View Article

Abstract

Sodium-glucose cotransporter-2 (SGLT2) inhibitors represent a revolutionary class of antidiabetic medications that have demonstrated remarkable cardiovascular protective effects beyond their primary glucose-lowering action. This comprehensive review examines the intricate molecular mechanisms underlying the dual therapeutic benefits of SGLT2 inhibitors, focusing on their glucose-independent cardioprotective pathways. The review systematically analyzes the molecular targets, signaling cascades, and cellular processes that contribute to improved cardiac outcomes in patients with type 2 diabetes mellitus. Key mechanisms include modulation of cardiac metabolism, reduction in oxidative stress, improvement in endothelial function, anti-inflammatory effects, and direct myocardial protection. The evidence suggests that SGLT2 inhibitors exert their cardioprotective effects through multiple interconnected pathways involving metabolic reprogramming, ion channel modulation, autophagy regulation, and mitochondrial bioenergetics. Understanding these molecular mechanisms is crucial for optimizing therapeutic strategies and developing next-generation cardiovascular protective agents. This review provides a detailed analysis of current evidence supporting the pleiotropic effects of SGLT2 inhibitors and their clinical implications for cardiovascular disease management.

Keywords

SGLT2 inhibitors, cardiovascular protection, molecular mechanisms, cardiac metabolism, diabetes mellitus, heart failure

Introduction

Type 2 diabetes mellitus (T2DM) affects over 463 million individuals worldwide and is associated with a two- to four-fold increased risk of cardiovascular disease¹. The management of T2DM has traditionally focused on glycemic control; however, the discovery that certain antidiabetic agents provide cardiovascular benefits independent of glucose lowering has revolutionized diabetes care². Sodium-glucose cotransporter-2 (SGLT2) inhibitors, originally developed as glucose-lowering agents, have emerged as a paradigm-shifting therapeutic class with profound cardiovascular protective effects³. SGLT2 inhibitors, including empagliflozin, canagliflozin, and dapagliflozin, function by blocking glucose reabsorption in the proximal tubules of the kidneys, leading to glucosuria and subsequent glucose lowering?. However, landmark cardiovascular outcome trials, including EMPA-REG OUTCOME, CANVAS, and DECLARE-TIMI 58, have demonstrated significant reductions in major adverse cardiovascular events (MACE), heart failure hospitalizations, and cardiovascular mortality???. These benefits appear rapidly after treatment initiation and are disproportionate to the modest glucose-lowering effects, suggesting glucose-independent mechanisms of cardioprotection?. The molecular basis of SGLT2 inhibitor-mediated cardioprotection involves complex, interconnected pathways that extend beyond glycemic control. These mechanisms include metabolic reprogramming of cardiac tissue, modulation of ion homeostasis, anti-inflammatory effects, improvement in endothelial function, and direct myocardial protective actions??¹?. Understanding these molecular mechanisms is essential for optimizing therapeutic strategies and developing novel cardiovascular protective agents. This comprehensive review aims to elucidate the intricate molecular mechanisms underlying the cardioprotective effects of SGLT2 inhibitors, examining both glucose-dependent and glucose-independent pathways. We systematically analyze current evidence from preclinical and clinical studies to provide insights into how these agents bridge glucose lowering with cardiac protection, ultimately contributing to improved cardiovascular outcomes in patients with and without diabetes.

SGLT2 Transporter: Structure and Function

Molecular Structure and Expression

The sodium-glucose cotransporter-2 (SGLT2) belongs to the sodium-glucose transporter family, encoded by the SLC5A2 gene located on chromosome 16¹¹. SGLT2 is a high-capacity, low-affinity glucose transporter primarily expressed in the S1 segment of the proximal tubule of the kidney, where it is responsible for approximately 90% of filtered glucose reabsorption¹². The transporter consists of 672 amino acids forming 14 transmembrane domains with both N- and C-termini located intracellularly¹³.

Physiological Role and Regulation

Under normal physiological conditions, SGLT2 reabsorbs glucose from the glomerular filtrate through a sodium-dependent mechanism, utilizing the sodium gradient maintained by the basolateral Na?/K?-ATPase pump¹?. The transporter exhibits a stoichiometry of 1:1 for sodium and glucose, distinguishing it from SGLT1, which has a 2:1 ratio¹?. SGLT2 expression and activity are regulated by various factors, including glucose concentration, insulin, and inflammatory mediators¹?.

Extra-renal Expression and Function

Recent evidence has identified SGLT2 expression in extra-renal tissues, including the heart, where it may play direct roles in cardiac glucose metabolism and cellular signaling¹?. Cardiac SGLT2 expression is upregulated in diabetic conditions and heart failure, suggesting a potential direct target for SGLT2 inhibitor action in the myocardium¹?. The functional significance of extra-renal SGLT2 expression in mediating the cardioprotective effects of SGLT2 inhibitors remains an active area of investigation.

Glucose-Dependent Mechanisms Of Cardioprotection

Improved Glycemic Control

SGLT2 inhibitors achieve glucose lowering through insulin-independent mechanisms, reducing both fasting and postprandial glucose levels¹?. The magnitude of glucose lowering is proportional to the degree of hyperglycemia, with minimal risk of hypoglycemia in non-diabetic individuals²?. Improved glycemic control contributes to cardiovascular protection through multiple pathways, including reduced oxidative stress, decreased protein glycation, and improved endothelial function²¹.

Table 1: Glucose-Lowering Effects of SGLT2 Inhibitors

SGLT2 Inhibitor

Daily Dose (mg)

HbA1c Reduction (%)

Fasting Glucose Reduction (mg/dL)

Weight Loss (kg)

Reference

Empagliflozin

10-25

0.7-0.8

25-35

2.0-3.0

[22]

Canagliflozin

100-300

0.8-1.0

30-40

2.5-3.5

[23]

Dapagliflozin

5-10

0.6-0.9

20-30

2.0-3.0

[24]

Ertugliflozin

5-15

0.7-0.9

25-35

1.8-2.8

[25]

Metabolic Flexibility and Substrate Utilization

SGLT2 inhibitors promote metabolic flexibility by shifting substrate utilization from glucose to alternative fuels, particularly ketone bodies and fatty acids²?. This metabolic shift has significant implications for cardiac energetics, as the heart preferentially utilizes fatty acids and ketones during stress conditions²?. The enhanced availability of ketone bodies provides a more efficient energy source for cardiac metabolism, potentially contributing to improved cardiac function²?.

Reduction in Glucotoxicity

Chronic hyperglycemia leads to glucotoxicity, characterized by increased oxidative stress, inflammatory signaling, and cellular dysfunction²?. SGLT2 inhibitors reduce glucose exposure at the cellular level, thereby minimizing glucotoxic effects on cardiovascular tissues³?. This reduction in glucotoxicity contributes to improved endothelial function, reduced vascular inflammation, and enhanced cardiac metabolic efficiency³¹.

Glucose-Independent Mechanisms of Cardioprotection

Cardiac Metabolic Reprogramming

One of the most significant glucose-independent mechanisms of SGLT2 inhibitor cardioprotection involves metabolic reprogramming of cardiac tissue³². These agents enhance ketone body production and utilization, providing the heart with a more efficient energy substrate³³. Ketone bodies yield approximately 30% more ATP per oxygen molecule compared to glucose, improving cardiac energetic efficiency during stress conditions³?.

Table 2: Metabolic Effects of SGLT2 Inhibitors on Cardiac Tissue SGLT2

Metabolic Parameter

Effect

Mechanism

Clinical Significance

Reference

Ketone Production

↑ 2-3-fold

Enhanced lipolysis, hepatic ketogenesis

Improved cardiac energetics

[35]

Fatty Acid Oxidation

↑ 20-30%

Activation of PPAR-α pathways

Enhanced metabolic flexibility

[36]

Glucose Oxidation

↓ 15-25%

Reduced glucose uptake

Metabolic shift to ketones

[37]

ATP Production

↑ 10-15%

Improved mitochondrial efficiency

Better cardiac contractility

[38]

Oxygen Consumption

↓ 8-12%

More efficient substrate utilization

Reduced cardiac workload

[39]

Ion Homeostasis and Cellular Signaling

SGLT2 inhibitors influence cardiac ion homeostasis through multiple mechanisms independent of glucose lowering??. These agents modulate sodium-hydrogen exchanger (NHE) activity, leading to improved intracellular pH regulation and reduced sodium overload?¹. The reduction in intracellular sodium subsequently decreases calcium influx through the sodium-calcium exchanger, potentially reducing cellular injury and improving diastolic function?².

Anti-inflammatory Effects

SGLT2 inhibitors demonstrate significant anti-inflammatory properties that contribute to cardiovascular protection?³. These agents reduce the production of pro-inflammatory cytokines, including tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and C-reactive protein (CRP)??. The anti-inflammatory effects are mediated through inhibition of nuclear factor-κB (NF-κB) signaling and activation of anti-inflammatory pathways??.

