Abstract
Despite current established therapy, heart failure (HF) remains a leading cause of hospitalization and mortality worldwide. Novel therapeutic targets are therefore needed to improve the prognosis of patients with HF. The EMPA-REG OUTCOME trial demonstrated significant reductions in mortality and HF hospitalization risk in patients with type 2 diabetes (T2D) and cardiovascular disease with the antihyperglycemic agent, empagliflozin – a sodium glucose co-transporter 2 (SGLT2) inhibitor. The CANVAS trial subsequently reported a reduction in 3-point MACE (major adverse cardiovascular events) and HF hospitalization risk. While SGLT2 inhibition may have potential application beyond T2D, including HF, the mechanisms responsible for the cardioprotective effects of SGLT2 inhibitors remain incompletely understood.
SGLT2 inhibition promotes natriuresis and osmotic diuresis, leading to plasma volume contraction and reduced preload, as well as decreases in blood pressure, arterial stiffness and afterload, thereby improving subendocardial blood flow in patients with HF. SGLT2 inhibition is also associated with preservation of renal function. Based on data from mechanistic studies and clinical trials, large clinical trials with SGLT2 inhibitors are now investigating the potential use of SGLT2 inhibition in patients with HF with and without T2D. Accordingly, in this review, we summarize key pharmacodynamic effects of SGLT2 inhibitors and the clinical evidence which support the rationale for the use of SGLT2 inhibitors in HF patients with T2D. Since presumably these favorable effects occur independent of blood-glucose lowering, we also explore the potential use of SGLT2 inhibition in patients without T2D with HF or at risk of HF, such as in patients with coronary artery disease or hypertension. Finally, we provide a detailed overview and summary of ongoing cardiovascular outcome trials with SGLT2 inhibitors.
Keywords: heart failure, SGLT2 inhibitors, clinical trials review
Introduction
Worldwide, 1-2% of the general adult population have heart failure (HF), which is accompanied by reduced quality of life, high morbidity, mortality and significant financial costs1. Existing therapies such as renin angiotensin aldosterone system (RAAS) inhibition, beta-blockade and angiotensin receptor blockers/neprilysin inhibitors (ARNI) reduce hospitalization and mortality risk in patients with HF and reduced ejection fraction (“HFrEF”)2. Despite important cardiovascular benefits with the use of these agents, patients still have an increased risk for morbidity and mortality. These therapies also carry the potential for serious adverse effects including hypotension, kidney dysfunction and electrolyte abnormalities2. Identification of novel therapeutic strategies to improve symptoms, reduce mortality, recurrent hospitalization, and acute decompensation is therefore critical to advance outcomes in HF patients.
Type 2 diabetes (T2D) is among the many comorbidities associated with cardiovascular disease (CVD) that contributes to end organ damage, and T2D also intensifies the risk for developing HF3 and HF-related complications, including death4. These risks are further compounded in the presence of diabetic nephropathy, highlighting an important interaction between T2D, chronic kidney disease (CKD) and HF5. The aims of this review are to summarize experimental and clinical evidence, which support the rationale for the use of antihyperglycemic sodium-glucose cotransport-2 (SGLT2) inhibitor agents in patients with T2D and HF, and to critically appraise whether SGLT2 inhibition may also be applicable in patients with HF without T2D.
HF Current Therapeutic Strategies and Unmet Needs
The two major types of HF, broadly categorized based on systolic function are: (1) HFrEF, left ventricular ejection fraction (LVEF) <40%, and (2) HF with preserved ejection fraction (HFpEF), LVEF ≥40%. Mechanistically, HF is accompanied by activation of several key neurohormonal regulatory systems, including the sympathetic nervous system (SNS), the renin-angiotensin-aldosterone system (RAAS), and by dysfunction of endogenous natriuretic mechanisms6. Activation of these compensatory pathways initially maintains blood pressure and preserves renal function. Over time, however, chronic neurohormonal activation increases left ventricular afterload and promotes vascular and cardiac remodeling and HF disease progression.
Current standards of care for HF patients with HFrEF include β-blockers, RAAS inhibitors, angiotensin receptor-neprilysin inhibitor (ARNI), diuretics and digoxin to suppress neurohormones, reduce volume overload and improve cardiac contractility6, 7. Conventional diuretics only provide symptomatic relief for HF patients, but do not impact mortality (Figure 1). In contrast to HFrEF, current HF therapies fail to improve outcomes in patients with HFpEF6. Accordingly, HFpEF management focuses on the treatment of co-morbidities such as T2D, hypertension, coronary artery disease, and obesity8. Despite overall clinical benefits, currently available HF therapies, such as RAAS inhibitors and diuretics, increase the risk of adverse effects due to hypotension, volume depletion and SNS activation, highlighting the urgent need for safe, novel therapies9,10, 11
Figure 1. Diuretic agents, their mechanisms of action and potential impact in patients with HF.
HF = heart failure; SGLT2i = sodium glucose co-transporter 2 inhibitor; PV = pressure volume; CV = cardiovascular; HHF = hospitalization for heart failure; LV = left ventricular.
Anti-hyperglycemic Agents and Cardiovascular Safety Trials
New insights into HF management have emerged somewhat unexpectedly from trials examining antihyperglycemic agents used in the treatment of T2D. While metformin and insulin may not impact HF progression, thiazolidinediones are associated with an increased risk of edema and HF12. These findings led to the requirement by regulatory agencies for cardiovascular safety studies for all new antihyperglycemic agents. The dipeptidyl-peptidase 4 inhibitors (DPP-4i), saxagliptin and alogliptin but not sitagliptin, have also been associated with increased risk for hospitalization of HF by the Food and Drug Administration (FDA) and European Medicines Agency (EMA); responsible mechanisms are, however, not currently known13, 14.
In contrast, liraglutide, a GLP-1 receptor agonist (GLP-1RA), was shown to reduce cardiovascular events by 13%, and also reduced all-cause mortality and nephropathy events in the Liraglutide Effect and Action in Diabetes: Evaluation of Cardiovascular Outcome Results (LEADER) trial, a cardiovascular safety study in patients with T2D at heightened CV (cardiovascular) risk15 (Figure 2). Importantly, once-daily liraglutide administration did not increase the risk of hospitalization for HF compared to placebo including in the subgroup of patients with pre-existing HF after 3.8 years follow-up15. The cardioprotective effects associated with liraglutide emerged at 12-18 months in the context of only modest blood pressure and glucose lowering effects, suggesting that clinical benefits were derived through non-hemodynamic, non-glycemic anti-atherosclerotic mechanisms. Favorable effects, including a reduction in 3-point MACE (26%), were also reported in the SUSTAIN-6 trial with the investigational GLP-1RA agent, semaglutide – effects driven primarily by a reduction in the risk for stroke, also over approximately 12-18 months16, 17. Consistent with observations from the LEADER trial, semaglutide did not significantly modify the risk of hospitalization for HF16, 17. Importantly, the results of LEADER and SUSTAIN-6, which were obtained predominantly in T2D patients without HF, have been mirrored in real-world studies and meta-analyses examining GLP-1RA18, 19. Unfortunately, in recently hospitalized patients with advanced HF and reduced LVEF (median 25%), dedicated studies with liraglutide were associated with overall neutral effects on cardiovascular death (at 180 days) and re-hospitalization rates compared to placebo. These data suggest that GLP1-RA agents are safe in HF20–24 and reduce risk factors for CVD (blood pressure, body weight), particularly among T2D patients, but are unlikely to have direct beneficial effects on myocardial function in HF patients with advanced disease despite the expression of GLP-1 receptors in the heart.
Figure 2.
Hazard ratio and 95% confidence intervals for outcomes examined in EMPA-REG Outcome, CANVAS, LEADER and SUSTAIN 6. CV = cardiovascular; CI = confidence interval112
In contrast to LEADER and SUSTAIN-6, SGLT2 inhibition with empagliflozin reduced the risk of hospitalization for HF in the EMPA-REG OUTCOME trial25, 26. This CV safety trial randomly assigned patients with T2D and established CVD (approximately 10% with established HF) to 10 or 25 mg of empagliflozin once daily versus placebo in addition to standard of care. Compared to placebo, participants treated with empagliflozin, pooled from both doses, had a 14% reduction in 3-point MACE (CV death, non-fatal myocardial infarction, non-fatal stroke). The 35% reduction in the risk of hospitalization for HF emerged within several months, suggesting a role for acute effects on renal and/or systemic hemodynamic function or direct effects on the cardiovascular system (Figure 3, “Mechanism of Action” section below). Benefits on 3-point MACE were similar across eGFR subgroups down to an eGFR of 30 ml/min/1.73m2, despite the fact that SGLT2 inhibitors are associated with less glucosuria as renal function declines. EMPA-REG OUTCOME also reported a 39% reduction in the composite renal endpoint, new onset or worsening nephropathy (defined as incident macroalbuminuria, doubling of serum creatinine, eGFR <45 ml/min/1.73m2, initiation of renal replacement therapy or a renal disease related death)26.
Figure 3. Integrated Physiological Basis for Proposed Mechanisms Leading to Cardiorenal Benefits and Risks Associated with Sodium Glucose Cotransport-2 Inhibition.
* = only reported in the CANVAS Program; RAGE = receptor for advanced glycation end products; PTH = parathyroid hormone; FGF-23 = fibroblast growth factor 23; Na+ = sodium; AKI = acute kidney injury.
The results of two additional SGLT2 inhibitor CV safety trials – CANagliflozin cardioVascular Assessment Study (CANVAS) and CANagliflozin cardioVascular Assessment Study-Renal (CANVAS-R)27 have also been reported. In CANVAS, 4,330 patients with T2D at high CV risk were randomly assigned 1:1:1 to 100, 300 mg/day canagliflozin or to placebo for 2 years to evalute 3-point MACE (Clinicaltrials.gov NCT01032629)28. In CANVAS-R, the effect of canagliflozin for 2.5 years was evaluated in 5,700 patients with T2D at elevated CV risk, with the primary outcome of kidney disease progresion (Clinicaltrials.gov NCT01989754)29. The secondary outcomes of CANVAS and CANVAS-R have been modified to prioritize vascular death, total mortality and HF and to refine the outcomes addressing myocardial infarction and stroke30. Rather than evaluating separate effects of each dose of canagliflozin, the results of both studies and doses of canagliflozin were combined and reported as the CANVAS Program, with an overall HF prevalence of 14.4%. In the CANVAS Program with canagliflozin (n=10,142 patients with T2D), the primary endpoint, 3-point MACE, was significantly reduced, and the risk of hospitalization for HF was reduced by 33%31. From a renal perspective, progression of albuminuria was reduced by 27%, and the composite outcome (sustained 40% reduction in eGFR, the need for renal-replacement therapy, or death from renal causes) was reduced by 40%31.
