higher volume depletion with SGLT2i compared to placebo[97]. Still, it is recommended to review the dose of diuretics while initiating a patient with SGLT2i to prevent postural hypotension.

The use of SGLT2i is associated with increased circulating ketone bodies. The incidence of euglycemic diabetic ketoacidosis varies from study to study. CANVAS and EMPAREG trials reported a nonsignificant increase of euglycemic DKA, but DECLARE-TIMI 58 reported a small but significant increase of the same[6-8]. Two meta-analyses reported a nonsignificant increase in the rates of DKA among SGLT2i versus placebo or other anti-diabetic agents[98,99]. Real-world data however suggests slightly higher rates of DKA among SGLT2i users[100,101]. This difference of results might be due to the controlled conditions of RCTs and the cautious selection of participants. Risk factors for euglycemic DKA included T1DM, presence of dehydration, excess alcohol intake, critical illness, post-operative period and intake of very low-carbohydrate diet.

The FDA had issued a warning for AKI with dapagliflozin and canagliflozin in 2016 based on few case reports submitted to the FDA adverse events reporting system (FARES). The possible mechanisms that mediate renal damage by SGLT2i could be related to volume depletion by osmotic diuresis, decreased trans glomerular pressure, and hypoxic injury to the renal medulla[102]. However, none of the CVOTs[6-8] reported a higher incidence of AKI with SGLT2i and the dedicated renal outcome trials reported SGLT2i to be beneficial for renal outcome[20,103,104]. The real-world data also suggest the use of SGLT2i is not associated with an increased risk for AKI[105]. However, a careful selection of patients initiated on an SGLT2i and close monitoring of eGFR would be useful to reduce the likelihood of AKI in the real-world clinical practice settings.

There was a postulation that SGLT2i may affect bone mineral density and bone quality, thereby increasing the risk for fragility fractures. Initial studies with canagliflozin reported a mild increase in serum phosphate, parathyroid hormone, bone resorption markers, and bone formation markers and a slight reduction in total hip bone mineral density without affecting the rest of the skeleton[106,107]. In the CANVAS trial, the risk for fracture was significantly higher with canagliflozin versus placebo but the same finding was not replicated in other two large CV outcome studies with empagliflozin and dapagliflozin[6-8]. The underlying mechanism for increased incidence of fracture with canagliflozin can be a direct effect on bone metabolism, or it can be due to an increased risk for falls due to orthostatic hypertension associated with SGLT2i. Recently three meta-analyses[108-110] did not find any increased incidence of bone fractures with SGLT2i compared to either placebo or active treatment, thus reassuring about the fracture risk of this class of drugs.

An increased incidence of lower limb amputations was seen with canagliflozin compared to placebo (6.3 vs 3.4 participants with amputation per 1000 patient-years) in CANVAS trial[7] as well as in a pharmacovigilance analysis of FARES data[111], but not with other SGLT2i. The difference can be partly explained by the differences in study design and data collection regarding lower limb amputation. A recent meta-analysis[112] of 14 RCTs reported no increase in lower limb amputation with SGLT2 inhibitor as a class. However, upon subgroup analysis there was a higher fracture risk with canagliflozin versus placebo or non-SGLT2i antidiabetic drugs. The fact that there is inter-SGLT2i differences in the risk for fractures and for lower limb amputation remains to be confirmed yet.

A concern regarding an increase in bladder malignancies in male patients receiving dapagliflozin was raised in initial studies but was not clear whether it was due to earlier diagnosis of malignancies in the sub-clinical phase or there indeed was a true increase in rates of bladder cancer[95]. Possible mechanisms for tumor genesis with an SGLT2i could be the enhanced tumor growth from bladder epithelium due to persistent glycosuria in as well as the effects of chronic cystitis or recurrent urinary tract infections. However, one metanalysis[113] did not confirm any significant increase in malignancies with the use of SGLT 2 inhibitors, and further research is needed in this field.

SGLT2 IN THE PREVENTION/MANAGEMENT OF HEART DISEASES: RECOMMENDATIONS

Table 3 summarises the current position of different bodies regarding the use of SGLT2i in patients with heart disease with and without diabetes. While they don’t specifically prefer one SGLT2i over another, for a particular indication, all the bodies recommend using SGLT2i that have proven benefits in that aspect.

Table 3 Role of sodium glucose cotransporter 2 inhibitors in the management a prevention of diabetes - position of different guidelines.
Organize groups
Position of different guidelines
ADA, 2023Among people with T2DM who have established ASCVD (a SGLT2i with demonstrated cardiovascular disease benefit is recommended as part of the comprehensive cardiovascular risk reduction and/or glucose - lowering regimens. (LOE: A)
In people with T2DM who have established ASCVD, multiple atherosclerotic cardiovascular disease risk factors, or DKD, a SGLT2i with demonstrated cardiovascular benefit is recommended to reduce the risk of major adverse cardiovascular events and/or heart failure hospitalization. (LOE: A)
In people with T2DM and established ASCVD or multiple risk factors for atherosclerotic cardiovascular disease, combined therapy with a SGLT2i and a GLP1-RA may be considered for additive reduction in the risk of adverse cardiovascular and kidney events. (LOE: A)
In people with T2DM and established heart failure with either preserved or reduced ejection fraction, a SGLT2i with proven benefit in this patient population is recommended to reduce risk of worsening heart failure and cardiovascular death. (LOE: A)
In people with T2DM and established heart failure with either preserved or reduced ejection fraction, a SGLT2i with proven benefit in this patient population is recommended to improve symptoms, physical limitations, and quality of life. (LOE: A)
AACE, 2023 SGLT2i should be started irrespective of glycemic target or other T2DM therapies in patients with T2DM and ASCVD or at high risk for ASCVD (albuminuria/proteinuria, hypertension and left ventricular hypertrophy, LV systolic or diastolic dysfunction, ankle-branchial index < 0.9)
ACC/AHA, 2022 In patients with symptomatic chronic HFrEF, SGLT2i are recommended to reduce hospitalisation for heart failure and CV mortality, irrespective of the presence of type 2 diabetes. (COR: 1, LOE: A)
In patients with HFmrEF, SGLT2i can be beneficial in decreasing HF hospitalisation and CV mortality (COR: 1, LOE: A)
In patients with HFpEF, SGLT2i can be beneficial in decreasing HF hospitalisation and CV mortality (COR: 1, LOE: A)
ESC, 2022SGLT2i are recommended in all patients with HFrEF and T2DM to reduce the risk of HF hospitalization and CV death. (COR: 1, LOE: A)
SGLT2i are recommended in patients with T2DM and LVEF > 40% (HFmrEF and HFpEF) to reduce the risk of HF hospitalization or CV death. (COR: 1, LOE: A)
SGLT2i are recommended in patients with T2DM with multiple ASCVD risk factors or established ASCVD to reduce the risk of HF hospitalization. (COR: 1, LOE: A)
CHOICE OF SGLT2I - ARE THEY ALL THE SAME?

Although an overall beneficial effect is obvious, there is some heterogeneity in the findings from different SGLT2i CVOTs. Also, although no head-to-head comparison data between the SGLT2i are available, there has been some numerical differences in their results. Whether the differences arise from variation in the pharmacologic properties of the various SGLT2 inhibitors or are the result of disparities in trial design and/or baseline characteristics of the study participants remain unclear. However, the latter limits direct comparability of the CVOTs.

One postulated mechanism has been the differences in specificity of the molecules to the SGLT2 receptor over SGLT1, which is greater than 2500-fold for empagliflozin, 2235-fold for ertugliflozin, 1200-fold for dapagliflozin and 200-fold for canagliflozin[114]. In the EMPA-REG trial, empagliflozin users showed a mean improvement in HbA1c by 0.24% over 206 wk, while canagliflozin over 188 wk resulted in improvement by 0.58% in the CANVAS program.

A network meta-analysis of 38 RCTs including canagliflozin, dapagliflozin or empagliflozin that were published up to November 2015 found that canagliflozin 300 mg reduced HbA1c, FPG and systolic blood pressure and increased LDL cholesterol to a greater extent compared to other SGLT2i or to 100 mg Canagliflozin[115].