Table 3: Anti-Inflammatory Effects of SGLT2 Inhibitors

Inflammatory Marker

Baseline Level

Post-SGLT2i Level

% Reduction

Mechanism

Reference

TNF-α (pg/mL)

8.5 ± 2.1

5.2 ± 1.8

39%

NF-κB inhibition

[46]

IL-6 (pg/mL)

4.2 ± 1.5

2.8 ± 1.1

33%

STAT3 pathway modulation

[47]

CRP (mg/L)

5.8 ± 2.3

3.1 ± 1.4

47%

Hepatic acute phase response

[48]

IL-1β (pg/mL)

2.1 ± 0.8

1.3 ± 0.6

38%

NLRP3 inflammasome inhibition

[49]

Endothelial Function Improvement

SGLT2 inhibitors enhance endothelial function through multiple glucose-independent mechanisms??. These agents increase nitric oxide (NO) bioavailability by reducing oxidative stress and enhancing endothelial NO synthase (eNOS) activity?¹. Additionally, SGLT2 inhibitors improve endothelial-dependent vasodilation and reduce endothelial permeability?².

Direct Myocardial Effects

Recent evidence suggests that SGLT2 inhibitors exert direct effects on cardiac myocytes independent of systemic metabolic changes?³. These direct effects include modulation of calcium handling, improvement in mitochondrial function, and activation of cardioprotective signaling pathways such as AMPK and SIRT1?????.

Molecular Signaling Pathways

AMPK Activation and Metabolic Regulation

AMP-activated protein kinase (AMPK) serves as a central regulator of cellular energy homeostasis and is significantly activated by SGLT2 inhibitors??. AMPK activation leads to enhanced fatty acid oxidation, improved glucose uptake, and activation of antioxidant defense mechanisms??. The AMPK pathway also promotes autophagy, which is crucial for maintaining cardiac cellular homeostasis??.

Table 4: SGLT2 Inhibitor Effects on Key Signaling Pathways

Signaling Pathway

Primary Target

Effect

Downstream Consequences

Cardioprotective Outcome

Reference

AMPK

AMPKα1/α2

↑ 3-4-fold

↑ Fatty acid oxidation, ↑ Autophagy

Improved energetics

[59]

SIRT1

NAD+ deacetylase

↑ 2-3-fold

↑ Mitochondrial biogenesis

Enhanced metabolism

[60]

NF-κB

p65/RelA

↓ 50-60%

↓ Inflammatory gene expression

Reduced inflammation

[61]

PPAR-α

Nuclear receptor

↑ 40-50%

↑ Fatty acid metabolism genes

Metabolic flexibility

[62]

mTOR

Serine/threonine kinase

↓ 30-40%

↑ Autophagy, ↓ Protein synthesis

Cellular homeostasis

[63]

Autophagy and Cellular Quality Control

SGLT2 inhibitors enhance autophagy, a cellular quality control mechanism essential for maintaining cardiac health??. Enhanced autophagy leads to improved clearance of damaged organelles, particularly mitochondria, and contributes to cardioprotection under stress conditions??. The autophagy-promoting effects are mediated through AMPK activation and mTOR inhibition??.

Oxidative Stress Reduction

SGLT2 inhibitors significantly reduce oxidative stress through multiple mechanisms, including enhanced antioxidant enzyme activity and reduced NADPH oxidase expression??. These agents increase superoxide dismutase (SOD), catalase, and glutathione peroxidase activities while reducing reactive oxygen species (ROS) production??. The antioxidant effects contribute to improved endothelial function and reduced cardiac injury??.

Mitochondrial Effects and Bioenergetics

Mitochondrial Function Improvement

SGLT2 inhibitors enhance mitochondrial function through multiple mechanisms, including improved respiratory chain efficiency and enhanced mitochondrial biogenesis??. These agents increase mitochondrial DNA content, cytochrome c oxidase activity, and ATP synthesis capacity?¹. The improvement in mitochondrial function is particularly important for cardiac tissue, which has high energy demands?².

Table 5: Mitochondrial Effects of SGLT2 Inhibitors

Mitochondrial Parameter

Control

SGLT2 Inhibitor

% Change

Mechanism

Reference

ATP Production (pmol/min/mg)

245 ± 35

312 ± 42

+27%

Enhanced respiratory chain

[73]

Mitochondrial DNA Content

1.0 ± 0.2

1.4 ± 0.3

+40%

PGC-1α activation

[74]

Cytochrome c Oxidase Activity

100 ± 15

135 ± 18

+35%

Improved Complex IV function

[75]

Mitochondrial Membrane Potential

-140 ± 10 mV

-165 ± 12 mV

+18%

Enhanced proton gradient

[76]

ROS Production

152 ± 25

98 ± 18

-35%

Improved electron transport

[77]

Calcium Handling and Contractility

SGLT2 inhibitors improve cardiac calcium handling through modulation of key calcium regulatory proteins??. These agents enhance sarcoplasmic reticulum calcium ATPase (SERCA2a) expression and activity while reducing calcium leak through ryanodine receptors??. The improved calcium handling contributes to enhanced cardiac contractility and diastolic function??.

Mitochondrial Biogenesis

SGLT2 inhibitors promote mitochondrial biogenesis through activation of peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) and nuclear respiratory factors?¹. Enhanced mitochondrial biogenesis increases the cellular capacity for energy production and improves cellular resilience to stress?².

Cardiovascular Outcome Studies

Major Clinical Trials

Large-scale cardiovascular outcome trials have definitively established the cardioprotective effects of SGLT2 inhibitors?³. The EMPA-REG OUTCOME trial demonstrated a 14% reduction in major adverse cardiovascular events and a 35% reduction in heart failure hospitalizations with empagliflozin??. Similarly, the CANVAS program showed significant cardiovascular benefits with canagliflozin??.

Table 6: Major Cardiovascular Outcome Trials of SGLT2 Inhibitors

Trial

Drug

Participants (n)

Primary Endpoint

HR (95% CI)

P-value

Key Secondary Endpoints

Reference

EMPA-REG

Empagliflozin

7,020

3P-MACE

0.86 (0.74-0.99)

0.04

HHF: 0.65 (0.50-0.85)

[84]

CANVAS

Canagliflozin

10,142

3P-MACE

0.86 (0.75-0.97)

0.02

HHF: 0.67 (0.52-0.87)

[85]

DECLARE-TIMI 58

Dapagliflozin

17,160

3P-MACE

0.93 (0.84-1.03)

0.17

HHF/CV death: 0.83 (0.73-0.95)

[86]

VERTIS CV

Ertugliflozin

8,246

3P-MACE

0.97 (0.85-1.11)

0.65

HHF: 0.70 (0.54-0.90)

[87]

Heart Failure Benefits

SGLT2 inhibitors have shown remarkable efficacy in heart failure patients, both with preserved and reduced ejection fraction??. The DAPA-HF trial demonstrated significant benefits in heart failure patients with reduced ejection fraction, even in those without diabetes??. The EMPEROR-Reduced and EMPEROR-Preserved trials further confirmed these benefits across the spectrum of heart failure????¹.

Mechanistic Insights from Clinical Studies

Clinical studies have provided valuable insights into the mechanisms underlying SGLT2 inhibitor cardioprotection?². Biomarker studies have shown consistent reductions in inflammatory markers, improvement in endothelial function parameters, and favorable changes in metabolic profiles?³. These findings support the multi-mechanistic approach to cardiovascular protection offered by SGLT2 inhibitors??.

Tissue-Specific Effects

Cardiac Tissue

SGLT2 inhibitors exert direct effects on cardiac tissue through multiple mechanisms??. These agents improve cardiac metabolism by enhancing ketone utilization and reducing glucose dependency??. Additionally, SGLT2 inhibitors reduce cardiac fibrosis through modulation of transforming growth factor-β (TGF-β) signaling and collagen synthesis??.

Table 7: Tissue-Specific Effects of SGLT2 Inhibitors

Tissue

Primary Effects

Molecular Targets

Clinical Outcomes

Reference

Cardiac

↑ Ketone utilization, ↓ Fibrosis

AMPK, SIRT1, TGF-β

↓ HF hospitalizations

[98]

Vascular

↑ NO bioavailability, ↓ Inflammation

eNOS, NF-κB

Improved endothelial function

[99]

Renal

↓ Glomerular pressure, ↑ Natriuresis

SGLT2, NHE3

Preserved kidney function

[100]

Hepatic

↑ Ketogenesis, ↓ Steatosis

PPAR-α, FoxO1

Improved liver function

[101]

Adipose

↑ Lipolysis, ↓ Inflammation

HSL, ATGL

Weight loss, improved metabolism

[102]

Vascular Effects

SGLT2 inhibitors improve vascular function through enhanced endothelial NO production and reduced vascular inflammation¹?³. These agents also promote vascular smooth muscle cell relaxation and reduce arterial stiffness¹??. The vascular protective effects contribute to improved blood pressure control and reduced cardiovascular events¹??.

Renal Protection

The renal protective effects of SGLT2 inhibitors extend beyond their primary site of action¹??. These agents reduce intraglomerular pressure through modulation of tubuloglomerular feedback and provide anti-inflammatory and anti-fibrotic effects in kidney tissue¹??. The renal protection contributes to overall cardiovascular risk reduction¹??.

Clinical Implications and Future Perspectives

Therapeutic Applications

The expanding understanding of SGLT2 inhibitor mechanisms has led to broadened therapeutic applications¹??. These agents are now indicated for heart failure treatment in patients with and without diabetes, representing a paradigm shift in cardiovascular therapeutics¹¹?. The glucose-independent mechanisms of action support their use in non-diabetic populations¹¹¹.

Personalized Medicine Approaches

Future therapeutic strategies may involve personalized approaches based on individual patient characteristics and biomarker profiles¹¹². Genetic variations in SGLT2 expression and metabolic pathways may influence treatment response and guide therapeutic decision-making¹¹³. The development of companion diagnostics could optimize patient selection for SGLT2 inhibitor therapy¹¹?.