The factors responsible for the acute beneficial effects on hospitalization for HF in EMPA-REG OUTCOME and the CANVAS Program are incompletely understood. The majority of patients (approximately 90%) in the EMPA-REG OUTCOME and CANVAS Program trials did not have HF at baseline, suggesting that the HF beneficial effects observed were due to a primary prevention effect. Importantly, SGLT2 inhibitors are associated with favorable effects on many of the comorbidities associated with HFpEF, including diabetes-related hyperglycemia, obesity and hypertension. Together with other potential protective mechanisms discussed in the next section,32 these effects may also reduce HF risk and/or CV death in HF patients without T2D, a demographic currently being evaluated in ongoing CV outcome trials (Table 1).
Table 1.
Clinical Trials with SGLT2i
Name of Clinical Trial |
Type | Treatment Arms |
Duration | Sample size | Diabetes Status |
Primary Outcomes/Results |
Secondary Outcomes |
NCT | Estimated Reporting |
---|---|---|---|---|---|---|---|---|---|
Cardiovascular Outcome Trials | |||||||||
Empagliflozin Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients (EMPA-REG OUTCOME) | Double blind, placebo controlled RCT (Phase 3) | Empagliflozin 10 mg or 25 mg daily vs. placebo | Up to 4.6 years | 7,020 patients with established CV complications (≥18 years) | All T2D | 14% reduction in 3-point MACE (CV death, non-fatal MI, non-fatal stroke) pooled from 10mg and 25mg empagliflozin doses | 35% reduction in hospitalization for HF, 39% reduction in the composite renal endpoint (new macroalbuminuria, doubling of serum creatinine and GFR ≤45, renal replacement therapy, renal death) | NCT01131676 | Reported in 2015 |
Canagliflozin Cardiovascular Assessment Study (CANVAS Program) | Double-blind, placebo controlled RCT (Phase 3) | Canagliflozin 100 mg or 300 mg daily vs. placebo | 3.6 years | 10,142 patients with established vascular complications or ≥2 CV risk factors (>30 years) | All T2D | 14% reduction in 3-point MACE (CV death, non-fatal MI, non-fatal stroke) | 27% reduction in progression of albuminuria, 70$ increase in regression of albuminuria, 40% reduction in the composite renal endpoint (40% reduction in eGFR, renal replacement therapy, renal death) | NCT01032629 | Reported in 2017 |
Evaluation of the Effects of Canagliflozin on Renal and Cardiovascular Outcomes in Participants with Diabetic Nephropathy (CREDENCE) | Double-blind, placebo controlled RCT (Phase 3) | Canagliflozin 100 mg daily vs. placebo | 4 years | 3,627 patients with Stage 2 or 3 CKD and macroalbuminuria and on ACEi/ARB (>30 years) | All T2D | ESKD, S-creatinine doubling, renal/CV death | CV death, non-fatal MI, non-fatal stroke, hospitalized UAP, hospitalized CHF, composite renal endpoint (ESKD, doubling of serum Cr. Renal death) | NCT02065791 | Anticipated 2019 |
Multicenter Trial to Evaluate the Effect of Dapagliflozin on the Incidence of Cardiovascular Events (DECLARE-TIMI 58) | Double-blind, placebo controlled RCT (Phase 3) | Dapagliflozin 10 mg vs. placebo | Up to 6 years | 17,276 patients with high risk for CV events (≥40 years) | All T2D | CV death, non-fatal MI, non-fatal ischemic stroke; CV death, hospitalization due to HF | Renal composite endpoint (≥40% decrease in eGFR to <60 and/or ESRD and/or renal or CV death, all-cause mortality | NCT01730534 | Anticipated 2019 |
Cardiovascular Outcomes Following Ertugliflozin Treatment in Type 2 Diabetes Mellitus Participants With Vascular Disease (VERTIS) | Double-blind, placebo controlled RCT (Phase 3) | Ertugliflozin vs. placebo | Up to 6.1 years | 8,000 patients with established vascular complications (≥40 years) | All T2D | CV death, non-fatal MI, non-fatal stroke | CV death, non-fatal MI, non-fatal stroke and hospitalized UAP | NCT01986881 | Anticipated 2019 |
Large Randomized Controlled Clinical Trials in Patients with Heart Failure | |||||||||
Dapagliflozin Effect on Symptoms and Biomarkers in Diabetes Patients With Heart Failure (DEFINE-HF) | Double blind, placebo controlled RCT (Phase 4) | Dapagliflozin 10 mg daily vs. placebo | 12 weeks | 250 patients with HF (≥19 years) | All T2D | Change in NTproBNP | Change in SBP, weight, HbA1c, BNP and QoL score by questionnaire | NCT02653482 | TBD |
Dapagliflozin in Type 2 Diabetes or Pre-diabetes, and Preserved Ejection Fraction Heart Failure (PRESERVED HF) | Double blind, placebo controlled RCT (Phase 4) | Dapagliflozin 10 mg daily vs. placebo | 12 weeks | 320 patients with HF (≥19 years) | T2D or pre-diabetes | Change in NTproBNP | Change in SBP, weight, HbA1c, BNP and QoL score by questionnaire | NCT03030235 | TBD |
Study to Evaluate the Effect of Dapagliflozin on Incidence of Worsening Heart failure or Cardiovascular Death in Patients with CHF (DAPA-HF) | Double blind, placebo controlled RCT (Phase 3) | Dapagliflozin 10 mg daily vs. placebo | 36 months | 4,500 patients with HFrEF (≥18 years) | Non-diabetic and T2D (T1D excluded) | Time to CV death or hospitalization for HF or an urgent HF visit. | Time to ≥50% sustained decline in eGFR or ESRD. QoL score by questionnaire. Time to death by any cause. | NCT03036124 | TBD |
EMPagliflozin outcomE tRial in Patients With chrOnic heaRt Failure With Preserved Ejection Fraction (EMPEROR-Preserved) | Double blind, placebo controlled RCT (Phase 3) | Empagliflozin 10 mg daily vs. placebo | 38 months | 4,126 patients with HFpEF (≥18 years) | Non-diabetic, T1D and T2D eligible | Time to first event of adjudicated CV death or adjudicated hospitalization for HF. | Change in eGFR. Time to sustained reduction in eGFR. Time to all-cause mortality. Time to DM. | NCT03057951 | TBD |
EMPagliflozin outcomE tRial in Patients With chrOnic heaRt Failure With Reduced Ejection Fraction (EMPEROR-Reduced) | Double blind, placebo controlled RCT (Phase 3) | Empagliflozin 10 mg daily vs. placebo | 38 months | 2,850 patients with HFrEF (≥18 years) | Non-diabetic, T1D and T2D eligible | Time to first event of adjudicated CV death or adjudicated hospitalization for HF. | Change in eGFR. Time to sustained reduction in eGFR. Time to all-cause mortality. Time to DM. | NCT03057977 | TBD |
Mechanistic Clinical Trials in Patients with Heart Failure | |||||||||
Empagliflozin Impact on Hemodynamics in Patients With Diabetes and Heart Failure (EMBRACE-F) | Double blind, placebo controlled RCT (Phase 4) | Empagliflozin 10mg daily vs. placebo | 12 weeks | 60 patients with HF (≥19 years) | All T2D | Change in pulmonary artery systolic and diastolic pressure. | Change in mean pulmonary artery pressure. Change in QoL score by questionnaire. | NCT03030222 | TBD |
Study and Effectiveness of SGLT-2 Inhibitors in Patients with Heart Failure and Diabetes (REFORM) | Double blind, placebo controlled RCT (Phase 4) | Dapagliflozin 10 mg daily vs. placebo | 12 months | 56 patients with HF (18-75 years) | All T2D | Change in LV ESV or EDV (primary) by cardiac MRI | Change in LVEF, LVMi, fluid status by BIA and QoL score by questionnaire | NCT02397421 | TBD |
Empagliflozin in Heart Failure: Diuretic and Cardio-Renal Effects (EMPA) | Double blind, placebo controlled RCT (Phase 2) | Empagliflozin 10mg daily vs. placebo | 14 days | 50 patients with stable HF (≥18 years) | All T2D | Changes in urine sodium concentrations via ion selective electrodes. | Changes in blood volume by radiolabeled albumin. | NCT03027960 | TBD |
SGLT2 Inhibition in Diabetes and Heart Failure | Prospective cohort study | SGLT2i | 4 weeks | 31 patients with HF (≥18 years) | All T2D | Changes in peak oxygen consumption (VO2)(mL/kg/min) and minute ventilation (VE)/carbon dioxide production (VCO2) slop | Not specified | NCT02862067 | TBD |
HF = heart failure; SGLT2i sodium-glucose co-transporter inhibitor; RCT = randomized controlled trial, T2D = type 2 diabetes, QoL = quality of life, LV = left ventricular, ESV = end-systolic volume, EDV = end-diastolic volume, LVMi = left ventricular mass indexed, EF = ejection fraction, CV = cardiovascular, UAP = unstable angina pectoris, CHF = congestive heart failure, ESKD = end-stage kidney disease; TBD = to be determined
The results of the EMPA-REG OUTCOME trial have translated into recognition of empagliflozin as a CV protective therapy for patients with T2D by the Food and Drug Administration (FDA), the American College of Cardiology/American Heart Association, Diabetes Canada, and the European Society of Cardiology; similar perspectives may soon emerge for canagliflozin based on the CANVAS Program. Consistent with the observations in EMPA-REG OUTCOME and CANVAS, the broad impact of SGLT2 inhibition on CV outcomes has been observed in several “real-world” studies such as CVD-REAL, which reported significant benefits on mortality and hospitalization for HF risk with several SGLT2 inhibitor agents in an analysis of administrative data from 6 countries33. Separate analyses examining dapagliflozin vs. other glucose lowering therapies revealed similar beneficial CV effects34, 35, and an additional analysis demonstrated a 56% decreased risk of all-cause mortality and CVD with SGLT2 inhibition in a general practice cohort in the Sweden36.