However, contrary to the biologic plausibility based on SGLT2 selectivity, the CV superiority for 3P-MACE has been seen with empagliflozin in the EMPA-REG and to some extent with canagliflozin use in the CANVAS and CREDENCE studies, but it has not been seen with dapagliflozin in the DECLARE-TIMI, or with ertugliflozin in the VERTIS-CV trials[6-9].

Although the half-lives, metabolism and elimination of these drugs are quite similar, their oral bioavailability is variable, lowest being for canagliflozin (65%) and the highest for ertugliflozin (100%). There is also some variation in the volume of distribution and plasma protein binding. But to what extent these differences translate into clinically important discrepancies is yet unknown[116].

All the SGLT2i so far have demonstrated non-inferiority for 3P-MACE compared to placebo, and superior outcomes with respect to HHF outcomes, except for ertugliflozin which failed to demonstrate superiority in the VERTIS-CV trial. The studies included population with different co-morbidities and risk factors which can significantly reduce the incidence of CV or renal events during the study period. In the EMPA-REG and VERTIS-CV trials, all the participants had established ASCVD whereas in the CANVAS trial, 66% had ASCVD and in the DECLARE-TIMI, 41% had ASCVD whereas the remaining participants had multiple CV risk factors[6-9]. With regards to HF, the initial CVOTs were inconsistent in their reporting of whether the baseline HF status was HFrEF or HFpEF. A higher proportion of participants in the VERTIS-CV trial had HF (24%) at baseline, compared to the other major CVOTs (approximately 10%-15%), of whom 80% had HFpEF, which might have skewed the results of the trial[9].

The beneficial effects seen in the DECLARE-TIMI trial with respect to the combined outcome of CV death or HHF was driven mainly by gross reduction in HHF. The decrease in all-cause mortality seen in the EMPA-REG and DAPA-HF trials was predominantly due to reduction in CV mortality. Regarding 3P-MACE, it is believed that the differences in the number of participants with HF, specifically HFpEF in the VERTIS-CV trial and also low rates of CV events in the DECLARE-TIMI trial might explain why the superiority of SGLT2i for 3-PMACE couldn’t be demonstrated in these trials although this was evident in the EMPA-REG and CANVAS trials.

Notably, conflicting results have been seen between different trials with the same drug, for example empagliflozin in the EMPA-REG and EMPEROR-preserved) or dapagliflozin in the DECLARE-TIMI and DAPA-HF trials with respect to CV or all-cause mortality[13,19]. Mortality reduction with empagliflozin was seen in the EMPAREG-OUTCOME trial but not in the EMPEROR-reduced trial. Participants in the HF outcome trials (DAPA-HF and EMPEROR-reduced) were different from participants of older CVOTs like EMPAREG or DECLARE-TIMI, as, in the former group, most of the participants had HFrEF, lower BMI, systolic blood pressure and mean eGFR, and a significant proportion was non-diabetic. However, on comparing the HF trials DAPA-HF (dapagliflozin) and EMPEROR-reduced (empagliflozin), significant reduction in the risk of CV deaths was seen in the DAPA-HF trial, but not with empagliflozin in the EMPEROR-reduced trial[35,45] thus raising a question on whether baseline characteristics alone account for the changes or there is a role of individual pharmacologic property of the SGLT2i. Following the results of the DAPA-HF and EMPEROR-reduced trials, it became clear that the beneficial effects on CV death or HHF was observed in participants irrespective of the presence of diabetes.

A recent real-world study involving 25315 patients (empagliflozin: 5302, dapagliflozin: 4681, canagliflozin: 4411, other SGLT2 inhibitors: 10921), the authors reported no significant differences in the risk of developing HF, MI, AP, stroke, and AF among the individual SGLT2 inhibitors. The robustness of the results was also confirmed through a multitude of sensitivity analyses[117].

Overall, till date, there is insufficient evidence to suggest the superiority of any SGLT2i over the other with regards to different CV outcomes and it appears to be a class-effect. Data from available CVOTs may aid in making a choice of one particular SGLT2i over the other depending on the clinical scenario and the purpose of use. Thus, in patients with established ASCVD, empagliflozin appears to have an upper hand when it comes to mortality reduction and empagliflozin and canagliflozin both seem to score over dapagliflozin in reduction of 3P-MACE or the composite of HHF and CV death. However, in those with HFrEF, dapagliflozin seems to score better than empagliflozin in mortality reduction. Ertugliflozin is not a prudent choice in any scenario, whereas for HFpEF, both dapagliflozin and empagliflozin are equal and can be used irrespective of the presence of diabetes. Talking of those without established ASCVD, a 300 mg dose of Canagliflozin can be a prudent choice based on its effects on the risk factors like the greater degrees of weight loss, BP and HbA1c% reduction. However, this is more of a personalised opinion rather than an evidence based one and the choice must be balanced against the cost in individual countries, availability, and the risk for adverse effects.

SGLT2I FOR CARDIO-PROTECTION IN TYPE 1 DIABETES? - THE CURRENT STATUS

It is a known fact now that children and adults with type 1 diabetes have insulin resistance and display features of metabolic syndrome like obesity, dyslipidemia, hypertension. A significant proportion of T1DM patients go on to develop ASCVD and HF, thus raising the question of the role of SGLT2i as adjuncts to insulin in these patients[118].

The metabolic benefits of SGLT2 inhibition like weight reduction and better Hba1c reduction have been demonstrated in type 1 diabetes in three phase 3 clinical trials- the EASE (empagliflozin), DEPICT (dapagliflozin), and inTandem (sotagliflozin, a dual SGLT1/2 inhibitor)[119].

In 2019, the European Medicines Agency (EMA) had approved dapagliflozin 5 mg as an adjunct pharmacotherapy for overweight-obese individuals with type 1 diabetes with overweight[120]. However, the FDA declined the applications for dapagliflozin and empagliflozin due to risk concerns for DKA[121]. Indeed, SGLT2i-induced glucosuria can lead to negative caloric balance and promote ketone generation. Ketosis can sometimes occur without hyperglycaemia, known as euglycemic DKA, which makes detection more difficult. In most clinical trials, the risk of DKA was dose-dependent and not seen in participants receiving very low doses of SGLT2 inhibitors like dapagliflozin 5 mg or empagliflozin 2.5 mg[118].

But again, it is unclear whether the cardiorenal protection seen with SGLT2i would manifest at such low doses. Also, the risk of DKA could be higher outside the ideal settings of a clinical trial. The EMA approval of dapagliflozin in T1DM was first made subject to a condition of strict risk mitigation strategy and close supervision by prescribers and consequently, was abruptly reversed in October 2021[122]. It is reassuring to note that most real-world data do not show an alarming increase in DKA risk with SGLT2i in T1DM[123].

CONCLUSION

The discovery of the cardioprotective effects of SGLT2i have brought about a paradigm shift in the management of T2DM with a shift of focus towards a holistic approach to target organ protection in T2DM rather than glycemic control alone. While their roles in HF and cardiac risk factors are well established, they have the potential to be used in other heart diseases like diabetic cardiomyopathy and cardiac autonomic neuropathy as well.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Cardiac and cardiovascular systems

Country of origin: United Kingdom

Peer-review report’s classification

Scientific Quality: Grade B

Novelty: Grade B

Creativity or Innovation: Grade B

Scientific Significance: Grade B

P-Reviewer: Lymperopoulos A, United States S-Editor: Li L L-Editor: A P-Editor: Yuan YY