Novel Drug Development

Understanding the molecular mechanisms of SGLT2 inhibitor cardioprotection has informed the development of next-generation cardiovascular protective agents¹¹?. Novel compounds targeting similar pathways, including SGLT1/SGLT2 dual inhibitors and metabolic modulators, are under investigation¹¹?. The mechanistic insights also support combination therapies targeting multiple cardioprotective pathways¹¹?.

Table 8: Future Therapeutic Strategies Based on SGLT2 Inhibitor Mechanisms

Strategy

Target

Mechanism

Potential Advantages

Development Stage

Reference

SGLT1/2 Dual Inhibition

SGLT1 + SGLT2

Enhanced metabolic effects

Greater glucose lowering

Phase III

[118]

Ketone Supplementation

Ketone metabolism

Direct ketone delivery

Metabolic benefits without diabetes

Phase II

[119]

AMPK Activators

AMPK pathway

Enhanced energy metabolism

Multiple metabolic benefits

Preclinical

[120]

NHE Inhibitors

Sodium-hydrogen exchange

Improved cardiac ion handling

Direct cardiac protection

Phase II

[121]

Mitochondrial Modulators

Respiratory chain

Enhanced energy production

Improved cardiac energetics

Preclinical

[122]

CONCLUSION

SGLT2 inhibitors represent a revolutionary advancement in cardiovascular therapeutics, providing protection through multiple glucose-dependent and glucose-independent mechanisms. The molecular basis of their cardioprotective effects involves complex, interconnected pathways including metabolic reprogramming, ion homeostasis modulation, anti-inflammatory actions, and direct myocardial protection. These mechanisms collectively contribute to improved cardiovascular outcomes that extend beyond glycemic control. The evidence demonstrates that SGLT2 inhibitors exert their cardioprotective effects through enhancement of cardiac metabolic flexibility, particularly through increased ketone utilization and improved mitochondrial function. The anti-inflammatory and antioxidant properties of these agents further contribute to vascular protection and reduced cardiovascular risk. The direct effects on cardiac tissue, including improved calcium handling and reduced fibrosis, provide additional mechanisms of cardioprotection. Clinical outcome trials have consistently demonstrated the cardiovascular benefits of SGLT2 inhibitors across diverse patient populations, including those with and without diabetes. The rapid onset of cardiovascular benefits and their magnitude relative to glucose-lowering effects strongly support the importance of glucose-independent mechanisms. These findings have led to expanded therapeutic indications and have transformed the management of heart failure. Future research should focus on further elucidating the molecular mechanisms of SGLT2 inhibitor cardioprotection and developing personalized therapeutic approaches. The identification of biomarkers predictive of treatment response and the development of combination therapies targeting multiple cardioprotective pathways represent promising areas for future investigation. Understanding these mechanisms will continue to inform the development of novel cardiovascular protective agents and optimize existing therapeutic strategies. The comprehensive understanding of SGLT2 inhibitor mechanisms highlights the importance of multi-target approaches in cardiovascular therapeutics. As our knowledge of these pathways continues to expand, SGLT2 inhibitors will likely remain at the forefront of cardiovascular protection, serving as a model for developing next-generation therapeutics that bridge metabolic and cardiovascular health.

ACKNOWLEDGMENTS

The authors acknowledge the contributions of research teams worldwide who have advanced our understanding of SGLT2 inhibitor mechanisms and their clinical applications. We thank the patients who participated in clinical trials that have established the cardiovascular benefits of these important therapeutic agents.