Mechanisms of Action of SGLT2 Inhibitors and Relationships with CV Outcomes (Figure 1)
High capacity, low-affinity SGLT2 are located in the renal proximal tubular epithelium and reabsorb filtered glucose37, 38. SGLT2 inhibition-related glycosuria results in an insulin-independent HbA1c reduction of approximately 0.7-1.0% in patients with T2D, and also a body weight loss (~2 to 3 kg) due to induction of a negative caloric balance in patents with normal renal function39 (Figure 3). SGLT2 inhibition-induced glycosuria also mediates a uricosuric effect40–42 via the GLUT9 transporter40, which may be cardioprotective, as increased plasma uric acid levels are associated with CV complications43 and congestive HF44. Despite being safely used in T2D patients with CKD in clinical trials, canagliflozin dosing reductions are required for patients with eGFR of <60 and >45 ml/min/1.73m2. Furthermore, empagliflozin and canagliflozin are contraindicated in patients with eGFR of <45 ml/min/1.73m2 and dapagliflozin is contraindicated in patients with eGFR of <60 ml/min/1.73m2 due to reduced glycemic efficacy. Therefore, based on these glucose-centric GFR cut-offs, SGLT2 inhibitor use in HF patients with co-existing CKD may be “off-label”. Based on available evidence, it is unlikely that SGLT2 inhibitor-mediated reductions in death, HF or nephropathy events are mediated by HbA1c lowering, since HbA1c was only marginally decreased by <0.4% according to the trial design. A number of trials with other antihyperglycemic agents, perhaps with the exception of LEADER, have failed to show a positive impact of improved glycemic control on cardiorenal endpoints of magnitude seen in EMPA-REG OUTCOME or CANVAS Program12. Similarly, glucosuria-associated weight loss was also unlikely to have accounted for the CV benefits with SGLT2 inhibition. While <3.5% decreases in total body weight was observed in EMPA-REG OUTCOME and CANVAS Program, trials such as LOOK AHEAD had no significant effects on lowering CV events over 10 years follow-up in overweight adults with T2D despite an average 6% loss of body weight45.
Aside from effects on glycemic control and weight loss, SGLT2 are responsible for ~5% of sodium reabsorption at the proximal tubule under normal conditions. In the setting of chronic hyperglycemia, the capacity of SGLT2 in the kidney is increased leading to more pronounced effects on sodium homeostasis which decreases plasma volume and blood pressure with SGLT2 inhibition, as reviewed in more detail elsewhere39, 46–48. Importantly for HF, SGLT2 inhibition-mediated effects on effective circulating volume contraction could reduce preload, thereby lowering ventricular filling pressure. Overall, the effects of SGLT2 inhibition on natriuresis and associated plasma volume contraction would be expected to reduce cardiac preload, while afterload reductions may occur through blood pressure and arterial stiffness lowering, thereby improving subendocardial blood flow.
In the kidney, SGLT2 inhibition-mediated proximal tubular natriuresis increases sodium delivery to the macula densa, which activates tubuloglomerular feedback, afferent vasoconstriction and reduces glomerular pressure, as reviewed elsewhere41, 49, 50. These intrarenal hemodynamic effects may account for the 30-40% reduction in albuminuria observed with SGLT2 inhibitor agents including empagliflozin, dapagliflozin and canagliflozin25. The hemodynamic effects of SGLT2 inhibition occur even in the presence of renal dysfunction, despite attenuated HbA1c lowering effects in this setting51. Preservation of GFR might be of particular importance in HF patients in order to avoid volume overload and complications arising from diuretic resistance (Figure 3). Therefore, natriuresis mediated by SGLT2 inhibition is likely a major factor leading to cardiovascular and renoprotective effects observed with empagliflozin and canagliflozin, which appears to extend across CKD stages52. Whether other SGLT2 inhibitors will also demonstrate cardiac protection remains to be determined in large prospective CV safety studies (Table 1)53.
Despite the clinical relevance of maintaining sodium and water homeostasis in HF patients, reliable measurements of changes in total body sodium in humans remains difficult due to the potential for multiple sodium reservoirs throughout the body. Recent studies suggest that tissues such as skin and muscle store sodium, and that sodium accumulation is associated with CV disease (CVD)54. In fact, the concentration of sodium in skin measured by 23sodium-magnetic resonance imaging (23Na-MRI) correlates strongly with left ventricular mass and blood pressure in CKD, suggesting that it is a surrogate marker for volume expanded states55. SGLT2 inhibition with dapagliflozin for 6 weeks significantly reduces skin sodium concentration measured by 23Na-MRI in 59 patients with T2D56 suggesting that SGLT2 inhibition decreases both plasma volume and total body sodium content, which may protect against volume expansion and the risk of being hospitalized for HF.
Treatment of T2D patients with SGLT2 inhibitors typically reduces SBP by 4-6 mmHg and DBP by 1-2 mmHg46, 47. In response to SGLT2 inhibition, similar magnitudes in SBP reduction (~2.7 mmHg) have also been observed in healthy individuals without T2D57–61. Apart from natriuresis and osmotic diuresis, other mechanisms that might contribute to blood pressure lowering in response to SGLT2 inhibition include a decrease in arterial stiffness62, and effects on endothelial function or vascular architecture63. Improvements in arterial stiffness with SGLT2 inhibition has been partly attributed to effective circulating volume contraction via mild diuretic effects and vascular smooth muscle relaxation64. Despite these antihypertensive effects, the risks of myocardial infarction, hospitalization for unstable angina, or stroke were not reduced in the EMPA-REG OUTCOME or CANVAS Program trials. This lack of effect on atherosclerosis-related endpoints argues against a major role for blood pressure lowering alone leading to CV benefits. Moreover, previous meta-analyses using other agents have suggested that modest blood pressure lowering effects in EMPA-REG OUTCOME and the CANVAS Program were probably insufficient to account for the large impact on CV or renal endpoints65. Therefore, based on available evidence from these clinical trials, it seems highly unlikely that the reduction in CV risk with empagliflozin or canagliflozin can be attributed to glycemic control, blood pressure, or body weight reduction suggesting the involvement of other SGLT2 inhibition-mediated mechanisms.
It is unclear whether SGLT2 inhibitors have direct effects on cardiomyocytes, since SGLT2 is not expressed in cardiac tissue in humans66. Recent preclinical studies in rat ventricular myocytes treated with empagliflozin however, demonstrated direct SGLT2 inhibition-mediated cardiac effects by lowering myocardial intracellular sodium concentrations via inhibition of the myocardial Na+/H+ exchanger flux with a secondary decrease in intracellular calcium and an increase in mitochondrial calcium67. Regulation of mitochondrial calcium concentrations by SGLT2 inhibition is of interest, because mitochondrial calcium is an activator of ATP synthesis and of antioxidant enzymatic pathways68, 69. In fact, increasing mitochondrial calcium concentrations prevents sudden death and overt HF in porcine models68. Additionally, increases in myocardial intracellular sodium and calcium concentrations are early hallmarks and drivers of cardiovascular death and HF70, 71 (Figure 3). Further cardioprotective effects associated with SGLT2 inhibition may be derived through attenuating the pro-thrombotic milieu associated with hyperglycemia, which may be achieved through dapagliflozin-mediated effects on neutrophil-derived S100 calcium-binding proteins A8/A9 (S100A8/A9), which interact with the receptor for advanced glycation end products (RAGE)72. In light of the rapid clinical benefits observed in EMPA-REG OUTCOME and the CANVAS Program in particular, and animal models demonstrating protective effects on left ventricular mass and left ventricular end diastolic diameter73, SGLT2 inhibition may have protective effects on cardiomyocytes or on inflamamtory pathways in the CV system – an intriguing possibility that merits further investigation.
Beyond effects on traditional CV risk factors or mitochondrial cardiomyocyte pathways, is the possibility that the reduction in HF risk in EMPA-REG OUTCOME and in the CANVAS Program were related to metabolic substrate shifts from conventional fatty acids to ketone bodies. SGLT2 inhibition is associated with increased glucagon (possibly via direct pancreatic α-cell effects) and reduced insulin concentrations, thereby promoting the production of ketones such as β-hydroxybutyrate74. Ketones are freely taken up by the heart and can act as an efficient substrate for myocardial energy generation, as discussed elsewhere75–77. This metabolic substrate shift leads to improved energy utilization, potentially contributing to improved cardiac efficiency, contractility and CV protection. In conjunction with increases in hematocrit leading to improved oxygen delivery to the heart78, SGLT2 inhibition may both increase oxygen supply to the heart, and reduce cardiac oxygen demand, thereby improving cardiac function.
Given the acute nature of the HF benefit in EMPA-REG OUTCOME25, discussion around CV benefits have generally focused on either volume/hemodynamic pathways or on changes in energy utilization in the heart. However, it is important to recognize and highlight the possibility that both of these pathways may have protective and injurious potential. For example, natriuresis may be beneficial for reasons discussed above, but may also increase the risk of volume depletion and acute kidney injury, and promote neurohormonal activation. In EMPA-REG OUTCOME, empagliflozin was well tolerated hemodynamically and decreased the risk of AKI, possibly due to reduced loop diuretic use, which could help preserve renal function by the avoidance of volume depletion79. In the CANVAS Program, canagliflozin also did not increase the risk of AKI. Fortunately, SGLT2 inhibition-mediated effects on the RAAS appear to be modest80–82 and effects on symapthetic activity are neutral in patients without established HF83.
Despite empirical evidence showing HF benefits in the EMPA-REG OUTCOME and CANVAS Program trials, volume-related effects of SGLT2 inhibition in patients with HF are not yet fully understood. For example, in a pilot study involving 20 Japaneses patients with T2D and HF, the addition of ipragliflozin to conventional loop or thiazide diuretics for 4 days decreased plasma natriuretic peptide levels without influencing plasma angiotensin II, aldosterone or noradrenaline levels84. It is not known whether participants in EMPA-REG OUTCOME had HFrEF or HFpEF, since echocardiography was not performed at baseline. In a subsequent report, the EMPA-REG OUTCOME investigators explored HF outcomes from this trial and concluded that a decrease in HF hospitalization and CV death was consistent among patients with and without HF at baseline. Empagliflozin may therefore prevent HF decompensation as well as incident HF in predisposed individuals79. The time course for the reduction in HF hospitalization risk with canaglifozin has not yet been reported in the CANVAS Program.
Impact of SGLT2 Inhibition on Cardiac Structure and Function
Until ongoing small mechanistic studies and clinical trials are completed (Table 1), the relative role(s) of SGLT2 inhibition on HF pathophysiology described above remains speculative. There are limited data evaluating whether SGLT2 inhibitors modify cardiac structure and function. In animals, SGLT2 inhibition reduces cardiac fibrosis, inflammation, and oxidative stress85. In several rodent models of T2D, SGLT2 inhibition improves diastolic function and left ventricular hypertrophy while simultaneously reducing myocardial fibrosis and expression of profibrotic/prohypertrophic proteins86–88. Verma et al. have also demonstrated cardioprotective effects in a zebrafish model of HF, suggesting conservation of cardiovascular protective effects across species89.