References
1.  Saisho Y. SGLT2 Inhibitors: the Star in the Treatment of Type 2 Diabetes? Diseases. 2020;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 55]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
2.  Packer M, Anker SD, Butler J, Filippatos G, Zannad F. 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;2:1025-1029.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 233]  [Cited by in F6Publishing: 255]  [Article Influence: 51.0]  [Reference Citation Analysis (0)]
3.  Perkovic V, Jardine MJ, Neal B, Bompoint S, Heerspink HJL, Charytan DM, Edwards R, Agarwal R, Bakris G, Bull S, Cannon CP, Capuano G, Chu PL, de Zeeuw D, Greene T, Levin A, Pollock C, Wheeler DC, Yavin Y, Zhang H, Zinman B, Meininger G, Brenner BM, Mahaffey KW; CREDENCE Trial Investigators. Canagliflozin and Renal Outcomes in Type 2 Diabetes and Nephropathy. N Engl J Med. 2019;380:2295-2306.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2826]  [Cited by in F6Publishing: 3329]  [Article Influence: 665.8]  [Reference Citation Analysis (0)]
4.  McMurray JJV, DeMets DL, Inzucchi SE, Køber L, Kosiborod MN, Langkilde AM, Martinez FA, Bengtsson O, Ponikowski P, Sabatine MS, Sjöstrand M, Solomon SD; DAPA-HF Committees and Investigators. The Dapagliflozin And Prevention of Adverse-outcomes in Heart Failure (DAPA-HF) trial: baseline characteristics. Eur J Heart Fail. 2019;21:1402-1411.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 126]  [Article Influence: 25.2]  [Reference Citation Analysis (0)]
5.  Heerspink HJL, Stefánsson BV, Correa-Rotter R, Chertow GM, Greene T, Hou FF, Mann JFE, McMurray JJV, Lindberg M, Rossing P, Sjöström CD, Toto RD, Langkilde AM, Wheeler DC; DAPA-CKD Trial Committees and Investigators. Dapagliflozin in Patients with Chronic Kidney Disease. N Engl J Med. 2020;383:1436-1446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1586]  [Cited by in F6Publishing: 2357]  [Article Influence: 589.3]  [Reference Citation Analysis (1)]
6.  Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE; EMPA-REG OUTCOME Investigators. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med. 2015;373:2117-2128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7124]  [Cited by in F6Publishing: 7460]  [Article Influence: 828.9]  [Reference Citation Analysis (0)]
7.  Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, Erondu N, Shaw W, Law G, Desai M, Matthews DR; CANVAS Program Collaborative Group. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N Engl J Med. 2017;377:644-657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4498]  [Cited by in F6Publishing: 4820]  [Article Influence: 688.6]  [Reference Citation Analysis (0)]
8.  Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Silverman MG, Zelniker TA, Kuder JF, Murphy SA, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Ruff CT, Gause-Nilsson IAM, Fredriksson M, Johansson PA, Langkilde AM, Sabatine MS; DECLARE-TIMI 58 Investigators. Dapagliflozin and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med. 2019;380:347-357.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3164]  [Cited by in F6Publishing: 3648]  [Article Influence: 729.6]  [Reference Citation Analysis (0)]
9.  Cannon CP, Pratley R, Dagogo-Jack S, Mancuso J, Huyck S, Masiukiewicz U, Charbonnel B, Frederich R, Gallo S, Cosentino F, Shih WJ, Gantz I, Terra SG, Cherney DZI, McGuire DK; VERTIS CV Investigators. Cardiovascular Outcomes with Ertugliflozin in Type 2 Diabetes. N Engl J Med. 2020;383:1425-1435.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 673]  [Cited by in F6Publishing: 842]  [Article Influence: 210.5]  [Reference Citation Analysis (0)]
10.  Bhatt DL, Szarek M, Pitt B, Cannon CP, Leiter LA, McGuire DK, Lewis JB, Riddle MC, Inzucchi SE, Kosiborod MN, Cherney DZI, Dwyer JP, Scirica BM, Bailey CJ, Díaz R, Ray KK, Udell JA, Lopes RD, Lapuerta P, Steg PG; SCORED Investigators. Sotagliflozin in Patients with Diabetes and Chronic Kidney Disease. N Engl J Med. 2021;384:129-139.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 434]  [Cited by in F6Publishing: 618]  [Article Influence: 206.0]  [Reference Citation Analysis (0)]
11.  Heidenreich PA, Bozkurt B, Aguilar D, Allen LA, Byun JJ, Colvin MM, Deswal A, Drazner MH, Dunlay SM, Evers LR, Fang JC, Fedson SE, Fonarow GC, Hayek SS, Hernandez AF, Khazanie P, Kittleson MM, Lee CS, Link MS, Milano CA, Nnacheta LC, Sandhu AT, Stevenson LW, Vardeny O, Vest AR, Yancy CW. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation. 2022;145:e895-e1032.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 340]  [Cited by in F6Publishing: 643]  [Article Influence: 321.5]  [Reference Citation Analysis (0)]
12.  Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, Januzzi J, Verma S, Tsutsui H, Brueckmann M, Jamal W, Kimura K, Schnee J, Zeller C, Cotton D, Bocchi E, Böhm M, Choi DJ, Chopra V, Chuquiure E, Giannetti N, Janssens S, Zhang J, Gonzalez Juanatey JR, Kaul S, Brunner-La Rocca HP, Merkely B, Nicholls SJ, Perrone S, Pina I, Ponikowski P, Sattar N, Senni M, Seronde MF, Spinar J, Squire I, Taddei S, Wanner C, Zannad F; EMPEROR-Reduced Trial Investigators. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N Engl J Med. 2020;383:1413-1424.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1875]  [Cited by in F6Publishing: 2590]  [Article Influence: 647.5]  [Reference Citation Analysis (0)]
13.  Anker SD, Butler J, Filippatos G, Ferreira JP, Bocchi E, Böhm M, Brunner-La Rocca HP, Choi DJ, Chopra V, Chuquiure-Valenzuela E, Giannetti N, Gomez-Mesa JE, Janssens S, Januzzi JL, Gonzalez-Juanatey JR, Merkely B, Nicholls SJ, Perrone SV, Piña IL, Ponikowski P, Senni M, Sim D, Spinar J, Squire I, Taddei S, Tsutsui H, Verma S, Vinereanu D, Zhang J, Carson P, Lam CSP, Marx N, Zeller C, Sattar N, Jamal W, Schnaidt S, Schnee JM, Brueckmann M, Pocock SJ, Zannad F, Packer M; EMPEROR-Preserved Trial Investigators. Empagliflozin in Heart Failure with a Preserved Ejection Fraction. N Engl J Med. 2021;385:1451-1461.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1192]  [Cited by in F6Publishing: 2014]  [Article Influence: 671.3]  [Reference Citation Analysis (0)]
14.  Solomon SD, McMurray JJV, Claggett B, de Boer RA, DeMets D, Hernandez AF, Inzucchi SE, Kosiborod MN, Lam CSP, Martinez F, Shah SJ, Desai AS, Jhund PS, Belohlavek J, Chiang CE, Borleffs CJW, Comin-Colet J, Dobreanu D, Drozdz J, Fang JC, Alcocer-Gamba MA, Al Habeeb W, Han Y, Cabrera Honorio JW, Janssens SP, Katova T, Kitakaze M, Merkely B, O'Meara E, Saraiva JFK, Tereshchenko SN, Thierer J, Vaduganathan M, Vardeny O, Verma S, Pham VN, Wilderäng U, Zaozerska N, Bachus E, Lindholm D, Petersson M, Langkilde AM; DELIVER Trial Committees and Investigators. Dapagliflozin in Heart Failure with Mildly Reduced or Preserved Ejection Fraction. N Engl J Med. 2022;387:1089-1098.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 874]  [Article Influence: 437.0]  [Reference Citation Analysis (0)]
15.  Salah HM, Al'Aref SJ, Khan MS, Al-Hawwas M, Vallurupalli S, Mehta JL, Mounsey JP, Greene SJ, McGuire DK, Lopes RD, Fudim M. Efficacy and safety of sodium-glucose cotransporter 2 inhibitors initiation in patients with acute heart failure, with and without type 2 diabetes: a systematic review and meta-analysis. Cardiovasc Diabetol. 2022;21:20.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 30]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
16.  Birkeland KI, Jørgensen ME, Carstensen B, Persson F, Gulseth HL, Thuresson M, Fenici P, Nathanson D, Nyström T, Eriksson JW, Bodegård J, Norhammar A. Cardiovascular mortality and morbidity in patients with type 2 diabetes following initiation of sodium-glucose co-transporter-2 inhibitors versus other glucose-lowering drugs (CVD-REAL Nordic): a multinational observational analysis. Lancet Diabetes Endocrinol. 2017;5:709-717.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 244]  [Cited by in F6Publishing: 250]  [Article Influence: 35.7]  [Reference Citation Analysis (0)]