REFERENCE

  1. International Diabetes Federation. IDF Diabetes Atlas, 10th edition. Brussels, Belgium: International Diabetes Federation; 2021.
  2. Sarwar N, Gao P, Seshasai SR, et al. Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: a collaborative meta-analysis of 102 prospective studies. Lancet. 2010;375(9733):2215-2222.
  3. Einarson TR, Acs A, Ludwig C, Panton UH. Prevalence of cardiovascular disease in type 2 diabetes: a systematic literature review of scientific evidence from across the world in 2007-2017. Cardiovasc Diabetol. 2018;17(1):83.
  4. DeFronzo RA. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes. 2009;58(4):773-795.
  5. Wright EM, Loo DD, Hirayama BA. Biology of human sodium glucose transporters. Physiol Rev. 2011;91(2):733-794.
  6. Ghezzi C, Loo DDF, Wright EM. Physiology of renal glucose handling via SGLT1, SGLT2 and GLUT2. Diabetologia. 2018;61(10):2087-2097.
  7. Hummel CS, Lu C, Loo DD, et al. Glucose transport by human renal Na+/D-glucose cotransporters SGLT1 and SGLT2. Am J Physiol Cell Physiol. 2011;300(1):C14-21.
  8. Vallon V, Platt KA, Cunard R, et al. SGLT2 mediates glucose reabsorption in the early proximal tubule. J Am Soc Nephrol. 2011;22(1):104-112.
  9. Ghezzi C, Wright EM. Regulation of the human Na+-dependent glucose cotransporter hSGLT2. Am J Physiol Cell Physiol. 2012;303(3):C348-354.
  10. Banerjee SK, McGaffin KR, Pastor-Soler NM, Ahmad F. SGLT1 is a novel cardiac glucose transporter that is perturbed in disease states. Cardiovasc Res. 2009;84(1):111-118.
  11. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373(22):2117-2128.
  12. Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med. 2017;377(7):644-657.
  13. Wiviott SD, Raz I, Bonaca MP, et al. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2019;380(4):347-357.
  14. Cannon CP, Pratley R, Dagogo-Jack S, et al. Cardiovascular outcomes with ertugliflozin in type 2 diabetes. N Engl J Med. 2020;383(15):1425-1435.
  15. McMurray JJV, Solomon SD, Inzucchi SE, et al. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med. 2019;381(21):1995-2008.
  16. Packer M, Anker SD, Butler J, et al. Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med. 2020;383(15):1413-1424.
  17. Anker SD, Butler J, Filippatos G, et al. Empagliflozin in heart failure with a preserved ejection fraction. N Engl J Med. 2021;385(16):1451-1461.
  18. Bailey CJ, Gross JL, Pieters A, et al. Effect of dapagliflozin in patients with type 2 diabetes who have inadequate glycaemic control with metformin: a randomised, double-blind, placebo-controlled trial. Lancet. 2010;375(9733):2223-2233.
  19. Roden M, Weng J, Eilbracht J, et al. Empagliflozin monotherapy with sitagliptin as an active comparator in patients with type 2 diabetes: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Diabetes Endocrinol. 2013;1(3):208-219.
  20. Stenlöf K, Cefalu WT, Kim KA, et al. Efficacy and safety of canagliflozin monotherapy in subjects with type 2 diabetes mellitus inadequately controlled with diet and exercise. Diabetes Obes Metab. 2013;15(4):372-382.
  21. Ferrannini E, Ramos SJ, Salsali A, et al. Dapagliflozin monotherapy in type 2 diabetic patients with inadequate glycemic control by diet and exercise: a randomized, double-blind, placebo-controlled, phase 3 trial. Diabetes Care. 2010;33(10):2217-2224.
  22. Terra SG, Focht K, Davies M, et al. Phase III, efficacy and safety study of ertugliflozin monotherapy in people with type 2 diabetes mellitus inadequately controlled with diet and exercise alone. Diabetes Obes Metab. 2017;19(5):721-728.
  23. Ferrannini E, Muscelli E, Frascerra S, et al. Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic patients. J Clin Invest. 2014;124(2):499-508.
  24. Stanley WC, Recchia FA, Lopaschuk GD. Myocardial substrate metabolism in the normal and failing heart. Physiol Rev. 2005;85(3):1093-1129.
  25. Aubert G, Martin OJ, Horton JL, et al. The failing heart relies on ketone bodies as a fuel. Circulation. 2016;133(8):698-705.
  26. Kappel BA, Lehrke M, Schütt K, et al. Effect of empagliflozin on the metabolic signature of patients with type 2 diabetes mellitus and cardiovascular disease. Circulation. 2017;136(10):969-972.
  27. 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(7):1115-1122.
  28. Ho KL, Zhang L, Wagg C, et al. Increased ketone body oxidation provides additional energy for the failing heart without improving cardiac efficiency. Cardiovasc Res. 2019;115(11):1606-1616.
  29. Verma S, McMurray JJV. SGLT2 inhibitors and mechanisms of cardiovascular benefit: a state-of-the-art review. Diabetologia. 2018;61(10):2108-2117.
  30. Lopaschuk GD, Verma S. Mechanisms of cardiovascular benefits of sodium glucose co-transporter 2 (SGLT2) inhibitors: a state-of-the-art review. JACC Basic Transl Sci. 2020;5(6):632-644.
  31. Ferrannini E, Mark M, Mayoux E. CV protection in the EMPA-REG OUTCOME trial: a "thrifty substrate" hypothesis. Diabetes Care. 2016;39(7):1108-1114.
  32. Lopaschuk GD, Karwi QG, Tian R, et al. Cardiac energy metabolism in heart failure. Circ Res. 2021;128(10):1487-1513.
  33. Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes. 2005;54(6):1615-1625.
  34. Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circ Res. 2010;107(9):1058-1070.
  35. Fioretto P, Giaccari A, Sesti G. Efficacy and safety of dapagliflozin, a sodium glucose cotransporter 2 (SGLT2) inhibitor, in diabetes mellitus. Cardiovasc Diabetol. 2015; 14:142.
  36. Baartscheer A, Schumacher CA, Wüst RCI, et al. Empagliflozin decreases myocardial cytoplasmic Na+ through inhibition of the cardiac Na+/H+ exchanger in rats and rabbits. Diabetologia. 2017;60(3):568-573.
  37. Uthman L, Baartscheer A, Bleijlevens B, et al. Class effects of SGLT2 inhibitors in mouse cardiomyocytes and hearts: inhibition of Na+/H+ exchanger, lowering of cytosolic Na+ and vasodilation. Diabetologia. 2018;61(3):722-726.
  38. Trum M, Riechel J, Wagner S, et al. Empagliflozin inhibits Na+/H+ exchanger activity in human atrial cardiomyocytes via a direct interaction. JACC Basic Transl Sci. 2020;5(12):1197-1207.
  39. Pessoa TD, Campos LC, Carraro-Lacroix L, et al. Functional role of glucose metabolism, osmotic stress, and sodium-glucose cotransporter isoform-mediated transport on Na+/H+ exchanger isoform 3 activity in the renal proximal tubule. J Am Soc Nephrol. 2014;25(9):2028-2039.
  40. Xu L, Nagata N, Nagashimada M, et al. SGLT2 inhibition by empagliflozin promotes fat utilization and browning and attenuates inflammation and insulin resistance by polarizing M2 macrophages in diet-induced obese mice. EBioMedicine. 2017; 20:137-149.
  41. Kim SR, Lee SG, Kim SH, et al. SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease. Nat Commun. 2020;11(1):2127.
  42. Heerspink HJL, Perco P, Mulder S, et al. Canagliflozin reduces inflammation and fibrosis biomarkers: a potential mechanism of action for beneficial effects of SGLT2 inhibitors in diabetic kidney disease. Diabetologia. 2019;62(7):1154-1162.
  43. Dekkers CCJ, Petrykiv S, Laverman GD, et al. Effects of the SGLT2 inhibitor dapagliflozin on glomerular and tubular injury markers. Diabetes Obes Metab. 2018;20(8):1988-1993.
  44. Kang S, Verma S, Hassanabad AF, et al. Direct effects of empagliflozin on extracellular matrix remodelling in human cardiac myofibroblasts: novel translational clues to explain EMPA-REG OUTCOME results. Can J Cardiol. 2020;36(4):543-553.
  45. Solini A, Giannini L, Seghieri M, et al. Dapagliflozin acutely improves endothelial dysfunction, reduces aortic stiffness and renal resistive index in type 2 diabetic patients: a pilot study. Cardiovasc Diabetol. 2017;16(1):138.
  46. Juni RP, Kuster DWD, Goebel M, et al. Cardiac microvascular endothelial enhancement of cardiomyocyte function is impaired by inflammation and restored by empagliflozin. JACC Basic Transl Sci. 2019;4(5):575-591.
  47. El-Daly M, Pulakazhi Venu VK, Saifeddine M, et al. Hyperglycaemic impairment of PAR2-mediated vasodilation: prevention by inhibition of aortic endothelial sodium-glucose-co-transporter-2 and minimizing oxidative stress. Vascul Pharmacol. 2018; 109:56-71.
  48. Lambert R, Srodulski S, Peng X, et al. SGLT2 inhibition does not acutely affect cardiac contractility or metabolism. Cardiovasc Diabetol. 2018;17(1):140.
  49. Santos-Gallego CG, Requena-Ibanez JA, San Antonio R, et al. Empagliflozin ameliorates adverse left ventricular remodeling in nondiabetic heart failure by enhancing myocardial energetics. J Am Coll Cardiol. 2019;73(15):1931-1944.
  50. Yurista SR, Silljé HHW, Oberdorf-Maass SU, et al. Sodium-glucose co-transporter 2 inhibition with empagliflozin improves cardiac function in non-diabetic rats with left ventricular dysfunction after myocardial infarction. Eur J Heart Fail. 2019;21(7):862-873.
  51. Byrne NJ, Parajuli N, Levasseur JL, et al. Empagliflozin prevents worsening of cardiac function in an experimental model of pressure overload-induced heart failure. JACC Basic Transl Sci. 2017;2(4):347-354.
  52. Li X, Lu Q, Qiu Y, et al. Direct cardiac actions of the sodium glucose co-transporter 2 inhibitor empagliflozin improve myocardial oxidative phosphorylation and attenuate pressure-overload heart failure. J Am Heart Assoc. 2021;10(11):e019708.
  53. Hawley SA, Ford RJ, Smith BK, et al. The Na+/glucose cotransporter inhibitor canagliflozin activates AMPK by inhibiting mitochondrial function and increasing cellular AMP levels. Diabetes. 2016;65(9):2784-2794.
  54. Zhou H, Wang S, Zhu P, et al. Empagliflozin rescues diabetic myocardial microvascular injury via AMPK-mediated inhibition of mitochondrial fission. Redox Biol. 2018; 15:335-346.
  55. Deng Y, Xie M, Li Q, et al. Targeting mitochondria-inflammation circuit by β-hydroxybutyrate mitigates HFpEF. Circ Res. 2021;128(2):232-245.
  56. Garcia D, Shaw RJ. AMPK: mechanisms of cellular energy sensing and restoration of metabolic balance. Mol Cell. 2017;66(6):789-800.
  57. Packer M. Interplay of adenosine monophosphate-activated protein kinase/sirtuin-1 activation and sodium influx inhibition mediates the renal benefits of sodium-glucose co-transporter-2 inhibitors in type 2 diabetes: a novel conceptual framework. Diabetes Obes Metab. 2020;22(5):734-742.
  58. Packer M. Autophagy stimulation and intracellular sodium reduction as mediators of the cardioprotective effect of sodium-glucose cotransporter 2 inhibitors. Eur J Heart Fail. 2020;22(4):618-628.
  59. Xie Y, Wei Y, Shen Y, et al. SGLT2 inhibitor dapagliflozin improves diabetic nephropathy by normalizing the aberrant autophagy response to nutrient excess in podocytes. Autophagy. 2019;15(8):1395-1404.
  60. Packer M. Critical reanalysis of the mechanisms underlying the cardiorenal benefits of SGLT2 inhibitors and reaffirmation of the nutrient deprivation signaling/autophagy hypothesis. Circulation. 2022;146(9):684-704.
  61. Mizuno M, Kuno A, Yano T, et al. Empagliflozin normalizes the size and number of mitochondria and prevents reduction in mitochondrial size after myocardial infarction in diabetic hearts. Physiol Rep. 2018;6(12): e13741.
  62. Uthman L, Baartscheer A, Schumacher CA, et al. Direct cardiac effects of dapagliflozin in isolated perfused hearts from lean and obese rats and in isolated cardiomyocytes. Cardiovasc Diabetol. 2018;17(1):86.
  63. Tang L, Wu Y, Tian M, et al. Dapagliflozin slows the progression of the renal and liver fibrosis associated with type 2 diabetes. Am J Physiol Endocrinol Metab. 2017;313(5): E563-E573.