In humans, an analysis of 10 patients with T2D (baseline HbA1c 7.3%) and established CVD who underwent transthoracic echocardiograms before and after three months of treatment with empagliflozin was recently reported90. The authors observed a significant reduction in left ventricular mass index, an established surrogate marker of CV risk and improved diastolic function as determined by early lateral annular tissue Doppler velocity. Considering SGLT2 are not expressed in the human myocardium91, it is difficult to reconcile whether and how SGLT2 inhibition could have direct effects on ventricular remodelling. It is possible these SGLT2 inhibition-mediated effects on ventricular remodelling occur secondary to indirect effects on sodium and calcium entry into cardiomyocyte cellular compartments or via unknown effects in the heart. Further mechanistic studies are needed to better interpret the exsiting clinical data and to better define the effects of SGLT2 inhibition in cardiovascular tissue.
SGLT2 Inhibition and Potential Side Effects
While SGLT2 inhibition has emerged as a promising new cardio-renal protective therapy, it is important to recognize potential adverse effects related to drugs in this class, including increased rates of genitourinary infections, postural hypotension, polyuria, diabetic ketoacidosis, acute kidney injury and possible increased rates of bone fractures (Table 2)31, 39, 92–95. The most commonly reported side effect due to glycosuria is genitourinary infections, mostly genital mycotic infections but in some studies bacterial urinary tract infections as well39, 92–94. Less common but perhaps more important in the HF population in the context of concomitant diuretic use, is the potential for volume depletion. Whether this hemodynamic effect will prove a serious risk for ill and elderly patients with comorbid medical conditions is unclear. Although the EMPA-REG OUTCOME trial did not report a specific approach to adjusting baseline diuretic agents, it reasonable to be cautious when combining SGLT2 inhibitors with other diuretics 96, as suggested elsewhere39, especially in patients with severe systolic dysfunction. While AKI has been reported with SGLT2 inhibition, rates of AKI were less common in empagliflozin versus placebo-treated patients in the EMPA-REG OUTCOME trial, and similar trends were observed in the CANVAS Program26. This is reassuring, since 80% of patients in EMPA-REG OUTCOME were taking concommitant RAAS inhibitors, which also decrease intraglomerular pressure25. From a safety point of view, SGLT2 inhibitors should be part of “sick day” T2D mangement strategies, and may need to be held in the context of NSAID use or radiocontrast administration due to the potential for volume depletion and hemodynamic side effects 96.
Table 2.
Summary of Adverse Effects of Sodium Glucose Co-Transport-2 Inhibitors
Type | Proposed Mechanism of Action | Drug vs. Class | References |
---|---|---|---|
Genitourinary Infections | Increased infection risk in the setting of glycosuria | SGLT2i | 39, 92–94 |
Polyuria/polakyuria | Glycosuria-induced osmotic diuresis, initial increase in natriuresis | SGLT2i | 39, 92–94 |
Postural hypotension | Volume depletion, blood pressure lowering effects through volume independent mechanisms (i.e., glucose lowering, weight loss, reduced arterial stiffness) possibly in the setting of concomitant diuretic, anti-hypertensive use | SGLT2i | 39, 92–94 |
Diabetic ketoacidosis | Ketoacidosis from reduced carbohydrate availability due to increased glycosuria and reduced insulin concentrations. SGLT2 inhibition is also associated with increase levels of counter-regulatory hormones such as glucagon, thereby increasing ketogenesis. | SGLT2i | 105 |
Decrease in eGFR | Decreased intraglomerular pressure due to afferent vasoconstriction, leading to an expected rise in creatinine (acute eGFR “dip” of 4-6 ml/min/1.73m2). Acute kidney injury risk is not increased with SGLT2 inhibition in clinical trials (EMPA-REG OUTCOME, CANVAS Program) but has been reported in post-marketing data. Further work is required to determine the long-term significance of the eGFR dip, and to determine whether or not it is protective (i.e. associated with reduced glomerular hypertension and proteinuria). | SGLT2i | 26 |
Fracture risk |
|
Only reported in the CANVAS Program | 108 |
Amputation risk |
|
Only reported in the CANVAS Program | 31 |
In the CANVAS Program, patients treated with canagliflozin had a significantly higher risk of lower extremity amputations compared to the placebo group31. The increased risk of lower extremity amputations observed in CANVAS with canagliflozin has not been reported with dapagliflozin, empagliflozin, or other SGLT2 inhibitors97. While the potential mechanisms responsible for differences in risk rates between members of this drug class for lower extremity amputations are not known, it may be relevant to recognize that canagliflozin has less SGLT2 selectivity compared to the other currently available SGLT2 inhibitors, and this may as a consequence induce more glucosuria98. While it is not yet known if this translates to greater volume depletion with canagliflozin, a more pronoucned rise in hematocrit and hemoconcentration could conceivably increase blood viscosity and hence tissue ischemia in the periphery. Interestingly, other diuretics that cause hemoconcetration may similarly increase amputation risk, although this has not been carefully studied99. In addition, canagliflozin, but not empagliflozin, impacts mitochondrial AMP-kinase activity, which is involved in mitochondrial energy pathways100. In animals, canagliflozin associated AMP kinase activation was associated with inhibition of Complex I of the respiratory chain, leading to increases in cellular AMP and ADP. While it is not known if these pharmacological effects observed preclinically impact lower extremity perfusion, oxygenation or energy delivery, these findings highlight the concept that SGLT2 inhibitors differ pharmacodynamically and thereby lead to differences in both benefit and adverse effect profiles. As an alternative explanation, data collection or reporting of amputations may have differed between the trials, leading to under-reporting with other agents. Whereas further work is required to determine whether or not increased amputation risk occurs across the SGLT2 inhibitor drug class, a recent administrative data analysis using FAERS reported a consistent increased risk of amputation with canagliflozin only, and no increased risk with either empagliflozin or dapagliflozin101. Whether the risk for amputation will be present in HF is unclear, but should be further explored to determine if hypotension and peripheral hypoperfusion, both characteristic of HF, impact amputation risk further.
Notwithstanding the theoretical benefits around energy utilization described above, increased ketonemia in patients with diabetes taking SGLT2 inhibitiors should raise concern for evolving diabetic ketoacidosis. Although not reported more frequently as an adverse effect in EMPA-REG OUTCOME or the CANVAS Program, the US FDA and EMA have issued advisories about the potential risk of diabetic ketoacidosis with SGLT2 inhibitors 102, 103. Fadini et al. recently published an analysis of the FDA Adverse Event Reporting System (FAERS), which demonstrated a proportional reporting ratio (PRR) of 7.9 (95% confidence interval, 7.5-8.4) for diabetic ketoacidosis in patients using SGLT2 inhibitors compared to those using other agents, with a higher risk in patients with type 1 diabetes104. An increase in the risk of diabetic ketoacidosis was also observed by Fralick et al. in > 76,000 commercially insured T2D patients in the United States taking SGLT2 inhibitors compared to those using DPP4 inhibitors (hazard ratio for diabetic ketoacidosis, 2.2 95% CI 1.4-3.6)95. In contrast, a separate meta-analysis of completed clinical trials in >10,000 T2D patients reported a reduction in risk of diabetic ketoacidosis, while a third administrative database analysis in >150,000 T2D patients reported an overall neutral risk of diabetic ketoacidosis, highlighting that more data are needed to better understand this rare but potentially serious risk105–107.
As a final comment, the CANVAS Program also reported an increased risk for bone fracture – an observation not yet reported with other agents in this drug class. Mechanistically, SGLT2 inhibition may exert adverse effects on bone physiology through increased concentrations of parathyroid hormone and fibroblast growth factor-23108. The impact on bone fracture risk does not appear to exist across this drug class, since a recent meta-analysis by Tang et al failed to broadly demonstrate bone fracture risk with SGLT2 inhibitor use109. Whether bone fracture risk is of specific concern with SGLT2 inhibitors in HF is not known. Nevertheless, these agents should be used with caution in frail individuals who are at risk of falling, including those with hypotension.
Ongoing SGLT2 Inhibitors Cardiovascular Trials
In light of the emerging evidence for CV protetction with SGLT2 inhibition, specifically empagliflozin and canagliflozin, clinical trials are currently underway to evaluate the impact of other SGLT2 inhibitors on primary CV outcomes in patients with T2D53. The “Multicenter Trial to Evaluate the Effect of Dapagliflozin on the Incidence of Cardiovascular Events” (DECLARE –TIMI58) is a Phase IIIb trial that is evaluting the effect of dapagliflozin (10 mg daily) versus placebo over 4.5 years on the composite 3-point MACE primary outcome in patients with T2D (Clinicaltrials.gov NCT01730534)53. This trial is enrolling 17,150 patients with T2D and either known CVD (secondary prevention cohort) or at least two risk factors for CVD (primary prevention cohort). The impact of ertugliflozin on CV and renal endpoints is similarly being examined in the VERTIS study, although results are not anticipated until after 202053.
In addition to these trials, CV endpoints have been included as secondary endpoints in primary renal outcome trials. In the Evaluation of the Effects of Canagliflozin on Renal and Cardiovascular Outcomes in Participants with Diabetic Nephropathy Study (CREDENCE), the objectives are to determine whether canagliflozin versus placebo for 5 years will reduce the primary composite endpoint of the study including end-stage kidney disease (ESKD), doubling of serum creatinine, renal or CV death in patients with T2D with stage 2 or 3 CKD and macroalbuminuria (Clinicaltrials.gov NCT02065791)53. The effects on progression of CKD or CV/renal death of dapagliflozin versus placebo as an addition to standard of care in 4,000 patients with CKD are being examined in a 4 year follow-up Study to Evaluate the Effect of Dapagliflozin on Renal Outcomes and Cardiovascular Mortality in Patients With Chronic Kidney Disease (DAPA-CKD) (Clinicaltrials.gov NCT03036150). Importantly DAPA-CKD is recruiting patients with and without T2D to better define the role of SGLT2 inhibition as a renal protective therapy in the absence of hyperglycemia – perhaps through the natriuresis-related pathways discussed above. A second large CKD prevention trial with empagliflozin was announced on June 11, 2017 that will include patients with and without diabetic kidney disease, although no details are currently available. To date, no human studies have reported renal or cardiovascular effects in the setting of non-diabetic kidney disease. Accordingly, planned and ongoing nephropathy trials with empagliflozin and dapagliflozin in non-diabetic patients are based on the premise that volume-related and other non-glycemic mechanisms will extend to patients without diabetes, since glucose-lowering effects are unlikely to be responsible for end-organ protection110.