17.  Zelniker TA, Wiviott SD, Raz I, Im K, Goodrich EL, Bonaca MP, Mosenzon O, Kato ET, Cahn A, Furtado RHM, Bhatt DL, Leiter LA, McGuire DK, Wilding JPH, Sabatine MS. 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;393:31-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1634]  [Cited by in F6Publishing: 1730]  [Article Influence: 346.0]  [Reference Citation Analysis (0)]
18.  Butler J, Usman MS, Filippatos G, Ferreira JP, Böhm M, Brueckmann M, Januzzi JL, Kaul S, Piña IL, Ponikowski P, Senni M, Sumin M, Verma S, Zaremba-Pechmann L, Pocock SJ, Packer M, Anker S. Safety and Efficacy of Empagliflozin and Diuretic Use in Patients with Heart Failure and Preserved Ejection Fraction: A Post Hoc Analysis of the EMPEROR-Preserved Trial. JAMA Cardiol. 2023;8:640-649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 3]  [Reference Citation Analysis (0)]
19.  Bhatt DL, Verma S, Pitt B. EMPEROR-Preserved: A promise fulfilled. Cell Metab. 2021;33:2099-2103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 7]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
20.  Zannad F, Ferreira JP, Pocock SJ, Anker SD, Butler J, Filippatos G, Brueckmann M, Ofstad AP, Pfarr E, Jamal W, Packer M. SGLT2 inhibitors in patients with heart failure with reduced ejection fraction: a meta-analysis of the EMPEROR-Reduced and DAPA-HF trials. Lancet. 2020;396:819-829.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 621]  [Cited by in F6Publishing: 744]  [Article Influence: 186.0]  [Reference Citation Analysis (0)]
21.  Jongs N, Greene T, Chertow GM, McMurray JJV, Langkilde AM, Correa-Rotter R, Rossing P, Sjöström CD, Stefansson BV, Toto RD, Wheeler DC, Heerspink HJL; DAPA-CKD Trial Committees and Investigators. Effect of dapagliflozin on urinary albumin excretion in patients with chronic kidney disease with and without type 2 diabetes: a prespecified analysis from the DAPA-CKD trial. Lancet Diabetes Endocrinol. 2021;9:755-766.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 84]  [Article Influence: 28.0]  [Reference Citation Analysis (0)]
22.  Mordi IR, Lang CC. Glucose-Lowering and Metabolic Effects of SGLT2 Inhibitors. Heart Fail Clin. 2022;18:529-538.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
23.  Lavalle-González FJ, Januszewicz A, Davidson J, Tong C, Qiu R, Canovatchel W, Meininger G. Efficacy and safety of canagliflozin compared with placebo and sitagliptin in patients with type 2 diabetes on background metformin monotherapy: a randomised trial. Diabetologia. 2013;56:2582-2592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 366]  [Cited by in F6Publishing: 378]  [Article Influence: 34.4]  [Reference Citation Analysis (0)]
24.  Schernthaner G, Gross JL, Rosenstock J, Guarisco M, Fu M, Yee J, Kawaguchi M, Canovatchel W, Meininger G. Canagliflozin compared with sitagliptin for patients with type 2 diabetes who do not have adequate glycemic control with metformin plus sulfonylurea: a 52-week randomized trial. Diabetes Care. 2013;36:2508-2515.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 362]  [Cited by in F6Publishing: 377]  [Article Influence: 34.3]  [Reference Citation Analysis (0)]
25.  Kamijo Y, Ishii H, Yamamoto T, Kobayashi K, Asano H, Miake S, Kanda E, Urata H, Yoshida M. Potential Impact on Lipoprotein Subfractions in Type 2 Diabetes. Clin Med Insights Endocrinol Diabetes. 2019;12:1179551419866811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
26.  Kalra S, Singh V, Nagrale D. Sodium-Glucose Cotransporter-2 Inhibition and the Glomerulus: A Review. Adv Ther. 2016;33:1502-1518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 28]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
27.  Szekeres Z, Toth K, Szabados E. The Effects of SGLT2 Inhibitors on Lipid Metabolism. Metabolites. 2021;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 72]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
28.  Palmer BF, Clegg DJ. Kidney-Protective Effects of SGLT2 Inhibitors. Clin J Am Soc Nephrol. 2023;18:279-289.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
29.  Cherney DZ, Perkins BA, Soleymanlou N, Maione M, Lai V, Lee A, Fagan NM, Woerle HJ, Johansen OE, Broedl UC, von Eynatten M. Renal hemodynamic effect of sodium-glucose cotransporter 2 inhibition in patients with type 1 diabetes mellitus. Circulation. 2014;129:587-597.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 847]  [Cited by in F6Publishing: 917]  [Article Influence: 83.4]  [Reference Citation Analysis (0)]
30.  Heerspink HJL, Perco P, Mulder S, Leierer J, Hansen MK, Heinzel A, Mayer G. Canagliflozin reduces inflammation and fibrosis biomarkers: a potential mechanism of action for beneficial effects of SGLT2 inhibitors in diabetic kidney disease. Diabetologia. 2019;62:1154-1166.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 248]  [Cited by in F6Publishing: 255]  [Article Influence: 51.0]  [Reference Citation Analysis (0)]
31.  Zhao Y, Xu L, Tian D, Xia P, Zheng H, Wang L, Chen L. Effects of sodium-glucose co-transporter 2 (SGLT2) inhibitors on serum uric acid level: A meta-analysis of randomized controlled trials. Diabetes Obes Metab. 2018;20:458-462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 185]  [Cited by in F6Publishing: 216]  [Article Influence: 36.0]  [Reference Citation Analysis (0)]
32.  Lai SW, Hwang BF, Kuo YH, Liu CS, Liao KF. Sodium-glucose cotransporter-2 inhibitors use and the risk of gout: a systematic review and meta-analysis. Front Endocrinol (Lausanne). 2023;14:1158153.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 4]  [Reference Citation Analysis (0)]
33.  Packer M. Uric Acid Is a Biomarker of Oxidative Stress in the Failing Heart: Lessons Learned from Trials With Allopurinol and SGLT2 Inhibitors. J Card Fail. 2020;26:977-984.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 42]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
34.  Chung MC, Hung PH, Hsiao PJ, Wu LY, Chang CH, Wu MJ, Shieh JJ, Chung CJ. Association of Sodium-Glucose Transport Protein 2 Inhibitor Use for Type 2 Diabetes and Incidence of Gout in Taiwan. JAMA Netw Open. 2021;4:e2135353.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
35.  Ekanayake P, Mudaliar S. Increase in hematocrit with SGLT-2 inhibitors - Hemoconcentration from diuresis or increased erythropoiesis after amelioration of hypoxia? Diabetes Metab Syndr. 2023;17:102702.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 9]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
36.  O'Neill J, Fasching A, Pihl L, Patinha D, Franzén S, Palm F. Acute SGLT inhibition normalizes O2 tension in the renal cortex but causes hypoxia in the renal medulla in anaesthetized control and diabetic rats. Am J Physiol Renal Physiol. 2015;309:F227-F234.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 171]  [Article Influence: 19.0]  [Reference Citation Analysis (0)]
37.  Nasiri-Ansari Ν, Dimitriadis GK, Agrogiannis G, Perrea D, Kostakis ID, Kaltsas G, Papavassiliou AG, Randeva HS, Kassi E. Canagliflozin attenuates the progression of atherosclerosis and inflammation process in APOE knockout mice. Cardiovasc Diabetol. 2018;17:106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 97]  [Article Influence: 16.2]  [Reference Citation Analysis (0)]
38.  Liu Y, Wu M, Xu B, Kang L. Empagliflozin Alleviates Atherosclerosis Progression by Inhibiting Inflammation and Sympathetic Activity in a Normoglycemic Mouse Model. J Inflamm Res. 2021;14:2277-2287.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 18]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