  64. Steven S, Oelze M, Hanf A, et al. The SGLT2 inhibitor empagliflozin improves the primary diabetic complications in ZDF rats. Redox Biol. 2017; 13:370-385.
  65. Aroor AR, Das NA, Carpenter AJ, et al. Glycemic control by the SGLT2 inhibitor empagliflozin decreases aortic stiffness, renal resistivity index and kidney injury. Cardiovasc Diabetol. 2018;17(1):108.
  66. Pabel S, Wagner S, Bollenberg H, et al. Empagliflozin directly improves diastolic function in human heart failure. Eur J Heart Fail. 2018;20(12):1690-1700.
  67. Durak A, Olgar Y, Degirmenci S, et al. A SGLT2 inhibitor dapagliflozin suppresses prolonged ventricular-repolarization through augmentation of voltage-gated K+ channel currents. Cardiovasc Diabetol. 2018;17(1):144.
  68. Connelly KA, Zhang Y, Desjardins JF, et al. Load-independent effects of empagliflozin contribute to improved diastolic function in experimental diabetes mellitus. Cardiovasc Diabetol. 2019;18(1):6.
  69. Osataphan S, Macchi C, Singhal G, et al. SGLT2 inhibition reprograms glucose metabolism and glycogen turnover in the liver and improves cyclic glucose tolerance. Diabetes. 2019;68(4):743-753.
  70. Rajasekeran H, Reich HN, Hladunewich MA, et al. Dapagliflozin in focal segmental glomerulosclerosis: a combined human-rodent pilot study. Am J Physiol Renal Physiol. 2018;314(3): F412-F420.
  71. Ridker PM, Everett BM, Thuren T, et al. Anti-inflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377(12):1119-1131.
  72. Guo H, Chen Y, Liao L, Wu W. Resveratrol protects HUVECs from oxidized-LDL induced oxidative damage by autophagy upregulation via the AMPK/SIRT1 pathway. Cardiovasc Drugs Ther. 2013;27(3):189-198.
  73. Paolisso P, Bergamaschi L, Gragnano F, et al. Outcomes in diabetic patients treated with SGLT2-Inhibitors with acute myocardial infarction undergoing PCI: The SGLT2-I AMI PROTECT Study. Pharmacol Res. 2023; 187:106597.
  74. Zelniker TA, Wiviott SD, Raz I, et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet. 2019;393(10166):31-39.
  75. McGuire DK, Shih WJ, Cosentino F, et al. Association of SGLT2 inhibitors with cardiovascular and kidney outcomes in patients with type 2 diabetes: a meta-analysis. JAMA Cardiol. 2021;6(2):148-158.
  76. Salah HM, Al'Aref SJ, Khan MS, et al. Effect of sodium-glucose cotransporter 2 inhibitors on cardiovascular and kidney outcomes-systematic review and meta-analysis of randomized placebo-controlled trials. Am Heart J. 2021; 232:10-22.
  77. Solomon SD, McMurray JJV, Anand IS, et al. Angiotensin-neprilysin inhibition in heart failure with preserved ejection fraction. N Engl J Med. 2019;381(17):1609-1620.
  78. Docherty KF, McMurray JJV. DAPA-HF and EMPEROR-Reduced: implications for clinical practice. Cardiovasc Drugs Ther. 2021;35(5):895-907.
  79. Butler J, Packer M, Filippatos G, et al. Effect of empagliflozin in patients with heart failure across the spectrum of kidney function: subgroup analysis of EMPEROR-Reduced and EMPEROR-Preserved. Lancet Diabetes Endocrinol. 2022;10(7):473-484.
  80. Heerspink HJL, Stefánsson BV, Correa-Rotter R, et al. Dapagliflozin in patients with chronic kidney disease. N Engl J Med. 2020;383(15):1436-1446.
  81. Perkovic V, Jardine MJ, Neal B, et al. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med. 2019;380(24):2295-2306.
  82. The EMPA-KIDNEY Collaborative Group. Empagliflozin in patients with chronic kidney disease. N Engl J Med. 2023;388(2):117-127.
  83. Bolinder J, Ljunggren Ö, Kullberg J, et al. Effects of dapagliflozin on body weight, total fat mass, and regional adipose tissue distribution in patients with type 2 diabetes mellitus with inadequate glycemic control on metformin. J Clin Endocrinol Metab. 2012;97(3):1020-1031.
  84. Cefalu WT, Leiter LA, Yoon KH, et al. Efficacy and safety of canagliflozin versus glimepiride in patients with type 2 diabetes inadequately controlled with metformin (CANTATA-SU): 52-week results from a randomised, double-blind, phase 3 non-inferiority trial. Lancet. 2013;382(9896):941-950.
  85. Matthews DR, Li Q, Perkovic V, et al. Effects of canagliflozin on amputation risk in type 2 diabetes: the CANVAS program. Diabetologia. 2019;62(6):926-938.
  86. Zhou Z, Jardine M, Perkovic V, et al. Canagliflozin and fracture risk in individuals with type 2 diabetes: results from the CANVAS Program. Diabetologia. 2019;62(10):1854-1867.
  87. Mahaffey KW, Jardine MJ, Bompoint S, et al. Canagliflozin and cardiovascular and renal outcomes in type 2 diabetes mellitus and chronic kidney disease in primary and secondary cardiovascular prevention groups. Circulation. 2019;140(9):739-750.
  88. Weber MA, Mansfield TA, Cain VA, et al. Blood pressure and glycaemic effects of dapagliflozin versus placebo in patients with type 2 diabetes on combination antihyperglycaemic therapy: a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Diabetes Endocrinol. 2016;4(3):211-220.
  89. Chilton R, Tikkanen I, Cannon CP, et al. 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(12):1180-1193.
  90. Yurista SR, Chong CR, Badimon JJ, et al. Therapeutic potential of ketone bodies for patients with cardiovascular disease: JACC state-of-the-art review. J Am Coll Cardiol. 2021;77(13):1660-1669.
  91. Brahma MK, Pepin ME, Wende AR. My sweetheart is broken: role of glucose in diabetic cardiomyopathy. Diabetes Metab J. 2017;41(1):1-9.
  92. Nielsen R, Møller N, Gormsen LC, et al. Cardiovascular effects of treatment with the ketone body 3-hydroxybutyrate in chronic heart failure patients. Circulation. 2019;139(18):2129-2141.
  93. Spertus JA, Birmingham MC, Nassif M, et al. The SGLT2 inhibitor canagliflozin in heart failure: the CHIEF-HF remote, patient-centered randomized trial. Nat Med. 2022;28(4):809-813.
  94. Kosiborod MN, Jhund PS, Docherty KF, et al. Effects of dapagliflozin on symptoms, function, and quality of life in patients with heart failure and reduced ejection fraction: results from the DAPA-HF trial. Circulation. 2020;141(2):90-99.
  95. Heise T, Seman L, Macha S, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of multiple rising doses of empagliflozin in patients with type 2 diabetes mellitus. Diabetes Ther. 2013;4(2):331-345.
  96. Liang Y, Arakawa K, Ueta K, et al. Effect of canagliflozin on renal threshold for glucose, glycemia, and body weight in normal and diabetic animal models. PLoS One. 2012;7(2):e30555.
  97. Rosenstock J, Hansen L, Zee P, et al. Dual add-on therapy in type 2 diabetes poorly controlled with metformin monotherapy: a randomized double-blind trial of saxagliptin plus dapagliflozin addition versus single addition of saxagliptin or dapagliflozin to metformin. Diabetes Care. 2015;38(3):376-383.
  98. Mathieu C, Ranetti AE, Li D, et al. Randomized, double-blind, phase 3 trial of triple therapy with dapagliflozin add-on to saxagliptin plus metformin in type 2 diabetes. Diabetes Care. 2015;38(11):2009-2017.
  99. Kadowaki T, Nangaku M, Hantel S, et al. Empagliflozin and kidney outcomes in Asian patients with type 2 diabetes and established cardiovascular disease: results from the EMPA-REG OUTCOME trial. J Diabetes Investig. 2019;10(3):760-770.
  100. Cherney DZI, Zinman B, Inzucchi SE, et al. Effects of empagliflozin on the urinary albumin-to-creatinine ratio in patients with type 2 diabetes and established cardiovascular disease: an exploratory analysis from the EMPA-REG OUTCOME randomised, placebo-controlled trial. Lancet Diabetes Endocrinol. 2017;5(8):610-621.
  101. Chua KP, Conti RM. Use and costs of diabetes medications, 2003-2017. JAMA Intern Med. 2020;180(12):1576-1577.
  102. Adler AI, McEwan P, Lamotte M, et al. Assessing the long-term cost-effectiveness of empagliflozin in patients with type 2 diabetes in the UK setting. Diabetes Ther. 2020;11(9):2025-2042.
  103. Januzzi JL Jr, Xu J, Li J, et al. Effects of canagliflozin on cardiovascular biomarkers in older adults with type 2 diabetes. J Am Coll Cardiol. 2017;70(6):704-712.
  104. Fitchett D, Inzucchi SE, Cannon CP, et al. Empagliflozin reduced mortality and hospitalization for heart failure across the spectrum of cardiovascular risk in the EMPA-REG OUTCOME trial. Circulation. 2019;139(11):1384-1395.
  105. Takasu T, Yokono M, Tahara A, et al. Effect of ipragliflozin, an SGLT2 inhibitor, on cardiac histopathological changes in type 2 diabetic db/db mice. Life Sci. 2017; 169:73-79.
  106. Cappetta D, De Angelis A, Sapio L, et al. Diabetic cardiomyopathy: the response of cardiac progenitor cells and its therapeutic implications. Cardiovasc Diabetol. 2016;15(1):134.
  107. Iorga A, Cunningham CM, Moazeni S, et al. The protective role of estrogen and estrogen receptors in cardiovascular disease and the controversial use of estrogen therapy. Biol Sex Differ. 2017;8(1):33.
  108. Chen J, Williams S, Ho S, et al. Quantitative PCR tissue distribution of human glucose transporter 1 (GLUT1) mRNA. Biochem Biophys Res Commun. 2001;284(2):399-408.
  109. Kern M, Klöting N, Niessen HG, et al. Linagliptin improves insulin sensitivity and hepatic steatosis in diet-induced obesity. PLoS One. 2012;7(6): e38744.
  110. Esterline R, Vaag A, Oscarsson J, Vora J. Mechanisms in endocrinology: SGLT2 inhibitors: clinical benefits by restoration of normal diurnal metabolism? Eur J Endocrinol. 2018;178(4): R113-R125.
  111. Nasiri-Ansari N, Androutsakos T, Flessa CM, et al. Endothelial function and arterial stiffness in metabolic syndrome and diabetes - impact of the Mediterranean diet. Nutrients. 2020;12(12):3745.
  112. Wu P, Wen W, Li J, et al. Systematic review and meta-analysis of randomized controlled trials on the effect of SGLT2 inhibitors on blood pressure in patients with type 2 diabetes mellitus. Cardiovasc Diabetol. 2020;19(1):27.
  113. Xu B, Li S, Kang B, Zhou J. The current role of sodium-glucose cotransporter 2 inhibitors in type 2 diabetes mellitus management. Cardiovasc Diabetol. 2022;21(1):83.
  114. Abdul-Ghani M, Del Prato S, Chilton R, DeFronzo RA. SGLT2 inhibitors and cardiovascular risk: lessons learned from the EMPA-REG OUTCOME study. Diabetes Care. 2016;39(5):717-725.
  115. Stubbs BJ, Cox PJ, Evans RD, et al. On the metabolism of exogenous ketones in humans. Front Physiol. 2017; 8:848.
  116. Puchalska P, Crawford PA. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. 2017;25(2):262-284.
  117. Newman JC, Verdin E. β-hydroxybutyrate: a signaling metabolite. Annu Rev Nutr. 2017; 37:51-76.
  118. Powell DR, Smith M, Greer J, et al. LX4211 increases serum glucagon-like peptide 1 and peptide YY levels by reducing sodium/glucose cotransporter 1 (SGLT1)-mediated absorption of intestinal glucose. J Pharmacol Exp Ther. 2013;345(2):250-259.
  119. Lapuerta P, Zambrowicz B, Strumph P, Sands A. Development of sotagliflozin, a dual sodium-dependent glucose transporter 1/2 inhibitor. Diab Vasc Dis Res. 2015;12(2):101-110.
  120. Viollet B, Horman S, Leclerc J, et al. AMPK inhibition in health and disease. Crit Rev Biochem Mol Biol. 2010;45(4):276-295.
  121. Karmazyn M, Gan XT, Humphreys RA, Yoshida H, Kusumoto K. The myocardial Na (+)-H (+) exchange: structure, regulation, and its role in heart disease. Circ Res. 1999;85(9):777-786.
  122. Schrauwen P, Schrauwen-Hinderling V, Hoeks J, Hesselink MK. Mitochondrial dysfunction and lipotoxicity. Biochim Biophys Acta. 2010;1801(3):266-271.