In addition to studies such as DAPA-CKD in patients without diabetes that will evaluate primary renal endpoints, several large clinical trials have been announced that will investigate the impact of SGLT2 inhibition in HF patients with and without T2D. These trials include DAPA-HF, which is enrolling HFrEF patients, and the EMPEROR trials, which are including patients with HFrEF and HFpEF (Table 1). In addition, there are a number of ongoing smaller mechanistic studies in patients with T2D in this area that will better define SGLT2 inhibition-mediated mechanisms resulting in clinical HF benefits – including natriuresis, plasma volume contraction and neurohomonal activation, as reviewed in Table 1. As in the case of renal trials including individuals without T2D, the rationale for undertaking HF trials with SGLT2 inhibition in patients who do not have T2D is to take advantage of glucose-independent cardiorenal protective pathways in response to SGLT2 inhibition. Once complete, these ongoing studies will help elucidate the complex pharmacodynamic effects of SGLT2 inhibition on CVD in patients with and without T2D.
Conclusions
SGLT2 inhibition may emerge as an effective and safe adjunctive therapy for HF; that promotes hemodynamic stability and helps correct volume overload, while avoiding the risks of volume depletion, independent of effects on hyperglycemia. Nevertheless, the mechanisms responsible for the acute cardioprotective effects of SGLT2 inhibition, such as sodium and water homeostasis and plasma volume regulation must be examined in diverse populations, including in patients with and without T2D, and in those with HFpEF and HFrEF. Furthermore, the therapeutic landscape will continue to evolve with the use of new agents that impact circulating volume, such endothelin-1A receptor antagonists (sodium retention) and ARNI (natriuresis). The diabetes research community will also need to focus on understanding how these various agents interact in combination – both in terms of blood pressure, albuminuria and cardiorenal benefits, and around the potential for adverse effects111.
In light of EMPA-REG OUTCOME, the CANVAS Program and studies with GLP-1RA agents, the management of patients with T2D and existing CVD is evolving. Since clinicians now have access to effective therapies to prevent cardiorenal events, future trials need to consider whether the demonstration of non-inferiority is sufficient, or whether superiority should be targeted, especially for HF events. In addition, while existing trial evidence has demonstrated unexpectedly robust protection in patients with existing complications, it remains to be determined if these agents will be similarly effective as primary prevention strategies.
Acknowledgments
FUNDING SOURCES:
Y.L. is supported by a Canadian Diabetes Association Fellowship. J.A.L. is supported by Sunnybrook Health Sciences Centre, University of Toronto. P.B. receives salary support by NIH (T32-DK063687), in addition to research support by Thrasher Foundation, Juvenile Diabetes Research Foundation (JDRF), International Society of Pediatric and Adolescent Diabetes (ISPAD) and Center for Women’s Health Research at University of Colorado. J.A.U. is supported in part by funding from a Heart and Stroke Foundation of Canada National New Investigator/Ontario Clinician Scientist Award; Ontario Ministry of Research and Innovation Early Researcher Award; Women’s College Research Institute and Department of Medicine, Women’s College Hospital; Peter Munk Cardiac Centre, University Health Network; Department of Medicine and Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research, University of Toronto. D.Z.I.C. receives support from the Canadian Institutes of Health Research, as well as Diabetes Action Canada, a Strategy for Patient-Oriented Research initiative supported by the Canadian Institutes for Health Research. D.Z.I.C. also receives operating funding from the Heart and Stroke Richard Lewar Centre of Excellence in Cardiovascular Research and Banting and Best Diabetes Centre, University of Toronto
D.Z.I.C. is the recipient of a University of Toronto, Department of Medicine Merit Award. The authors were fully responsible for all content and editorial decisions, were involved at all stages of manuscript development and have approved the final version.
DISCLOSURES:
J.A.U. has received consulting fees or speaking honoraria from Amgen, Janssen, Merck, Novartis, Sanofi Pasteur; and grant support from Novartis. J.A.L. receives honorarium and/or consulting fees from Novo Nordisk, Merck Sharpe and Dohme, Eli Lilly and Co., and AstraZeneca. D.Z.I.C. receives operating funding from Boehringer Ingelheim, Janssen, AstraZeneca, Merck. D.Z.I.C. has received consulting fees or speaking honoraria from Boehringer Ingelheim, Janssen, AstraZeneca, Merck, Mitsubishi-Tanabe, Sanofi and Abbvie.
References
- 1.Metra M, Carubelli V, Ravera A, Stewart Coats AJ. Heart failure 2016: still more questions than answers. Internationalj cardiology. 2017;227:766–777. doi: 10.1016/j.ijcard.2016.10.060. [DOI] [PubMed] [Google Scholar]
- 2.AbdAlla S, Lother H, el Massiery A, Quitterer U. Increased AT(1) receptor heterodimers in preeclampsia mediate enhanced angiotensin II responsiveness. Nat Med. 2001;7:1003–1009. doi: 10.1038/nm0901-1003. [DOI] [PubMed] [Google Scholar]
- 3.Mentz RJ, Kelly JP, von Lueder TG, Voors AA, Lam CS, Cowie MR, Kjeldsen K, Jankowska EA, Atar D, Butler J, Fiuzat M, Zannad F, Pitt B, O’Connor CM. Noncardiac comorbidities in heart failure with reduced versus preserved ejection fraction. J Am Coll Cardiol. 2014;64:2281–2293. doi: 10.1016/j.jacc.2014.08.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Kannel WB, Hjortland M, Castelli WP. Role of diabetes in congestive heart failure: the Framingham study. Am J Cardiol. 1974;34:29–34. doi: 10.1016/0002-9149(74)90089-7. [DOI] [PubMed] [Google Scholar]
- 5.Patel PA, Liang L, Khazanie P, Hammill BG, Fonarow GC, Yancy CW, Bhatt DL, Curtis LH, Hernandez AF. Antihyperglycemic Medication Use Among Medicare Beneficiaries With Heart Failure, Diabetes Mellitus, and Chronic Kidney Disease. Circ Heart Fail. 2016;9 doi: 10.1161/CIRCHEARTFAILURE.115.002638. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE, Jr, Drazner MH, Fonarow GC, Geraci SA, Horwich T, Januzzi JL, Johnson MR, Kasper EK, Levy WC, Masoudi FA, McBride PE, McMurray JJ, Mitchell JE, Peterson PN, Riegel B, Sam F, Stevenson LW, Tang WH, Tsai EJ, Wilkoff BL. 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2013;62:e147–239. doi: 10.1016/j.jacc.2013.05.019. [DOI] [PubMed] [Google Scholar]
- 7.Lytvyn Y, Bjornstad P, Pun N, Cherney DZ. New and old agents in the management of diabetic nephropathy. Curr Opin Nephrol Hypertens. 2016;25:232–239. doi: 10.1097/MNH.0000000000000214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Mentz RJ, Broderick S, Shaw LK, Fiuzat M, O’Connor CM. Heart failure with preserved ejection fraction: comparison of patients with and without angina pectoris (from the Duke Databank for Cardiovascular Disease) J Am Coll Cardiol. 2014;63:251–258. doi: 10.1016/j.jacc.2013.09.039. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Cagnoni F, Destro M, Bontempelli E, Locatelli G, Hering D, Schlaich MP. Central Sympathetic Inhibition: a Neglected Approach for Treatment of Cardiac Arrhythmias? Curr Hypertens Rep. 2016;18:13–24. doi: 10.1007/s11906-015-0619-0. [DOI] [PubMed] [Google Scholar]
- 10.Hillege HL, Nitsch D, Pfeffer MA, Swedberg K, McMurray JJ, Yusuf S, Granger CB, Michelson EL, Ostergren J, Cornel JH, de Zeeuw D, Pocock S, van Veldhuisen DJ. Renal function as a predictor of outcome in a broad spectrum of patients with heart failure. Circulation. 2006;113:671–678. doi: 10.1161/CIRCULATIONAHA.105.580506. [DOI] [PubMed] [Google Scholar]
- 11.Ronco C, Haapio M, House AA, Anavekar N, Bellomo R. Cardiorenal syndrome. J Am Coll Cardiol. 2008;52:1527–1539. doi: 10.1016/j.jacc.2008.07.051. [DOI] [PubMed] [Google Scholar]
- 12.Fitchett DH, Udell JA, Inzucchi SE. Heart failure outcomes in clinical trials of glucose-lowering agents in patients with diabetes. Eur J Heart Fail. 2017;19:43–53. doi: 10.1002/ejhf.633. [DOI] [PubMed] [Google Scholar]
- 13.Groop PH, Cooper ME, Perkovic V, Hocher B, Kanasaki K, Schernthaner G, Sharma K, Stanton RC, Toro R, Cescutti J, Gordat M, Meinicke T, Koitka-Weber A, W HJ, Eynatten M. Effects of linagliptin on glycemic control and albuminuria in type 2 diabetes – the MARLINA–T2D™ trial. ADS-ADEA Meeting. 2016 Poster #62. [Google Scholar]
- 14.Son JW, Kim S. Dipeptidyl Peptidase 4 Inhibitors and the Risk of Cardiovascular Disease in Patients with Type 2 Diabetes: A Tale of Three Studies. Diabetes Metab J. 2015;39:373–383. doi: 10.4093/dmj.2015.39.5.373. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, Steinberg WM, Stockner M, Zinman B, Bergenstal RM, Buse JB. Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes. N Engl J Med. 2016;375:311–322. doi: 10.1056/NEJMoa1603827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jodar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, Woo V, Hansen O, Holst AG, Pettersson J, Vilsboll T. Semaglutide and Cardiovascular Outcomes in Patients with Type 2 Diabetes. N Engl J Med. 2016;375:1834–1844. doi: 10.1056/NEJMoa1607141. [DOI] [PubMed] [Google Scholar]