39.  Symeonidis A, Kouraklis-Symeonidis A, Psiroyiannis A, Leotsinidis M, Kyriazopoulou V, Vassilakos P, Vagenakis A, Zoumbos N. Inappropriately low erythropoietin response for the degree of anemia in patients with noninsulin-dependent diabetes mellitus. Ann Hematol. 2006;85:79-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 44]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
40.  Daniele G, Xiong J, Solis-Herrera C, Merovci A, Eldor R, Tripathy D, DeFronzo RA, Norton L, Abdul-Ghani M. Dapagliflozin Enhances Fat Oxidation and Ketone Production in Patients With Type 2 Diabetes. Diabetes Care. 2016;39:2036-2041.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 146]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
41.  Bonner C, Kerr-Conte J, Gmyr V, Queniat G, Moerman E, Thévenet J, Beaucamps C, Delalleau N, Popescu I, Malaisse WJ, Sener A, Deprez B, Abderrahmani A, Staels B, Pattou F. Inhibition of the glucose transporter SGLT2 with dapagliflozin in pancreatic alpha cells triggers glucagon secretion. Nat Med. 2015;21:512-517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 446]  [Cited by in F6Publishing: 465]  [Article Influence: 51.7]  [Reference Citation Analysis (0)]
42.  Androutsakos T, Nasiri-Ansari N, Bakasis AD, Kyrou I, Efstathopoulos E, Randeva HS, Kassi E. SGLT-2 Inhibitors in NAFLD: Expanding Their Role beyond Diabetes and Cardioprotection. Int J Mol Sci. 2022;23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 49]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
43.  Ito D, Shimizu S, Inoue K, Saito D, Yanagisawa M, Inukai K, Akiyama Y, Morimoto Y, Noda M, Shimada A. Comparison of Ipragliflozin and Pioglitazone Effects on Nonalcoholic Fatty Liver Disease in Patients With Type 2 Diabetes: A Randomized, 24-Week, Open-Label, Active-Controlled Trial. Diabetes Care. 2017;40:1364-1372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 169]  [Cited by in F6Publishing: 195]  [Article Influence: 27.9]  [Reference Citation Analysis (1)]
44.  Itani T, Ishihara T. Efficacy of canagliflozin against nonalcoholic fatty liver disease: a prospective cohort study. Obes Sci Pract. 2018;4:477-482.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 52]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
45.  Eriksson JW, Lundkvist P, Jansson PA, Johansson L, Kvarnström M, Moris L, Miliotis T, Forsberg GB, Risérus U, Lind L, Oscarsson J. Effects of dapagliflozin and n-3 carboxylic acids on non-alcoholic fatty liver disease in people with type 2 diabetes: a double-blind randomised placebo-controlled study. Diabetologia. 2018;61:1923-1934.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 200]  [Cited by in F6Publishing: 228]  [Article Influence: 38.0]  [Reference Citation Analysis (1)]
46.  Akuta N, Watanabe C, Kawamura Y, Arase Y, Saitoh S, Fujiyama S, Sezaki H, Hosaka T, Kobayashi M, Suzuki Y, Suzuki F, Ikeda K, Kumada H. Effects of a sodium-glucose cotransporter 2 inhibitor in nonalcoholic fatty liver disease complicated by diabetes mellitus: Preliminary prospective study based on serial liver biopsies. Hepatol Commun. 2017;1:46-52.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 76]  [Article Influence: 10.9]  [Reference Citation Analysis (0)]
47.  Yeghiazarians Y, Jneid H, Tietjens JR, Redline S, Brown DL, El-Sherif N, Mehra R, Bozkurt B, Ndumele CE, Somers VK. Obstructive Sleep Apnea and Cardiovascular Disease: A Scientific Statement From the American Heart Association. Circulation. 2021;144:e56-e67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 382]  [Article Influence: 127.3]  [Reference Citation Analysis (0)]
48.  Tang Y, Sun Q, Bai XY, Zhou YF, Zhou QL, Zhang M. Effect of dapagliflozin on obstructive sleep apnea in patients with type 2 diabetes: a preliminary study. Nutr Diabetes. 2019;9:32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
49.  Sawada K, Karashima S, Kometani M, Oka R, Takeda Y, Sawamura T, Fujimoto A, Demura M, Wakayama A, Usukura M, Yagi K, Yoneda T. Effect of sodium glucose cotransporter 2 inhibitors on obstructive sleep apnea in patients with type 2 diabetes. Endocr J. 2018;65:461-467.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
50.  Patoulias D, Dimosiari A, Michailidis T, Roditis P, Lampropoulos S. Meta-analysis Evaluating the Effect of Sodium-Glucose Co-Transporter-2 Inhibitors on the Risk for New-Onset Obstructive Sleep Apnea in Patients With Type 2 Diabetes Mellitus Regardless of Cardiovascular Disease at Baseline. Am J Cardiol. 2022;179:116-117.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
51.  Tanriover C, Ucku D, Akyol M, Cevik E, Kanbay A, Sridhar VS, Cherney DZI, Kanbay M. Potential Use of SGLT-2 Inhibitors in Obstructive Sleep Apnea: A new treatment on the horizon. Sleep Breath. 2023;27:77-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
52.  Pellicori P, Ofstad AP, Fitchett D, Zeller C, Wanner C, George J, Zinman B, Brueckmann M, Lindenfeld J. Early benefits of empagliflozin in patients with or without heart failure: findings from EMPA-REG OUTCOME. ESC Heart Fail. 2020;7:3401-3407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
53.  Inzucchi SE, Khunti K, Fitchett DH, Wanner C, Mattheus M, George JT, Ofstad AP, Zinman B. Cardiovascular Benefit of Empagliflozin Across the Spectrum of Cardiovascular Risk Factor Control in the EMPA-REG OUTCOME Trial. J Clin Endocrinol Metab. 2020;105:3025-3035.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 15]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
54.  Carluccio E, Biagioli P, Reboldi G, Mengoni A, Lauciello R, Zuchi C, D'Addario S, Bardelli G, Ambrosio G. Left ventricular remodeling response to SGLT2 inhibitors in heart failure: an updated meta-analysis of randomized controlled studies. Cardiovasc Diabetol. 2023;22:235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 8]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
55.  Kondo H, Akoumianakis I, Badi I, Akawi N, Kotanidis CP, Polkinghorne M, Stadiotti I, Sommariva E, Antonopoulos AS, Carena MC, Oikonomou EK, Reus EM, Sayeed R, Krasopoulos G, Srivastava V, Farid S, Chuaiphichai S, Shirodaria C, Channon KM, Casadei B, Antoniades C. Effects of canagliflozin on human myocardial redox signalling: clinical implications. Eur Heart J. 2021;42:4947-4960.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 52]  [Article Influence: 17.3]  [Reference Citation Analysis (0)]
56.  Uthman L, Baartscheer A, Schumacher CA, Fiolet JWT, Kuschma MC, Hollmann MW, Coronel R, Weber NC, Zuurbier CJ. Direct Cardiac Actions of Sodium Glucose Cotransporter 2 Inhibitors Target Pathogenic Mechanisms Underlying Heart Failure in Diabetic Patients. Front Physiol. 2018;9:1575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 119]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
57.  Philippaert K, Kalyaanamoorthy S, Fatehi M, Long W, Soni S, Byrne NJ, Barr A, Singh J, Wong J, Palechuk T, Schneider C, Darwesh AM, Maayah ZH, Seubert JM, Barakat K, Dyck JRB, Light PE. Cardiac Late Sodium Channel Current Is a Molecular Target for the Sodium/Glucose Cotransporter 2 Inhibitor Empagliflozin. Circulation. 2021;143:2188-2204.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 98]  [Article Influence: 32.7]  [Reference Citation Analysis (0)]
58.  Amaral N, Okonko DO. Metabolic abnormalities of the heart in type II diabetes. Diab Vasc Dis Res. 2015;12:239-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
59.  Tabatabaei Dakhili SA, Greenwell AA, Ussher JR. Pyruvate Dehydrogenase Complex and Glucose Oxidation as a Therapeutic Target in Diabetic Heart Disease. J Lipid Atheroscler. 2023;12:47-57.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