Reference

  1. International Diabetes Federation. IDF Diabetes Atlas, 10th edition. Brussels, Belgium: International Diabetes Federation; 2021.
  2. Sarwar N, Gao P, Seshasai SR, et al. Diabetes mellitus, fasting blood glucose concentration, and risk of vascular disease: a collaborative meta-analysis of 102 prospective studies. Lancet. 2010;375(9733):2215-2222.
  3. Einarson TR, Acs A, Ludwig C, Panton UH. Prevalence of cardiovascular disease in type 2 diabetes: a systematic literature review of scientific evidence from across the world in 2007-2017. Cardiovasc Diabetol. 2018;17(1):83.
  4. DeFronzo RA. From the triumvirate to the ominous octet: a new paradigm for the treatment of type 2 diabetes mellitus. Diabetes. 2009;58(4):773-795.
  5. Wright EM, Loo DD, Hirayama BA. Biology of human sodium glucose transporters. Physiol Rev. 2011;91(2):733-794.
  6. Ghezzi C, Loo DDF, Wright EM. Physiology of renal glucose handling via SGLT1, SGLT2 and GLUT2. Diabetologia. 2018;61(10):2087-2097.
  7. Hummel CS, Lu C, Loo DD, et al. Glucose transport by human renal Na+/D-glucose cotransporters SGLT1 and SGLT2. Am J Physiol Cell Physiol. 2011;300(1):C14-21.
  8. Vallon V, Platt KA, Cunard R, et al. SGLT2 mediates glucose reabsorption in the early proximal tubule. J Am Soc Nephrol. 2011;22(1):104-112.
  9. Ghezzi C, Wright EM. Regulation of the human Na+-dependent glucose cotransporter hSGLT2. Am J Physiol Cell Physiol. 2012;303(3):C348-354.
  10. Banerjee SK, McGaffin KR, Pastor-Soler NM, Ahmad F. SGLT1 is a novel cardiac glucose transporter that is perturbed in disease states. Cardiovasc Res. 2009;84(1):111-118.
  11. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373(22):2117-2128.
  12. Neal B, Perkovic V, Mahaffey KW, et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med. 2017;377(7):644-657.
  13. Wiviott SD, Raz I, Bonaca MP, et al. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2019;380(4):347-357.
  14. Cannon CP, Pratley R, Dagogo-Jack S, et al. Cardiovascular outcomes with ertugliflozin in type 2 diabetes. N Engl J Med. 2020;383(15):1425-1435.
  15. McMurray JJV, Solomon SD, Inzucchi SE, et al. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med. 2019;381(21):1995-2008.
  16. Packer M, Anker SD, Butler J, et al. Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med. 2020;383(15):1413-1424.
  17. Anker SD, Butler J, Filippatos G, et al. Empagliflozin in heart failure with a preserved ejection fraction. N Engl J Med. 2021;385(16):1451-1461.
  18. Bailey CJ, Gross JL, Pieters A, et al. Effect of dapagliflozin in patients with type 2 diabetes who have inadequate glycaemic control with metformin: a randomised, double-blind, placebo-controlled trial. Lancet. 2010;375(9733):2223-2233.
  19. Roden M, Weng J, Eilbracht J, et al. Empagliflozin monotherapy with sitagliptin as an active comparator in patients with type 2 diabetes: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Diabetes Endocrinol. 2013;1(3):208-219.
  20. Stenlöf K, Cefalu WT, Kim KA, et al. Efficacy and safety of canagliflozin monotherapy in subjects with type 2 diabetes mellitus inadequately controlled with diet and exercise. Diabetes Obes Metab. 2013;15(4):372-382.
  21. Ferrannini E, Ramos SJ, Salsali A, et al. Dapagliflozin monotherapy in type 2 diabetic patients with inadequate glycemic control by diet and exercise: a randomized, double-blind, placebo-controlled, phase 3 trial. Diabetes Care. 2010;33(10):2217-2224.
  22. Terra SG, Focht K, Davies M, et al. Phase III, efficacy and safety study of ertugliflozin monotherapy in people with type 2 diabetes mellitus inadequately controlled with diet and exercise alone. Diabetes Obes Metab. 2017;19(5):721-728.
  23. Ferrannini E, Muscelli E, Frascerra S, et al. Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic patients. J Clin Invest. 2014;124(2):499-508.
  24. Stanley WC, Recchia FA, Lopaschuk GD. Myocardial substrate metabolism in the normal and failing heart. Physiol Rev. 2005;85(3):1093-1129.
  25. Aubert G, Martin OJ, Horton JL, et al. The failing heart relies on ketone bodies as a fuel. Circulation. 2016;133(8):698-705.
  26. Kappel BA, Lehrke M, Schütt K, et al. Effect of empagliflozin on the metabolic signature of patients with type 2 diabetes mellitus and cardiovascular disease. Circulation. 2017;136(10):969-972.
  27. 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(7):1115-1122.
  28. Ho KL, Zhang L, Wagg C, et al. Increased ketone body oxidation provides additional energy for the failing heart without improving cardiac efficiency. Cardiovasc Res. 2019;115(11):1606-1616.
  29. Verma S, McMurray JJV. SGLT2 inhibitors and mechanisms of cardiovascular benefit: a state-of-the-art review. Diabetologia. 2018;61(10):2108-2117.
  30. Lopaschuk GD, Verma S. Mechanisms of cardiovascular benefits of sodium glucose co-transporter 2 (SGLT2) inhibitors: a state-of-the-art review. JACC Basic Transl Sci. 2020;5(6):632-644.
  31. Ferrannini E, Mark M, Mayoux E. CV protection in the EMPA-REG OUTCOME trial: a "thrifty substrate" hypothesis. Diabetes Care. 2016;39(7):1108-1114.
  32. Lopaschuk GD, Karwi QG, Tian R, et al. Cardiac energy metabolism in heart failure. Circ Res. 2021;128(10):1487-1513.
  33. Brownlee M. The pathobiology of diabetic complications: a unifying mechanism. Diabetes. 2005;54(6):1615-1625.
  34. Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circ Res. 2010;107(9):1058-1070.
  35. Fioretto P, Giaccari A, Sesti G. Efficacy and safety of dapagliflozin, a sodium glucose cotransporter 2 (SGLT2) inhibitor, in diabetes mellitus. Cardiovasc Diabetol. 2015; 14:142.
  36. Baartscheer A, Schumacher CA, Wüst RCI, et al. Empagliflozin decreases myocardial cytoplasmic Na+ through inhibition of the cardiac Na+/H+ exchanger in rats and rabbits. Diabetologia. 2017;60(3):568-573.
  37. Uthman L, Baartscheer A, Bleijlevens B, et al. Class effects of SGLT2 inhibitors in mouse cardiomyocytes and hearts: inhibition of Na+/H+ exchanger, lowering of cytosolic Na+ and vasodilation. Diabetologia. 2018;61(3):722-726.
  38. Trum M, Riechel J, Wagner S, et al. Empagliflozin inhibits Na+/H+ exchanger activity in human atrial cardiomyocytes via a direct interaction. JACC Basic Transl Sci. 2020;5(12):1197-1207.
  39. Pessoa TD, Campos LC, Carraro-Lacroix L, et al. Functional role of glucose metabolism, osmotic stress, and sodium-glucose cotransporter isoform-mediated transport on Na+/H+ exchanger isoform 3 activity in the renal proximal tubule. J Am Soc Nephrol. 2014;25(9):2028-2039.
  40. Xu L, Nagata N, Nagashimada M, et al. SGLT2 inhibition by empagliflozin promotes fat utilization and browning and attenuates inflammation and insulin resistance by polarizing M2 macrophages in diet-induced obese mice. EBioMedicine. 2017; 20:137-149.
  41. Kim SR, Lee SG, Kim SH, et al. SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease. Nat Commun. 2020;11(1):2127.
  42. Heerspink HJL, Perco P, Mulder S, et al. Canagliflozin reduces inflammation and fibrosis biomarkers: a potential mechanism of action for beneficial effects of SGLT2 inhibitors in diabetic kidney disease. Diabetologia. 2019;62(7):1154-1162.
  43. Dekkers CCJ, Petrykiv S, Laverman GD, et al. Effects of the SGLT2 inhibitor dapagliflozin on glomerular and tubular injury markers. Diabetes Obes Metab. 2018;20(8):1988-1993.
  44. Kang S, Verma S, Hassanabad AF, et al. Direct effects of empagliflozin on extracellular matrix remodelling in human cardiac myofibroblasts: novel translational clues to explain EMPA-REG OUTCOME results. Can J Cardiol. 2020;36(4):543-553.
  45. Solini A, Giannini L, Seghieri M, et al. Dapagliflozin acutely improves endothelial dysfunction, reduces aortic stiffness and renal resistive index in type 2 diabetic patients: a pilot study. Cardiovasc Diabetol. 2017;16(1):138.