- 17.Lovshin JA. Glucagon-like Peptide-1 Receptor Agonists: A Class Update for Treating Type 2 Diabetes. Can J Diabetes. 2017 doi: 10.1016/j.jcjd.2017.08.242. [DOI] [PubMed] [Google Scholar]
- 18.Filion KB, Azoulay L, Platt RW, Dahl M, Dormuth CR, Clemens KK, Hu N, Paterson JM, Targownik L, Turin TC, Udell JA, Ernst P. A Multicenter Observational Study of Incretin-based Drugs and Heart Failure. N Engl J Med. 2016;374:1145–1154. doi: 10.1056/NEJMoa1506115. [DOI] [PubMed] [Google Scholar]
- 19.Monami M, Zannoni S, Pala L, Silverii A, Andreozzi F, Sesti G, Mannucci E. Effects of glucagon-like peptide-1 receptor agonists on mortality and cardiovascular events: A comprehensive meta-analysis of randomized controlled trials. Int J Cardiol. 2017;240:414–421. doi: 10.1016/j.ijcard.2017.03.163. [DOI] [PubMed] [Google Scholar]
- 20.Scheen AJ. GLP-1 receptor agonists and heart failure in diabetes. Diabetes & metabolism. 2017;43(Suppl 1):2s13–2s19. doi: 10.1016/S1262-3636(17)30068-X. [DOI] [PubMed] [Google Scholar]
- 21.Komajda M. Liraglutide in heart failure: caution is needed. Eur J Heart Fail. 2017;19:78–79. doi: 10.1002/ejhf.707. [DOI] [PubMed] [Google Scholar]
- 22.Jorsal A, Kistorp C, Holmager P, Tougaard RS, Nielsen R, Hanselmann A, Nilsson B, Moller JE, Hjort J, Rasmussen J, Boesgaard TW, Schou M, Videbaek L, Gustafsson I, Flyvbjerg A, Wiggers H, Tarnow L. Effect of liraglutide, a glucagon-like peptide-1 analogue, on left ventricular function in stable chronic heart failure patients with and without diabetes (LIVE)-a multicentre, double-blind, randomised, placebo-controlled trial. Eur J Heart Fail. 2017;19:69–77. doi: 10.1002/ejhf.657. [DOI] [PubMed] [Google Scholar]
- 23.Margulies KB, Hernandez AF, Redfield MM, Givertz MM, Oliveira GH, Cole R, Mann DL, Whellan DJ, Kiernan MS, Felker GM, McNulty SE, Anstrom KJ, Shah MR, Braunwald E, Cappola TP. Effects of Liraglutide on Clinical Stability Among Patients With Advanced Heart Failure and Reduced Ejection Fraction: A Randomized Clinical Trial. JAMA. 2016;316:500–508. doi: 10.1001/jama.2016.10260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Nielsen R, Jorsal A, Iversen P, Tolbod LP, Bouchelouche K, Sorensen J, Harms HJ, Flyvbjerg A, Tarnow L, Kistorp C, Gustafsson I, Botker HE, Wiggers H. Effect of liraglutide on myocardial glucose uptake and blood flow in stable chronic heart failure patients: A double-blind, randomized, placebo-controlled LIVE sub-study. J Nucl Cardiol. 2017 doi: 10.1007/s12350-017-1000-2. [DOI] [PubMed] [Google Scholar]
- 25.Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. The New England journal of medicine. 2015;373:2117–2128. doi: 10.1056/NEJMoa1504720. [DOI] [PubMed] [Google Scholar]
- 26.Wanner C, Inzucchi SE, Lachin JM, Fitchett D, von Eynatten M, Mattheus M, Johansen OE, Woerle HJ, Broedl UC, Zinman B. Empagliflozin and Progression of Kidney Disease in Type 2 Diabetes. N Engl J Med. 2016;375:323–334. doi: 10.1056/NEJMoa1515920. [DOI] [PubMed] [Google Scholar]
- 27.Neal B, Perkovic V, Mahaffey KW, Fulcher G, Erondu N, Desai M, Shaw W, Law G, Walton MK, Rosenthal N, Zeeuw D, Matthews DR. Optimising the analysis strategy for the CANVAS Program – a pre-specified plan for the integrated analyses of the CANVAS and CANVAS-R trials. Diabetes Obes Metab. 2017;19:926–935. doi: 10.1111/dom.12924. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Neal B, Perkovic V, de Zeeuw D, Mahaffey KW, Fulcher G, Stein P, Desai M, Shaw W, Jiang J, Vercruysse F, Meininger G, Matthews D. Rationale, design, and baseline characteristics of the Canagliflozin Cardiovascular Assessment Study (CANVAS)–a randomized placebo-controlled trial. Am Heart J. 2013;166:217–223.e11. doi: 10.1016/j.ahj.2013.05.007. [DOI] [PubMed] [Google Scholar]
- 29.Neal B, Perkovic V, Matthews DR, Mahaffey KW, Fulcher G, Meininger G, Erondu N, Desai M, Shaw W, Vercruysse F, Yee J, Deng H, de Zeeuw D. Rationale, design and baseline characteristics of the CANagliflozin cardioVascular Assessment Study-Renal (CANVAS-R): A randomized, placebo-controlled trial. Diabetes Obes Metab. 2017;19:387–393. doi: 10.1111/dom.12829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Neal B, Perkovic V, Matthews DR, Mahaffey KW, Fulcher G, Meininger G, Erondu N, Desai M, Shaw W, Vercruysse F, Yee J, Deng H, de Zeeuw D, Group C-RTC Rationale, design and baseline characteristics of the CANagliflozin cardioVascular Assessment Study-Renal (CANVAS-R): A randomized, placebo-controlled trial. Diabetes Obes Metab. 2017;19:387–393. doi: 10.1111/dom.12829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Neal B, Perkovic V, Mahaffey KW, de Zeeuw D, Fulcher G, Erondu N, Shaw W, Law G, Desai M, Matthews DR, Group CPC Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N Engl J Med. 2017;377:644–657. doi: 10.1056/NEJMoa1611925. [DOI] [PubMed] [Google Scholar]
- 32.Heerspink HJ, Perkins BA, Fitchett DH, Husain M, Cherney DZ. Sodium Glucose Cotransporter 2 Inhibitors in the Treatment of Diabetes: Cardiovascular and Kidney Effects, Potential Mechanisms and Clinical Applications. Circulation. 2016;134:752–772. doi: 10.1161/CIRCULATIONAHA.116.021887. [DOI] [PubMed] [Google Scholar]
- 33.Kosiborod M, Cavender MA, Fu AZ, Wilding JP, Khunti K, Holl RW, Norhammar A, Birkeland KI, Jorgensen M, Thuresson M, Arya N, Bodegard J, Hammar N, Fenici P. Lower Risk of Heart Failure and Death in Patients Initiated on SGLT-2 Inhibitors Versus Other Glucose-Lowering Drugs: The CVD-REAL Study. Circulation. 2017;136:249–259. doi: 10.1161/CIRCULATIONAHA.117.029190. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Birkeland KI, Jorgensen ME, Carstensen B, Persson F, Gulseth HL, Thuresson M, Fenici P, Nathanson D, Nystrom T, Eriksson JW, Bodegard 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. doi: 10.1016/S2213-8587(17)30258-9. [DOI] [PubMed] [Google Scholar]
- 35.Persson F, Nystrom T, Jorgensen ME, Carstensen B, Gulseth HL, Thuresson M, Fenici P, Nathanson D, Eriksson JW, Norhammar A, Bodegard J, Birkeland KI. Dapagliflozin Compared to DPP-4 inhibitors is Associated with Lower Risk of Cardiovascular Events and All-cause Mortality in Type 2 Diabetes Patients (CVD-REAL Nordic): a multinational observational study. Diabetes Obes Metab. 2017 doi: 10.1111/dom.13077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Nystrom T, Bodegard J, Nathanson D, Thuresson M, Norhammar A, Eriksson JW. Novel oral glucose-lowering drugs are associated with lower risk of all-cause mortality, cardiovascular events and severe hypoglycaemia compared with insulin in patients with type 2 diabetes. Diabetes Obes Metab. 2017;19:831–841. doi: 10.1111/dom.12889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Liu JJ, Lee T, DeFronzo RA. Why Do SGLT2 inhibitors inhibit only 30-50% of renal glucose reabsorption in humans? Diabetes. 2012;61:2199–2204. doi: 10.2337/db12-0052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bakris GL, Fonseca VA, Sharma K, Wright EM. Renal sodium-glucose transport: role in diabetes mellitus and potential clinical implications. Kidney Int. 2009;75:1272–1277. doi: 10.1038/ki.2009.87. [DOI] [PubMed] [Google Scholar]
- 39.Heerspink HJ, Perkins BA, Fitchett DH, Husain M, Cherney DZ. Sodium Glucose Cotransporter 2 Inhibitors in the Treatment of Diabetes Mellitus: Cardiovascular and Kidney Effects, Potential Mechanisms, and Clinical Applications. Circulation. 2016;134:752–772. doi: 10.1161/CIRCULATIONAHA.116.021887. [DOI] [PubMed] [Google Scholar]
- 40.Lytvyn Y, Skrtic M, Yang GK, Yip PM, Perkins BA, Cherney DZ. Glycosuria-mediated urinary uric acid excretion in patients with uncomplicated type 1 diabetes mellitus. Am J Physiol Renal Physiol. 2015;308:F77–83. doi: 10.1152/ajprenal.00555.2014. [DOI] [PubMed] [Google Scholar]
- 41.Ferrannini E, Solini A. SGLT2 inhibition in diabetes mellitus: rationale and clinical prospects. Nat Rev Endocrinol. 2012;8:495–502. doi: 10.1038/nrendo.2011.243. [DOI] [PubMed] [Google Scholar]
- 42.Musso C, Javor E, Cochran E, Balow JE, Gorden P. Spectrum of renal diseases associated with extreme forms of insulin resistance. Clin J Am Soc Nephrol. 2006;1:616–622. doi: 10.2215/CJN.01271005. [DOI] [PubMed] [Google Scholar]
- 43.Ndrepepa G, Braun S, King L, Hadamitzky M, Haase HU, Birkmeier KA, Schomig A, Kastrati A. Association of uric acid with mortality in patients with stable coronary artery disease. Metabolism. 2012;61:1780–1786. doi: 10.1016/j.metabol.2012.05.014. [DOI] [PubMed] [Google Scholar]
- 44.Huang H, Huang B, Li Y, Huang Y, Li J, Yao H, Jing X, Chen J, Wang J. Uric acid and risk of heart failure: a systematic review and meta-analysis. Eur J Heart Fail. 2014;16:15–24. doi: 10.1093/eurjhf/hft132. [DOI] [PubMed] [Google Scholar]