60.  Mustroph J, Lücht CM, Wagemann O, Sowa T, Hammer KP, Sag CM, Tarnowski D, Holzamer A, Pabel S, Beuthner BE, Sossalla S, Maier LS, Wagner S. Empagliflozin enhances human and murine cardiomyocyte glucose uptake by increased expression of GLUT1. Diabetologia. 2019;62:726-729.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 17]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
61.  Verma S, Rawat S, Ho KL, Wagg CS, Zhang L, Teoh H, Dyck JE, Uddin GM, Oudit GY, Mayoux E, Lehrke M, Marx N, Lopaschuk GD. Empagliflozin Increases Cardiac Energy Production in Diabetes: Novel Translational Insights Into the Heart Failure Benefits of SGLT2 Inhibitors. JACC Basic Transl Sci. 2018;3:575-587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 250]  [Article Influence: 41.7]  [Reference Citation Analysis (0)]
62.  Oshima H, Miki T, Kuno A, Mizuno M, Sato T, Tanno M, Yano T, Nakata K, Kimura Y, Abe K, Ohwada W, Miura T. Empagliflozin, an SGLT2 Inhibitor, Reduced the Mortality Rate after Acute Myocardial Infarction with Modification of Cardiac Metabolomes and Antioxidants in Diabetic Rats. J Pharmacol Exp Ther. 2019;368:524-534.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 61]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
63.  Aragón-Herrera A, Feijóo-Bandín S, Otero Santiago M, Barral L, Campos-Toimil M, Gil-Longo J, Costa Pereira TM, García-Caballero T, Rodríguez-Segade S, Rodríguez J, Tarazón E, Roselló-Lletí E, Portolés M, Gualillo O, González-Juanatey JR, Lago F. Empagliflozin reduces the levels of CD36 and cardiotoxic lipids while improving autophagy in the hearts of Zucker diabetic fatty rats. Biochem Pharmacol. 2019;170:113677.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 71]  [Article Influence: 14.2]  [Reference Citation Analysis (0)]
64.  Wu P, Wen W, Li J, Xu J, Zhao M, Chen H, Sun J. Systematic Review and Meta-Analysis of Randomized Controlled Trials on the Effect of SGLT2 Inhibitor on Blood Leptin and Adiponectin Level in Patients with Type 2 Diabetes. Horm Metab Res. 2019;51:487-494.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 64]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
65.  Kim JH, Lee M, Kim SH, Kim SR, Lee BW, Kang ES, Cha BS, Cho JW, Lee YH. Sodium-glucose cotransporter 2 inhibitors regulate ketone body metabolism via inter-organ crosstalk. Diabetes Obes Metab. 2019;21:801-811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 40]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
66.  Pietschner R, Kolwelter J, Bosch A, Striepe K, Jung S, Kannenkeril D, Ott C, Schiffer M, Achenbach S, Schmieder RE. Effect of empagliflozin on ketone bodies in patients with stable chronic heart failure. Cardiovasc Diabetol. 2021;20:219.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 16]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
67.  Tanajak P, Sa-Nguanmoo P, Sivasinprasasn S, Thummasorn S, Siri-Angkul N, Chattipakorn SC, Chattipakorn N. Cardioprotection of dapagliflozin and vildagliptin in rats with cardiac ischemia-reperfusion injury. J Endocrinol. 2018;236:69-84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 84]  [Article Influence: 14.0]  [Reference Citation Analysis (0)]
68.  Shao Q, Meng L, Lee S, Tse G, Gong M, Zhang Z, Zhao J, Zhao Y, Li G, Liu T. Empagliflozin, a sodium glucose co-transporter-2 inhibitor, alleviates atrial remodeling and improves mitochondrial function in high-fat diet/streptozotocin-induced diabetic rats. Cardiovasc Diabetol. 2019;18:165.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 160]  [Article Influence: 32.0]  [Reference Citation Analysis (0)]
69.  Hodrea J, Balogh DB, Hosszu A, Lenart L, Besztercei B, Koszegi S, Sparding N, Genovese F, Wagner LJ, Szabo AJ, Fekete A. Reduced O-GlcNAcylation and tubular hypoxia contribute to the antifibrotic effect of SGLT2 inhibitor dapagliflozin in the diabetic kidney. Am J Physiol Renal Physiol. 2020;318:F1017-F1029.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 29]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
70.  Huang K, Luo X, Liao B, Li G, Feng J. Insights into SGLT2 inhibitor treatment of diabetic cardiomyopathy: focus on the mechanisms. Cardiovasc Diabetol. 2023;22:86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 16]  [Reference Citation Analysis (0)]
71.  Shigiyama F, Kumashiro N, Miyagi M, Ikehara K, Kanda E, Uchino H, Hirose T. Effectiveness of dapagliflozin on vascular endothelial function and glycemic control in patients with early-stage type 2 diabetes mellitus: DEFENCE study. Cardiovasc Diabetol. 2017;16:84.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 157]  [Article Influence: 22.4]  [Reference Citation Analysis (0)]
72.  Oelze M, Kröller-Schön S, Welschof P, Jansen T, Hausding M, Mikhed Y, Stamm P, Mader M, Zinßius E, Agdauletova S, Gottschlich A, Steven S, Schulz E, Bottari SP, Mayoux E, Münzel T, Daiber A. The sodium-glucose co-transporter 2 inhibitor empagliflozin improves diabetes-induced vascular dysfunction in the streptozotocin diabetes rat model by interfering with oxidative stress and glucotoxicity. PLoS One. 2014;9:e112394.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 218]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
73.  Ojima A, Matsui T, Nishino Y, Nakamura N, Yamagishi S. Empagliflozin, an Inhibitor of Sodium-Glucose Cotransporter 2 Exerts Anti-Inflammatory and Antifibrotic Effects on Experimental Diabetic Nephropathy Partly by Suppressing AGEs-Receptor Axis. Horm Metab Res. 2015;47:686-692.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 161]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
74.  Paolisso P, Bergamaschi L, Santulli G, Gallinoro E, Cesaro A, Gragnano F, Sardu C, Mileva N, Foà A, Armillotta M, Sansonetti A, Amicone S, Impellizzeri A, Casella G, Mauro C, Vassilev D, Marfella R, Calabrò P, Barbato E, Pizzi C. Infarct size, inflammatory burden, and admission hyperglycemia in diabetic patients with acute myocardial infarction treated with SGLT2-inhibitors: a multicenter international registry. Cardiovasc Diabetol. 2022;21:77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 76]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
75.  Kolijn D, Pabel S, Tian Y, Lódi M, Herwig M, Carrizzo A, Zhazykbayeva S, Kovács Á, Fülöp GÁ, Falcão-Pires I, Reusch PH, Linthout SV, Papp Z, van Heerebeek L, Vecchione C, Maier LS, Ciccarelli M, Tschöpe C, Mügge A, Bagi Z, Sossalla S, Hamdani N. Empagliflozin improves endothelial and cardiomyocyte function in human heart failure with preserved ejection fraction via reduced pro-inflammatory-oxidative pathways and protein kinase Gα oxidation. Cardiovasc Res. 2021;117:495-507.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 161]  [Article Influence: 53.7]  [Reference Citation Analysis (0)]
76.  Brown AJM, Gandy S, McCrimmon R, Houston JG, Struthers AD, Lang CC. A randomized controlled trial of dapagliflozin on left ventricular hypertrophy in people with type two diabetes: the DAPA-LVH trial. Eur Heart J. 2020;41:3421-3432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 128]  [Article Influence: 42.7]  [Reference Citation Analysis (0)]
77.  Hasan R, Lasker S, Hasan A, Zerin F, Zamila M, Parvez F, Rahman MM, Khan F, Subhan N, Alam MA. Canagliflozin ameliorates renal oxidative stress and inflammation by stimulating AMPK-Akt-eNOS pathway in the isoprenaline-induced oxidative stress model. Sci Rep. 2020;10:14659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 61]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
78.  Li C, Zhang J, Xue M, Li X, Han F, Liu X, Xu L, Lu Y, Cheng Y, Li T, Yu X, Sun B, Chen L. SGLT2 inhibition with empagliflozin attenuates myocardial oxidative stress and fibrosis in diabetic mice heart. Cardiovasc Diabetol. 2019;18:15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 198]  [Cited by in F6Publishing: 334]  [Article Influence: 66.8]  [Reference Citation Analysis (0)]