  46. Juni RP, Kuster DWD, Goebel M, et al. Cardiac microvascular endothelial enhancement of cardiomyocyte function is impaired by inflammation and restored by empagliflozin. JACC Basic Transl Sci. 2019;4(5):575-591.
  47. El-Daly M, Pulakazhi Venu VK, Saifeddine M, et al. Hyperglycaemic impairment of PAR2-mediated vasodilation: prevention by inhibition of aortic endothelial sodium-glucose-co-transporter-2 and minimizing oxidative stress. Vascul Pharmacol. 2018; 109:56-71.
  48. Lambert R, Srodulski S, Peng X, et al. SGLT2 inhibition does not acutely affect cardiac contractility or metabolism. Cardiovasc Diabetol. 2018;17(1):140.
  49. Santos-Gallego CG, Requena-Ibanez JA, San Antonio R, et al. Empagliflozin ameliorates adverse left ventricular remodeling in nondiabetic heart failure by enhancing myocardial energetics. J Am Coll Cardiol. 2019;73(15):1931-1944.
  50. Yurista SR, Silljé HHW, Oberdorf-Maass SU, et al. Sodium-glucose co-transporter 2 inhibition with empagliflozin improves cardiac function in non-diabetic rats with left ventricular dysfunction after myocardial infarction. Eur J Heart Fail. 2019;21(7):862-873.
  51. Byrne NJ, Parajuli N, Levasseur JL, et al. Empagliflozin prevents worsening of cardiac function in an experimental model of pressure overload-induced heart failure. JACC Basic Transl Sci. 2017;2(4):347-354.
  52. Li X, Lu Q, Qiu Y, et al. Direct cardiac actions of the sodium glucose co-transporter 2 inhibitor empagliflozin improve myocardial oxidative phosphorylation and attenuate pressure-overload heart failure. J Am Heart Assoc. 2021;10(11):e019708.
  53. Hawley SA, Ford RJ, Smith BK, et al. The Na+/glucose cotransporter inhibitor canagliflozin activates AMPK by inhibiting mitochondrial function and increasing cellular AMP levels. Diabetes. 2016;65(9):2784-2794.
  54. Zhou H, Wang S, Zhu P, et al. Empagliflozin rescues diabetic myocardial microvascular injury via AMPK-mediated inhibition of mitochondrial fission. Redox Biol. 2018; 15:335-346.
  55. Deng Y, Xie M, Li Q, et al. Targeting mitochondria-inflammation circuit by β-hydroxybutyrate mitigates HFpEF. Circ Res. 2021;128(2):232-245.
  56. Garcia D, Shaw RJ. AMPK: mechanisms of cellular energy sensing and restoration of metabolic balance. Mol Cell. 2017;66(6):789-800.
  57. Packer M. Interplay of adenosine monophosphate-activated protein kinase/sirtuin-1 activation and sodium influx inhibition mediates the renal benefits of sodium-glucose co-transporter-2 inhibitors in type 2 diabetes: a novel conceptual framework. Diabetes Obes Metab. 2020;22(5):734-742.
  58. Packer M. Autophagy stimulation and intracellular sodium reduction as mediators of the cardioprotective effect of sodium-glucose cotransporter 2 inhibitors. Eur J Heart Fail. 2020;22(4):618-628.
  59. Xie Y, Wei Y, Shen Y, et al. SGLT2 inhibitor dapagliflozin improves diabetic nephropathy by normalizing the aberrant autophagy response to nutrient excess in podocytes. Autophagy. 2019;15(8):1395-1404.
  60. Packer M. Critical reanalysis of the mechanisms underlying the cardiorenal benefits of SGLT2 inhibitors and reaffirmation of the nutrient deprivation signaling/autophagy hypothesis. Circulation. 2022;146(9):684-704.
  61. Mizuno M, Kuno A, Yano T, et al. Empagliflozin normalizes the size and number of mitochondria and prevents reduction in mitochondrial size after myocardial infarction in diabetic hearts. Physiol Rep. 2018;6(12): e13741.
  62. Uthman L, Baartscheer A, Schumacher CA, et al. Direct cardiac effects of dapagliflozin in isolated perfused hearts from lean and obese rats and in isolated cardiomyocytes. Cardiovasc Diabetol. 2018;17(1):86.
  63. Tang L, Wu Y, Tian M, et al. Dapagliflozin slows the progression of the renal and liver fibrosis associated with type 2 diabetes. Am J Physiol Endocrinol Metab. 2017;313(5): E563-E573.
  64. Steven S, Oelze M, Hanf A, et al. The SGLT2 inhibitor empagliflozin improves the primary diabetic complications in ZDF rats. Redox Biol. 2017; 13:370-385.
  65. Aroor AR, Das NA, Carpenter AJ, et al. Glycemic control by the SGLT2 inhibitor empagliflozin decreases aortic stiffness, renal resistivity index and kidney injury. Cardiovasc Diabetol. 2018;17(1):108.
  66. Pabel S, Wagner S, Bollenberg H, et al. Empagliflozin directly improves diastolic function in human heart failure. Eur J Heart Fail. 2018;20(12):1690-1700.
  67. Durak A, Olgar Y, Degirmenci S, et al. A SGLT2 inhibitor dapagliflozin suppresses prolonged ventricular-repolarization through augmentation of voltage-gated K+ channel currents. Cardiovasc Diabetol. 2018;17(1):144.
  68. Connelly KA, Zhang Y, Desjardins JF, et al. Load-independent effects of empagliflozin contribute to improved diastolic function in experimental diabetes mellitus. Cardiovasc Diabetol. 2019;18(1):6.
  69. Osataphan S, Macchi C, Singhal G, et al. SGLT2 inhibition reprograms glucose metabolism and glycogen turnover in the liver and improves cyclic glucose tolerance. Diabetes. 2019;68(4):743-753.
  70. Rajasekeran H, Reich HN, Hladunewich MA, et al. Dapagliflozin in focal segmental glomerulosclerosis: a combined human-rodent pilot study. Am J Physiol Renal Physiol. 2018;314(3): F412-F420.
  71. Ridker PM, Everett BM, Thuren T, et al. Anti-inflammatory therapy with canakinumab for atherosclerotic disease. N Engl J Med. 2017;377(12):1119-1131.
  72. Guo H, Chen Y, Liao L, Wu W. Resveratrol protects HUVECs from oxidized-LDL induced oxidative damage by autophagy upregulation via the AMPK/SIRT1 pathway. Cardiovasc Drugs Ther. 2013;27(3):189-198.
  73. Paolisso P, Bergamaschi L, Gragnano F, et al. Outcomes in diabetic patients treated with SGLT2-Inhibitors with acute myocardial infarction undergoing PCI: The SGLT2-I AMI PROTECT Study. Pharmacol Res. 2023; 187:106597.
  74. Zelniker TA, Wiviott SD, Raz I, et al. SGLT2 inhibitors for primary and secondary prevention of cardiovascular and renal outcomes in type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials. Lancet. 2019;393(10166):31-39.
  75. McGuire DK, Shih WJ, Cosentino F, et al. Association of SGLT2 inhibitors with cardiovascular and kidney outcomes in patients with type 2 diabetes: a meta-analysis. JAMA Cardiol. 2021;6(2):148-158.
  76. Salah HM, Al'Aref SJ, Khan MS, et al. Effect of sodium-glucose cotransporter 2 inhibitors on cardiovascular and kidney outcomes-systematic review and meta-analysis of randomized placebo-controlled trials. Am Heart J. 2021; 232:10-22.
  77. Solomon SD, McMurray JJV, Anand IS, et al. Angiotensin-neprilysin inhibition in heart failure with preserved ejection fraction. N Engl J Med. 2019;381(17):1609-1620.
  78. Docherty KF, McMurray JJV. DAPA-HF and EMPEROR-Reduced: implications for clinical practice. Cardiovasc Drugs Ther. 2021;35(5):895-907.
  79. Butler J, Packer M, Filippatos G, et al. Effect of empagliflozin in patients with heart failure across the spectrum of kidney function: subgroup analysis of EMPEROR-Reduced and EMPEROR-Preserved. Lancet Diabetes Endocrinol. 2022;10(7):473-484.
  80. Heerspink HJL, Stefánsson BV, Correa-Rotter R, et al. Dapagliflozin in patients with chronic kidney disease. N Engl J Med. 2020;383(15):1436-1446.
  81. Perkovic V, Jardine MJ, Neal B, et al. Canagliflozin and renal outcomes in type 2 diabetes and nephropathy. N Engl J Med. 2019;380(24):2295-2306.
  82. The EMPA-KIDNEY Collaborative Group. Empagliflozin in patients with chronic kidney disease. N Engl J Med. 2023;388(2):117-127.
  83. Bolinder J, Ljunggren Ö, Kullberg J, et al. Effects of dapagliflozin on body weight, total fat mass, and regional adipose tissue distribution in patients with type 2 diabetes mellitus with inadequate glycemic control on metformin. J Clin Endocrinol Metab. 2012;97(3):1020-1031.