- 45.Wing RR, Bolin P, Brancati FL, Bray GA, Clark JM, Coday M, Crow RS, Curtis JM, Egan CM, Espeland MA, Evans M, Foreyt JP, Ghazarian S, Gregg EW, Harrison B, Hazuda HP, Hill JO, Horton ES, Hubbard VS, Jakicic JM, Jeffery RW, Johnson KC, Kahn SE, Kitabchi AE, Knowler WC, Lewis CE, Maschak-Carey BJ, Montez MG, Murillo A, Nathan DM, Patricio J, Peters A, Pi-Sunyer X, Pownall H, Reboussin D, Regensteiner JG, Rickman AD, Ryan DH, Safford M, Wadden TA, Wagenknecht LE, West DS, Williamson DF, Yanovski SZ. Cardiovascular effects of intensive lifestyle intervention in type 2 diabetes. N Engl J Med. 2013;369:145–154. doi: 10.1056/NEJMoa1212914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wilding JP, Norwood P, T’Joen C, Bastien A, List JF, Fiedorek FT. A study of dapagliflozin in patients with type 2 diabetes receiving high doses of insulin plus insulin sensitizers: applicability of a novel insulin-independent treatment. Diabetes care. 2009;32:1656–1662. doi: 10.2337/dc09-0517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.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;8:262–275.e9. doi: 10.1016/j.jash.2014.01.007. [DOI] [PubMed] [Google Scholar]
- 48.Tanaka H, Takano K, Iijima H, Kubo H, Maruyama N, Hashimoto T, Arakawa K, Togo M, Inagaki N, Kaku K. Factors Affecting Canagliflozin-Induced Transient Urine Volume Increase in Patients with Type 2 Diabetes Mellitus. Adv Ther. 2017;34:436–451. doi: 10.1007/s12325-016-0457-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Cherney DZI, Perkins BA, Soleymanlou N, Maione M, Lai V, Lee A, Fagan NM, Woerle HJ, Johansen OE, Broedl UC, Eynatten M. The Renal Hemodynamic Effect of SGLT2 Inhibition in Patients with Type 1 Diabetes. Circulation. 2014;129:587–597. doi: 10.1161/CIRCULATIONAHA.113.005081. [DOI] [PubMed] [Google Scholar]
- 50.Kim Y, Babu AR. Clinical potential of sodium-glucose cotransporter 2 inhibitors in the management of type 2 diabetes. Diabetes Metab Syndr Obes. 2012;5:313–327. doi: 10.2147/DMSO.S22545. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gilbert RE. The perils of clinical trials. Kidney Int. 2014;85:745–747. doi: 10.1038/ki.2013.406. [DOI] [PubMed] [Google Scholar]
- 52.Petrykiv S, Sjostrom CD, Greasley PJ, Xu J, Persson F, Heerspink HJ. Differential Effects of Dapagliflozin on Cardiovascular Risk Factors at Varying Degrees of Renal Function. Clin J Am Soc Nephrol. 2017;12:751–759. doi: 10.2215/CJN.10180916. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Inzucchi SE, Zinman B, Wanner C, Ferrari R, Fitchett D, Hantel S, Espadero RM, Woerle HJ, Broedl UC, Johansen OE. SGLT-2 inhibitors and cardiovascular risk: proposed pathways and review of ongoing outcome trials. Diab Vasc Dis Res. 2015;12:90–100. doi: 10.1177/1479164114559852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Titze J. A different view on sodium balance. Curr Opin Nephrol Hypertens. 2015;24:14–20. doi: 10.1097/MNH.0000000000000085. [DOI] [PubMed] [Google Scholar]
- 55.Schneider MP, Raff U, Kopp C, Scheppach JB, Toncar S, Wanner C, Schlieper G, Saritas T, Floege J, Schmid M, Birukov A, Dahlmann A, Linz P, Janka R, Uder M, Schmieder RE, Titze JM, Eckardt KU. Skin Sodium Concentration Correlates with Left Ventricular Hypertrophy in CKD. J Am Soc Nephrol. 2017;28:1867–1876. doi: 10.1681/ASN.2016060662. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Schmieder R, Ott C, Linz P, Jumar A, Friedrich S, Titze J, Hammon M, Uder M, Kistner I. OS 12-03 SGLT-2-INHIBITION WITH DAPAGLIFLOZIN REDUCES TISSUE SODIUM CONTENT. J Hypertens. 2016;34(Suppl 1):e76. ISH 2016 Abstract Book. [Google Scholar]
- 57.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. doi: 10.1161/CIRCULATIONAHA.113.005081. [DOI] [PubMed] [Google Scholar]
- 58.Rajasekeran H, Lytvyn Y, Cherney DZ. Sodium-glucose cotransporter 2 inhibition and cardiovascular risk reduction in patients with type 2 diabetes: the emerging role of natriuresis. Kidney Int. 2016;89:524–526. doi: 10.1016/j.kint.2015.12.038. [DOI] [PubMed] [Google Scholar]
- 59.Perkins BA, Udell JA, Cherney DZ. No Need to Sugarcoat the Message: Is Cardiovascular Risk Reduction From SGLT2 Inhibition Related to Natriuresis? Am J Kidney Dis. 2016;68:349–352. doi: 10.1053/j.ajkd.2016.03.410. [DOI] [PubMed] [Google Scholar]
- 60.Townsend RR, Machin I, Ren J, Trujillo A, Kawaguchi M, Vijapurkar U, Damaraju CV, Pfeifer M. Reductions in Mean 24-Hour Ambulatory Blood Pressure After 6-Week Treatment With Canagliflozin in Patients With Type 2 Diabetes Mellitus and Hypertension. J Clin Hypertens (Greenwich) 2016;18:43–52. doi: 10.1111/jch.12747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Tikkanen I, Narko K, Zeller C, Green A, Salsali A, Broedl UC, Woerle HJ. Empagliflozin reduces blood pressure in patients with type 2 diabetes and hypertension. Diabetes care. 2015;38:420–428. doi: 10.2337/dc14-1096. [DOI] [PubMed] [Google Scholar]
- 62.Chilton R, Tikkanen I, Cannon CP, Crowe S, Woerle HJ, Broedl UC, Johansen OE. Effects of empagliflozin on blood pressure and markers of arterial stiffness and vascular resistance in patients with type 2 diabetes. Diabetes Obes Metab. 2015;17:1180–1193. doi: 10.1111/dom.12572. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zimlichman R. Treatment of hypertension and metabolic syndrome: lowering blood pressure is not enough for organ protection, new approach-arterial destiffening. Curr Hypertens Rep. 2014;16:479–484. doi: 10.1007/s11906-014-0479-z. [DOI] [PubMed] [Google Scholar]
- 64.Cherney DZI, 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. doi: 10.1161/CIRCULATIONAHA.113.005081. [DOI] [PubMed] [Google Scholar]
- 65.Xie X, Atkins E, Lv J, Bennett A, Neal B, Ninomiya T, Woodward M, MacMahon S, Turnbull F, Hillis GS, Chalmers J, Mant J, Salam A, Rahimi K, Perkovic V, Rodgers A. Effects of intensive blood pressure lowering on cardiovascular and renal outcomes: updated systematic review and meta-analysis. Lancet. 2016;387:4354–43. doi: 10.1016/S0140-6736(15)00805-3. [DOI] [PubMed] [Google Scholar]
- 66.Lambert R, Srodulski S, Peng X, Margulies KB, Despa F, Despa S. Intracellular Na+ Concentration ([Na+]i) Is Elevated in Diabetic Hearts Due to Enhanced Na+-Glucose Cotransport. J Am Heart Assoc. 2015;4:e002183. doi: 10.1161/JAHA.115.002183. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Baartscheer A, Schumacher CA, Wust 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. doi: 10.1007/s00125-016-4134-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Liu T, Takimoto E, Dimaano VL, DeMazumder D, Kettlewell S, Smith G, Sidor A, Abraham TP, O’Rourke B. Inhibiting mitochondrial Na+/Ca2+ exchange prevents sudden death in a Guinea pig model of heart failure. Circulation research. 2014;115:44–54. doi: 10.1161/CIRCRESAHA.115.303062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Kohlhaas M, Liu T, Knopp A, Zeller T, Ong MF, Bohm M, O’Rourke B, Maack C. Elevated cytosolic Na+ increases mitochondrial formation of reactive oxygen species in failing cardiac myocytes. Circulation. 2010;121:1606–1613. doi: 10.1161/CIRCULATIONAHA.109.914911. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Erhardt LR. GUARD During Ischemia Against Necrosis (GUARDIAN) trial in acute coronary syndromes. Am J Cardiol. 1999;83:23G–25G. doi: 10.1016/s0002-9149(99)00216-7. [DOI] [PubMed] [Google Scholar]
- 71.Theroux P, Chaitman BR, Danchin N, Erhardt L, Meinertz T, Schroeder JS, Tognoni G, White HD, Willerson JT, Jessel A. Inhibition of the sodium-hydrogen exchanger with cariporide to prevent myocardial infarction in high-risk ischemic situations. Main results of the GUARDIAN trial. Guard during ischemia against necrosis (GUARDIAN) Investigators. Circulation. 2000;102:3032–3038. doi: 10.1161/01.cir.102.25.3032. [DOI] [PubMed] [Google Scholar]
- 72.Kraakman MJ, Lee MK, Al-Sharea A, Dragoljevic D, Barrett TJ, Montenont E, Basu D, Heywood S, Kammoun HL, Flynn M, Whillas A, Hanssen NM, Febbraio MA, Westein E, Fisher EA, Chin-Dusting J, Cooper ME, Berger JS, Goldberg IJ, Nagareddy PR, Murphy AJ. Neutrophil-derived S100 calcium-binding proteins A8/A9 promote reticulated thrombocytosis and atherogenesis in diabetes. J Clin Invest. 2017;127:2133–2147. doi: 10.1172/JCI92450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Abassi Z, Leor J, Landa N, Younis F, Hollander K, Mayoux E, Rath-Wolfson L, Rosenthal T. OS 05-04 EMPAGLIFLOZIN EXERTS CARDIO- AND NEPHRO-PROTECTIVE EFFECTS IN COHEN-ROSENTHAL DIABETIC HYPERTENSIVE RATS. J Hypertens. 2016;34(Suppl 1):e58–e59. ISH 2016 Abstract Book. [Google Scholar]
- 74.Bonner C, Kerr-Conte J, Gmyr V, Queniat G, Moerman E, Thevenet 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. doi: 10.1038/nm.3828. [DOI] [PubMed] [Google Scholar]
- 75.Ferrannini E, Mark M, Mayoux E. CV Protection in the EMPA-REG OUTCOME Trial: A “Thrifty Substrate” Hypothesis. Diabetes Care. 2016;39:1108–1114. doi: 10.2337/dc16-0330. [DOI] [PubMed] [Google Scholar]
- 76.Mudaliar S, Alloju S, Henry RR. Can a Shift in Fuel Energetics Explain the Beneficial Cardiorenal Outcomes in the EMPA-REG OUTCOME Study? A Unifying Hypothesis. Diabetes Care. 2016;39:1115–1122. doi: 10.2337/dc16-0542. [DOI] [PubMed] [Google Scholar]