79.  Trang NN, Chung CC, Lee TW, Cheng WL, Kao YH, Huang SY, Lee TI, Chen YJ. Empagliflozin and Liraglutide Differentially Modulate Cardiac Metabolism in Diabetic Cardiomyopathy in Rats. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 22]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
80.  Ye Y, Bajaj M, Yang HC, Perez-Polo JR, Birnbaum Y. SGLT-2 Inhibition with Dapagliflozin Reduces the Activation of the Nlrp3/ASC Inflammasome and Attenuates the Development of Diabetic Cardiomyopathy in Mice with Type 2 Diabetes. Further Augmentation of the Effects with Saxagliptin, a DPP4 Inhibitor. Cardiovasc Drugs Ther. 2017;31:119-132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 180]  [Cited by in F6Publishing: 256]  [Article Influence: 42.7]  [Reference Citation Analysis (0)]
81.  Baartscheer A, Schumacher CA, Wüst RC, Fiolet JW, Stienen GJ, Coronel R, Zuurbier CJ. Empagliflozin decreases myocardial cytoplasmic Na(+) through inhibition of the cardiac Na(+)/H(+) exchanger in rats and rabbits. Diabetologia. 2017;60:568-573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 396]  [Cited by in F6Publishing: 432]  [Article Influence: 61.7]  [Reference Citation Analysis (0)]
82.  Wang CC, Li Y, Qian XQ, Zhao H, Wang D, Zuo GX, Wang K. Empagliflozin alleviates myocardial I/R injury and cardiomyocyte apoptosis via inhibiting ER stress-induced autophagy and the PERK/ATF4/Beclin1 pathway. J Drug Target. 2022;30:858-872.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 16]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
83.  Chang WT, Lin YW, Ho CH, Chen ZC, Liu PY, Shih JY. Dapagliflozin suppresses ER stress and protects doxorubicin-induced cardiotoxicity in breast cancer patients. Arch Toxicol. 2021;95:659-671.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 30]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
84.  Li J, Lin S, Vanhoutte PM, Woo CW, Xu A. Akkermansia Muciniphila Protects Against Atherosclerosis by Preventing Metabolic Endotoxemia-Induced Inflammation in Apoe-/- Mice. Circulation. 2016;133:2434-2446.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 378]  [Cited by in F6Publishing: 465]  [Article Influence: 58.1]  [Reference Citation Analysis (0)]
85.  Hata S, Okamura T, Kobayashi A, Bamba R, Miyoshi T, Nakajima H, Kitagawa N, Hashimoto Y, Majima S, Senmaru T, Okada H, Ushigome E, Nakanishi N, Takakuwa H, Sasano R, Hamaguchi M, Fukui M. Gut Microbiota Changes by an SGLT2 Inhibitor, Luseogliflozin, Alters Metabolites Compared with Those in a Low Carbohydrate Diet in db/db Mice. Nutrients. 2022;14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 19]  [Reference Citation Analysis (0)]
86.  Lymperopoulos A, Borges JI, Cora N, Sizova A. Sympatholytic Mechanisms for the Beneficial Cardiovascular Effects of SGLT2 Inhibitors: A Research Hypothesis for Dapagliflozin's Effects in the Adrenal Gland. Int J Mol Sci. 2021;22.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 24]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
87.  Herat LY, Matthews J, Azzam O, Schlaich MP, Matthews VB. Targeting Features of the Metabolic Syndrome Through Sympatholytic Effects of SGLT2 Inhibition. Curr Hypertens Rep. 2022;24:67-74.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 10]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
88.  Böhm M, Slawik J, Brueckmann M, Mattheus M, George JT, Ofstad AP, Inzucchi SE, Fitchett D, Anker SD, Marx N, Wanner C, Zinman B, Verma S. Efficacy of empagliflozin on heart failure and renal outcomes in patients with atrial fibrillation: data from the EMPA-REG OUTCOME trial. Eur J Heart Fail. 2020;22:126-135.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 57]  [Article Influence: 11.4]  [Reference Citation Analysis (0)]
89.  Sardu C, Massimo Massetti M, Rambaldi P, Gatta G, Cappabianca S, Sasso FC, Santamaria M, Volpicelli M, Ducceschi V, Signoriello G, Paolisso G, Marfella R. SGLT2-inhibitors reduce the cardiac autonomic neuropathy dysfunction and vaso-vagal syncope recurrence in patients with type 2 diabetes mellitus: the SCAN study. Metabolism. 2022;137:155243.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 22]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
90.  Garg V, Verma S, Connelly KA, Yan AT, Sikand A, Garg A, Dorian P, Zuo F, Leiter LA, Zinman B, Jüni P, Verma A, Teoh H, Quan A, Mazer CD, Ha ACT. Does empagliflozin modulate the autonomic nervous system among individuals with type 2 diabetes and coronary artery disease? The EMPA-HEART CardioLink-6 Holter analysis. Metabol Open. 2020;7:100039.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
91.  Balcıoğlu AS, Çelik E, Şahin M, Göçer K, Aksu E, Aykan AÇ. Dapagliflozin Improves Cardiac Autonomic Function Measures in Type 2 Diabetic Patients with Cardiac Autonomic Neuropathy. Anatol J Cardiol. 2022;26:832-840.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 2]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
92.  Vasilakou D, Karagiannis T, Athanasiadou E, Mainou M, Liakos A, Bekiari E, Sarigianni M, Matthews DR, Tsapas A. Sodium-glucose cotransporter 2 inhibitors for type 2 diabetes: a systematic review and meta-analysis. Ann Intern Med. 2013;159:262-274.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 614]  [Cited by in F6Publishing: 627]  [Article Influence: 57.0]  [Reference Citation Analysis (0)]
93.  Liu J, Li L, Li S, Jia P, Deng K, Chen W, Sun X. Effects of SGLT2 inhibitors on UTIs and genital infections in type 2 diabetes mellitus: a systematic review and meta-analysis. Sci Rep. 2017;7:2824.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 140]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
94.  Puckrin R, Saltiel MP, Reynier P, Azoulay L, Yu OHY, Filion KB. SGLT-2 inhibitors and the risk of infections: a systematic review and meta-analysis of randomized controlled trials. Acta Diabetol. 2018;55:503-514.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 133]  [Article Influence: 22.2]  [Reference Citation Analysis (0)]
95.  Ueda P, Svanström H, Melbye M, Eliasson B, Svensson AM, Franzén S, Gudbjörnsdottir S, Hveem K, Jonasson C, Pasternak B. Sodium glucose cotransporter 2 inhibitors and risk of serious adverse events: nationwide register based cohort study. BMJ. 2018;363:k4365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 191]  [Article Influence: 31.8]  [Reference Citation Analysis (0)]
96.  Gadzhanova S, Pratt N, Roughead E. Use of SGLT2 inhibitors for diabetes and risk of infection: Analysis using general practice records from the NPS MedicineWise MedicineInsight program. Diabetes Res Clin Pract. 2017;130:180-185.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 39]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
97.  Arakaki RF. Sodium-glucose cotransporter-2 inhibitors and genital and urinary tract infections in type 2 diabetes. Postgrad Med. 2016;128:409-417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 21]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
98.  Baker WL, Smyth LR, Riche DM, Bourret EM, Chamberlin KW, White WB. Effects of sodium-glucose co-transporter 2 inhibitors on blood pressure: a systematic review and meta-analysis. J Am Soc Hypertens. 2014;8:262-75.e9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 317]  [Article Influence: 31.7]  [Reference Citation Analysis (1)]
99.  Wang Y, Desai M, Ryan PB, DeFalco FJ, Schuemie MJ, Stang PE, Berlin JA, Yuan Z. Incidence of diabetic ketoacidosis among patients with type 2 diabetes mellitus treated with SGLT2 inhibitors and other antihyperglycemic agents. Diabetes Res Clin Pract. 2017;128:83-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 45]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
100.  Monami M, Nreu B, Zannoni S, Lualdi C, Mannucci E. Effects of SGLT-2 inhibitors on diabetic ketoacidosis: A meta-analysis of randomised controlled trials. Diabetes Res Clin Pract. 2017;130:53-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 74]  [Article Influence: 10.6]  [Reference Citation Analysis (0)]
101.  Fadini GP, Bonora BM, Avogaro A. SGLT2 inhibitors and diabetic ketoacidosis: data from the FDA Adverse Event Reporting System. Diabetologia. 2017;60:1385-1389.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 149]  [Cited by in F6Publishing: 150]  [Article Influence: 21.4]  [Reference Citation Analysis (1)]
102.  Fralick M, Schneeweiss S, Patorno E. Risk of Diabetic Ketoacidosis after Initiation of an SGLT2 Inhibitor. N Engl J Med. 2017;376:2300-2302.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 213]  [Cited by in F6Publishing: 199]  [Article Influence: 28.4]  [Reference Citation Analysis (0)]
103.  Szalat A, Perlman A, Muszkat M, Khamaisi M, Abassi Z, Heyman SN. Can SGLT2 Inhibitors Cause Acute Renal Failure? Plausible Role for Altered Glomerular Hemodynamics and Medullary Hypoxia. Drug Saf. 2018;41:239-252.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 59]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
104.  Herrington WG, Staplin N, Wanner C, Green JB, Hauske SJ, Emberson JR, Preiss D, Judge P, Mayne KJ, Ng SYA, Sammons E, Zhu D, Hill M, Stevens W, Wallendszus K, Brenner S, Cheung AK, Liu ZH, Li J, Hooi LS, Liu W, Kadowaki T, Nangaku M, Levin A, Cherney D, Maggioni AP, Pontremoli R, Deo R, Goto S, Rossello X, Tuttle KR, Steubl D, Petrini M, Massey D, Eilbracht J, Brueckmann M, Landray MJ, Baigent C, Haynes R; The EMPA-KIDNEY Collaborative Group. Empagliflozin in Patients with Chronic Kidney Disease. N Engl J Med. 2023;388:117-127.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 636]  [Article Influence: 636.0]  [Reference Citation Analysis (2)]
105.  Islam L, Jose D, Alkhalifah M, Blaibel D, Chandrabalan V, Pappachan JM. Comparative efficacy of sodium glucose cotransporter-2 inhibitors in the management of type 2 diabetes mellitus: A real-world experience. World J Diabetes. 2024;15:463-474.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
106.  Nadkarni GN, Ferrandino R, Chang A, Surapaneni A, Chauhan K, Poojary P, Saha A, Ferket B, Grams ME, Coca SG. Acute Kidney Injury in Patients on SGLT2 Inhibitors: A Propensity-Matched Analysis. Diabetes Care. 2017;40:1479-1485.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 112]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
107.  Blevins TC, Farooki A. Bone effects of canagliflozin, a sodium glucose co-transporter 2 inhibitor, in patients with type 2 diabetes mellitus. Postgrad Med. 2017;129:159-168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 25]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
108.  The Lancet Diabetes Endocrinology. Dying for a good night's sleep. Lancet Diabetes Endocrinol. 2015;3:1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
109.  Tang HL, Li DD, Zhang JJ, Hsu YH, Wang TS, Zhai SD, Song YQ. Lack of evidence for a harmful effect of sodium-glucose co-transporter 2 (SGLT2) inhibitors on fracture risk among type 2 diabetes patients: a network and cumulative meta-analysis of randomized controlled trials. Diabetes Obes Metab. 2016;18:1199-1206.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 120]  [Cited by in F6Publishing: 119]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
110.  Ruanpeng D, Ungprasert P, Sangtian J, Harindhanavudhi T. Sodium-glucose cotransporter 2 (SGLT2) inhibitors and fracture risk in patients with type 2 diabetes mellitus: A meta-analysis. Diabetes Metab Res Rev. 2017;33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 90]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
111.  Azharuddin M, Adil M, Ghosh P, Sharma M. Sodium-glucose cotransporter 2 inhibitors and fracture risk in patients with type 2 diabetes mellitus: A systematic literature review and Bayesian network meta-analysis of randomized controlled trials. Diabetes Res Clin Pract. 2018;146:180-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 27]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
112.  The Lancet Diabetes Endocrinology. Glucose-lowering drugs: balancing risks and benefits. Lancet Diabetes Endocrinol. 2017;5:669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Reference Citation Analysis (0)]
113.  Li D, Yang JY, Wang T, Shen S, Tang H. Risks of diabetic foot syndrome and amputation associated with sodium glucose co-transporter 2 inhibitors: A Meta-analysis of Randomized Controlled Trials. Diabetes Metab. 2018;44:410-414.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 35]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
114.  Anker SD, Butler J. Empagliflozin, calcium, and SGLT1/2 receptor affinity: another piece of the puzzle. ESC Heart Fail. 2018;5:549-551.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 23]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
115.  Zaccardi F, Webb DR, Htike ZZ, Youssef D, Khunti K, Davies MJ. Efficacy and safety of sodium-glucose co-transporter-2 inhibitors in type 2 diabetes mellitus: systematic review and network meta-analysis. Diabetes Obes Metab. 2016;18:783-794.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 282]  [Cited by in F6Publishing: 320]  [Article Influence: 40.0]  [Reference Citation Analysis (0)]
116.  Gregg LP, Navaneethan SD. Are All SGLT2 Inhibitors Created Equal? Clin J Am Soc Nephrol. 2021;16:1309-1311.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1]  [Cited by in F6Publishing: 1]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
117.  Suzuki Y, Kaneko H, Okada A, Itoh H, Matsuoka S, Fujiu K, Michihata N, Jo T, Takeda N, Morita H, Kamiya K, Matsunaga A, Ako J, Node K, Yasunaga H, Komuro I. Comparison of cardiovascular outcomes between SGLT2 inhibitors in diabetes mellitus. Cardiovasc Diabetol. 2022;21:67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 22]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
118.  Lee AS, Twigg SM, Flack JR. Metabolic syndrome in type 1 diabetes and its association with diabetes complications. Diabet Med. 2021;38:e14376.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 17]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
119.  Snaith JR, Holmes-Walker DJ, Greenfield JR. Reducing Type 1 Diabetes Mortality: Role for Adjunctive Therapies? Trends Endocrinol Metab. 2020;31:150-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 19]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
120.  European Medicines Agency  First oral add-on treatment to insulin for of certain patients with type 1 diabetes [media release]. Feb 1, 2019. [cited 21 March 2024]. Available from: https://www.ema.europa.eu/en/news/first-oral-add-treatment-insulin-treatment-certain-patients-type-1-diabetes.  [PubMed]  [DOI]  [Cited in This Article: ]
121.  Ault A  FDA panel rejects empagliflozin for use in type 1 diabetes. Nov 14, 2019. [cited 21 March 2024]. Available from: http://www.medscape.com/viewarticle/921303.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  European Medicines Agency  Forxiga (dapagliflozin 5 mg should no longer be used for the treatment of type 1 diabetes mellitus. [cited 21 March 2024]. Available from: https://www.ema.europa.eu/en/medicines/dhpc/forxiga-dapagliflozin-5mg-should-no-longer-be-used-treatment-type-1-diabetes-mellitus.  [PubMed]  [DOI]  [Cited in This Article: ]
123.  Seufert J, Lanzinger S, Danne T, Bramlage P, Schmid SM, Kopp F, Kress S, Fasching P, Schäfer C, Holl RW; DPV Initiative. Real-world data of 12-month adjunct sodium-glucose co-transporter-2 inhibitor treatment in type 1 diabetes from the German/Austrian DPV registry: Improved HbA1c without diabetic ketoacidosis. Diabetes Obes Metab. 2022;24:742-746.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 13]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]