- 77.Newman JC, Verdin E. beta-hydroxybutyrate: much more than a metabolite. Diabetes Res Clin Pract. 2014;106:173–181. doi: 10.1016/j.diabres.2014.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Martens P, Mathieu C, Verbrugge FH. Promise of SGLT2 Inhibitors in Heart Failure: Diabetes and Beyond. Curr Treat Options Cardiovasc Med. 2017;19:23. doi: 10.1007/s11936-017-0522-x. [DOI] [PubMed] [Google Scholar]
- 79.Fitchett D, Zinman B, Wanner C, Lachin JM, Hantel S, Salsali A, Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE. Heart failure outcomes with empagliflozin in patients with type 2 diabetes at high cardiovascular risk: results of the EMPA-REG OUTCOME(R) trial. Eur Heart J. 2016;37:1526–1534. doi: 10.1093/eurheartj/ehv728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Cherney DZ, Perkins BA, Soleymanlou N, Xiao F, Zimpelmann J, Woerle HJ, Johansen OE, Broedl UC, von Eynatten M, Burns KD. Sodium glucose cotransport-2 inhibition and intrarenal RAS activity in people with type 1 diabetes. Kidney Int. 2014;86:1057–1058. doi: 10.1038/ki.2014.246. [DOI] [PubMed] [Google Scholar]
- 81.McMurray J. EMPA-REG – the “diuretic hypothesis”. J Diabetes Complications. 2016;30:3–4. doi: 10.1016/j.jdiacomp.2015.10.012. [DOI] [PubMed] [Google Scholar]
- 82.Skrtic M, Cherney DZ. Sodium-glucose cotransporter-2 inhibition and the potential for renal protection in diabetic nephropathy. Curr Opin Nephrol Hypertens. 2015;24:96–103. doi: 10.1097/MNH.0000000000000084. [DOI] [PubMed] [Google Scholar]
- 83.Marx N, McGuire DK. Sodium-glucose cotransporter-2 inhibition for the reduction of cardiovascular events in high-risk patients with diabetes mellitus. Eur Heart J. 2016;37:3192–3200. doi: 10.1093/eurheartj/ehw110. [DOI] [PubMed] [Google Scholar]
- 84.Takeuchi T, Dohi K, Omori T, Moriwaki K, Sato Y, Nakamori S, Fujimoto N, Fujii E, Yamada N, Ito M. Diuretic effects of sodium-glucose cotransporter 2 inhibitor in patients with type 2 diabetes mellitus and heart failure. Int J Cardiol. 2015;201:1–3. doi: 10.1016/j.ijcard.2015.07.072. [DOI] [PubMed] [Google Scholar]
- 85.de Leeuw AE, de Boer RA. Sodium-glucose cotransporter 2 inhibition: cardioprotection by treating diabetes-a translational viewpoint explaining its potential salutary effects. Eur Heart J Cardiovasc Pharmacother. 2016;2:244–255. doi: 10.1093/ehjcvp/pvw009. [DOI] [PubMed] [Google Scholar]
- 86.Habibi J, Aroor AR, Sowers JR, Jia G, Hayden MR, Garro M, Barron B, Mayoux E, Rector RS, Whaley-Connell A, DeMarco VG. Sodium glucose transporter 2 (SGLT2) inhibition with empagliflozin improves cardiac diastolic function in a female rodent model of diabetes. Cardiovasc Diabetol. 2017;16:9–23. doi: 10.1186/s12933-016-0489-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Joubert M, Jagu B, Montaigne D, Marechal X, Tesse A, Ayer A, Dollet L, Le May C, Toumaniantz G, Manrique A, Charpentier F, Staels B, Magre J, Cariou B, Prieur X. The SGLT2 Inhibitor Dapagliflozin Prevents Cardiomyopathy in a DiabeticLipodystrophic Mouse Model. Diabetes. 2017;66:1030–1040. doi: 10.2337/db16-0733. [DOI] [PubMed] [Google Scholar]
- 88.Lee TM, Chang NC, Lin SZ. Dapagliflozin, a selective SGLT2 Inhibitor, attenuated cardiac fibrosis by regulating the macrophage polarization via STAT3 signaling in infarcted rat hearts. Free Radic Biol Med. 2017;104:298–310. doi: 10.1016/j.freeradbiomed.2017.01.035. [DOI] [PubMed] [Google Scholar]
- 89.Shi X, Verma S, Yun J, Brand-Arzamendi K, Singh KK, Liu X, Garg A, Quan A, Wen XY. Effect of empagliflozin on cardiac biomarkers in a zebrafish model of heart failure: clues to the EMPA-REG OUTCOME trial? Mol Cell Biochem. 2017 doi: 10.1007/s11010-017-3018-9. [DOI] [PubMed] [Google Scholar]
- 90.Verma S, Garg A, Yan AT, Gupta AK, Al-Omran M, Sabongui A, Teoh H, Mazer CD, Connelly KA. Effect of Empagliflozin on Left Ventricular Mass and Diastolic Function in Individuals With Diabetes: An Important Clue to the EMPA-REG OUTCOME Trial? Diabetes Care. 2016;39:e212–e213. doi: 10.2337/dc16-1312. [DOI] [PubMed] [Google Scholar]
- 91.Di Franco A, Cantini G, Tani A, Coppini R, Zecchi-Orlandini S, Raimondi L, Luconi M, Mannucci E. Sodium-dependent glucose transporters (SGLT) in human ischemic heart: A new potential pharmacological target. Int J Cardiol. 2017;243:86–90. doi: 10.1016/j.ijcard.2017.05.032. [DOI] [PubMed] [Google Scholar]
- 92.Wu JH, Foote C, Blomster J, Toyama T, Perkovic V, Sundstrom J, Neal B. Effects of sodium-glucose cotransporter-2 inhibitors on cardiovascular events, death, and major safety outcomes in adults with type 2 diabetes: a systematic review and meta-analysis. Lancet Diabetes Endocrinol. 2016;4:411–419. doi: 10.1016/S2213-8587(16)00052-8. [DOI] [PubMed] [Google Scholar]
- 93.Kohan DE, Fioretto P, Tang W, List JF. Long-term study of patients with type 2 diabetes and moderate renal impairment shows that dapagliflozin reduces weight and blood pressure but does not improve glycemic control. Kidney Int. 2014;85:962–971. doi: 10.1038/ki.2013.356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Williamson JR, Chang K, Frangos M, Hasan KS, Ido Y, Kawamura T, Nyengaard JR, van den Enden M, Kilo C, Tilton RG. Hyperglycemic pseudohypoxia and diabetic complications. Diabetes. 1993;42:801–813. doi: 10.2337/diab.42.6.801. [DOI] [PubMed] [Google Scholar]
- 95.Fralick M, Schneeweiss S, Patorno E. Risk of Diabetic Ketoacidosis after Initiation of an SGLT2 Inhibitor. N Engl J Med. 2017;376:2300–2302. doi: 10.1056/NEJMc1701990. [DOI] [PubMed] [Google Scholar]
- 96.Cherney DZ, Udell JA. Use of Sodium Glucose Cotransporter 2 Inhibitors in the Hands of Cardiologists: With Great Power Comes Great Responsibility. Circulation. 2016;134:1915–1917. doi: 10.1161/CIRCULATIONAHA.116.024764. [DOI] [PubMed] [Google Scholar]
- 97.DTB Select: 3 | March 2017. Drug and therapeutics bulletin. 2017;55:26–29. doi: 10.1136/dtb.2014.3.0239. [DOI] [PubMed] [Google Scholar]
- 98.Sha S, Polidori D, Farrell K, Ghosh A, Natarajan J, Vaccaro N, Pinheiro J, Rothenberg P, Plum-Morschel L. Pharmacodynamic differences between canagliflozin and dapagliflozin: results of a randomized, double-blind, crossover study. Diabetes Obes Metab. 2015;17:188–197. doi: 10.1111/dom.12418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Erkens JA, Klungel OH, Stolk RP, Spoelstra JA, Grobbee DE, Leufkens HG. Antihypertensive drug therapy and the risk of lower extremity amputations in pharmacologically treated type 2 diabetes patients. Pharmacoepidemiol Drug Saf. 2004;13:139–146. doi: 10.1002/pds.932. [DOI] [PubMed] [Google Scholar]
- 100.Hawley SA, Ford RJ, Smith BK, Gowans GJ, Mancini SJ, Pitt RD, Day EA, Salt IP, Steinberg GR, Hardie DG. The Na+/Glucose Cotransporter Inhibitor Canagliflozin Activates AMPK by Inhibiting Mitochondrial Function and Increasing Cellular AMP Levels. Diabetes. 2016;65:2784–2794. doi: 10.2337/db16-0058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Fadini GP, Avogaro A. SGTL2 inhibitors and amputations in the US FDA Adverse Event Reporting System. Lancet Diabetes Endocrinol. 2017;5:680–681. doi: 10.1016/S2213-8587(17)30257-7. [DOI] [PubMed] [Google Scholar]
- 102.Drug safety communications. FDA warns that SGLT2 inhibitors for diabetes may result in a serious condition of too much acid in the blood. US Food and Drug Administration. 2015;2017 [Google Scholar]
- 103.Review of diabetes medicines called SGLT2 inhibitors started: risk of diabetic ketoacidosis to be examined. European Medicines Agency. 2015;2017 [Google Scholar]
- 104.Fadini GP, Bonora BM, Avogaro A. SGLT2 inhibitors and diabetic ketoacidosis: data from the FDA Adverse Event Reporting System. Diabetologia. 2017;60:1385–1389. doi: 10.1007/s00125-017-4301-8. [DOI] [PubMed] [Google Scholar]
- 105.Rosenstock J, Ferrannini E. Euglycemic Diabetic Ketoacidosis: A Predictable, Detectable, and Preventable Safety Concern With SGLT2 Inhibitors. Diabetes care. 2015;38:1638–1642. doi: 10.2337/dc15-1380. [DOI] [PubMed] [Google Scholar]
- 106.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. doi: 10.1016/j.diabres.2017.04.004. [DOI] [PubMed] [Google Scholar]
- 107.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. doi: 10.1016/j.diabres.2017.04.017. [DOI] [PubMed] [Google Scholar]
- 108.Taylor SI, Blau JE, Rother KI. Possible adverse effects of SGLT2 inhibitors on bone. Lancet Diabetes Endocrinol. 2015;3:8–10. doi: 10.1016/S2213-8587(14)70227-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 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. doi: 10.1111/dom.12742. [DOI] [PubMed] [Google Scholar]
- 110.Rajasekeran H, Cherney DZ, Lovshin JA. Do effects of sodium-glucose cotransporter-2 inhibitors in patients with diabetes give insight into potential use in non-diabetic kidney disease? Curr Opin Nephrol Hypertens. 2017;26:358–367. doi: 10.1097/MNH.0000000000000343. [DOI] [PubMed] [Google Scholar]
- 111.Mann JF, Green D, Jamerson K, Ruilope LM, Kuranoff SJ, Littke T, Viberti G. Avosentan for overt diabetic nephropathy. J Am Soc Nephrol. 2010;21:527–535. doi: 10.1681/ASN.2009060593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.MacIsaac RJ, Jerums G, Ekinci EI. Effects of glycaemic management on diabetic kidney disease. World J Diabetes. 2017;8:172–186. doi: 10.4239/wjd.v8.i5.172. [DOI] [PMC free article] [PubMed] [Google Scholar]