  84. Cefalu WT, Leiter LA, Yoon KH, et al. Efficacy and safety of canagliflozin versus glimepiride in patients with type 2 diabetes inadequately controlled with metformin (CANTATA-SU): 52-week results from a randomised, double-blind, phase 3 non-inferiority trial. Lancet. 2013;382(9896):941-950.
  85. Matthews DR, Li Q, Perkovic V, et al. Effects of canagliflozin on amputation risk in type 2 diabetes: the CANVAS program. Diabetologia. 2019;62(6):926-938.
  86. Zhou Z, Jardine M, Perkovic V, et al. Canagliflozin and fracture risk in individuals with type 2 diabetes: results from the CANVAS Program. Diabetologia. 2019;62(10):1854-1867.
  87. Mahaffey KW, Jardine MJ, Bompoint S, et al. Canagliflozin and cardiovascular and renal outcomes in type 2 diabetes mellitus and chronic kidney disease in primary and secondary cardiovascular prevention groups. Circulation. 2019;140(9):739-750.
  88. Weber MA, Mansfield TA, Cain VA, et al. Blood pressure and glycaemic effects of dapagliflozin versus placebo in patients with type 2 diabetes on combination antihyperglycaemic therapy: a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Diabetes Endocrinol. 2016;4(3):211-220.
  89. Chilton R, Tikkanen I, Cannon CP, et al. 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(12):1180-1193.
  90. Yurista SR, Chong CR, Badimon JJ, et al. Therapeutic potential of ketone bodies for patients with cardiovascular disease: JACC state-of-the-art review. J Am Coll Cardiol. 2021;77(13):1660-1669.
  91. Brahma MK, Pepin ME, Wende AR. My sweetheart is broken: role of glucose in diabetic cardiomyopathy. Diabetes Metab J. 2017;41(1):1-9.
  92. Nielsen R, Møller N, Gormsen LC, et al. Cardiovascular effects of treatment with the ketone body 3-hydroxybutyrate in chronic heart failure patients. Circulation. 2019;139(18):2129-2141.
  93. Spertus JA, Birmingham MC, Nassif M, et al. The SGLT2 inhibitor canagliflozin in heart failure: the CHIEF-HF remote, patient-centered randomized trial. Nat Med. 2022;28(4):809-813.
  94. Kosiborod MN, Jhund PS, Docherty KF, et al. Effects of dapagliflozin on symptoms, function, and quality of life in patients with heart failure and reduced ejection fraction: results from the DAPA-HF trial. Circulation. 2020;141(2):90-99.
  95. Heise T, Seman L, Macha S, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of multiple rising doses of empagliflozin in patients with type 2 diabetes mellitus. Diabetes Ther. 2013;4(2):331-345.
  96. Liang Y, Arakawa K, Ueta K, et al. Effect of canagliflozin on renal threshold for glucose, glycemia, and body weight in normal and diabetic animal models. PLoS One. 2012;7(2):e30555.
  97. Rosenstock J, Hansen L, Zee P, et al. Dual add-on therapy in type 2 diabetes poorly controlled with metformin monotherapy: a randomized double-blind trial of saxagliptin plus dapagliflozin addition versus single addition of saxagliptin or dapagliflozin to metformin. Diabetes Care. 2015;38(3):376-383.
  98. Mathieu C, Ranetti AE, Li D, et al. Randomized, double-blind, phase 3 trial of triple therapy with dapagliflozin add-on to saxagliptin plus metformin in type 2 diabetes. Diabetes Care. 2015;38(11):2009-2017.
  99. Kadowaki T, Nangaku M, Hantel S, et al. Empagliflozin and kidney outcomes in Asian patients with type 2 diabetes and established cardiovascular disease: results from the EMPA-REG OUTCOME trial. J Diabetes Investig. 2019;10(3):760-770.
  100. Cherney DZI, Zinman B, Inzucchi SE, et al. Effects of empagliflozin on the urinary albumin-to-creatinine ratio in patients with type 2 diabetes and established cardiovascular disease: an exploratory analysis from the EMPA-REG OUTCOME randomised, placebo-controlled trial. Lancet Diabetes Endocrinol. 2017;5(8):610-621.
  101. Chua KP, Conti RM. Use and costs of diabetes medications, 2003-2017. JAMA Intern Med. 2020;180(12):1576-1577.
  102. Adler AI, McEwan P, Lamotte M, et al. Assessing the long-term cost-effectiveness of empagliflozin in patients with type 2 diabetes in the UK setting. Diabetes Ther. 2020;11(9):2025-2042.
  103. Januzzi JL Jr, Xu J, Li J, et al. Effects of canagliflozin on cardiovascular biomarkers in older adults with type 2 diabetes. J Am Coll Cardiol. 2017;70(6):704-712.
  104. Fitchett D, Inzucchi SE, Cannon CP, et al. Empagliflozin reduced mortality and hospitalization for heart failure across the spectrum of cardiovascular risk in the EMPA-REG OUTCOME trial. Circulation. 2019;139(11):1384-1395.
  105. Takasu T, Yokono M, Tahara A, et al. Effect of ipragliflozin, an SGLT2 inhibitor, on cardiac histopathological changes in type 2 diabetic db/db mice. Life Sci. 2017; 169:73-79.
  106. Cappetta D, De Angelis A, Sapio L, et al. Diabetic cardiomyopathy: the response of cardiac progenitor cells and its therapeutic implications. Cardiovasc Diabetol. 2016;15(1):134.
  107. Iorga A, Cunningham CM, Moazeni S, et al. The protective role of estrogen and estrogen receptors in cardiovascular disease and the controversial use of estrogen therapy. Biol Sex Differ. 2017;8(1):33.
  108. Chen J, Williams S, Ho S, et al. Quantitative PCR tissue distribution of human glucose transporter 1 (GLUT1) mRNA. Biochem Biophys Res Commun. 2001;284(2):399-408.
  109. Kern M, Klöting N, Niessen HG, et al. Linagliptin improves insulin sensitivity and hepatic steatosis in diet-induced obesity. PLoS One. 2012;7(6): e38744.
  110. Esterline R, Vaag A, Oscarsson J, Vora J. Mechanisms in endocrinology: SGLT2 inhibitors: clinical benefits by restoration of normal diurnal metabolism? Eur J Endocrinol. 2018;178(4): R113-R125.
  111. Nasiri-Ansari N, Androutsakos T, Flessa CM, et al. Endothelial function and arterial stiffness in metabolic syndrome and diabetes - impact of the Mediterranean diet. Nutrients. 2020;12(12):3745.
  112. Wu P, Wen W, Li J, et al. Systematic review and meta-analysis of randomized controlled trials on the effect of SGLT2 inhibitors on blood pressure in patients with type 2 diabetes mellitus. Cardiovasc Diabetol. 2020;19(1):27.
  113. Xu B, Li S, Kang B, Zhou J. The current role of sodium-glucose cotransporter 2 inhibitors in type 2 diabetes mellitus management. Cardiovasc Diabetol. 2022;21(1):83.
  114. Abdul-Ghani M, Del Prato S, Chilton R, DeFronzo RA. SGLT2 inhibitors and cardiovascular risk: lessons learned from the EMPA-REG OUTCOME study. Diabetes Care. 2016;39(5):717-725.
  115. Stubbs BJ, Cox PJ, Evans RD, et al. On the metabolism of exogenous ketones in humans. Front Physiol. 2017; 8:848.
  116. Puchalska P, Crawford PA. Multi-dimensional roles of ketone bodies in fuel metabolism, signaling, and therapeutics. Cell Metab. 2017;25(2):262-284.
  117. Newman JC, Verdin E. β-hydroxybutyrate: a signaling metabolite. Annu Rev Nutr. 2017; 37:51-76.
  118. Powell DR, Smith M, Greer J, et al. LX4211 increases serum glucagon-like peptide 1 and peptide YY levels by reducing sodium/glucose cotransporter 1 (SGLT1)-mediated absorption of intestinal glucose. J Pharmacol Exp Ther. 2013;345(2):250-259.
  119. Lapuerta P, Zambrowicz B, Strumph P, Sands A. Development of sotagliflozin, a dual sodium-dependent glucose transporter 1/2 inhibitor. Diab Vasc Dis Res. 2015;12(2):101-110.
  120. Viollet B, Horman S, Leclerc J, et al. AMPK inhibition in health and disease. Crit Rev Biochem Mol Biol. 2010;45(4):276-295.
  121. Karmazyn M, Gan XT, Humphreys RA, Yoshida H, Kusumoto K. The myocardial Na (+)-H (+) exchange: structure, regulation, and its role in heart disease. Circ Res. 1999;85(9):777-786.
  122. Schrauwen P, Schrauwen-Hinderling V, Hoeks J, Hesselink MK. Mitochondrial dysfunction and lipotoxicity. Biochim Biophys Acta. 2010;1801(3):266-271.

Photo
Rushikesh Bhosle
Corresponding author

Pes Modern College of Pharmacy Nigdi Pune 44

Photo
Darshil Ingale
Co-author

Pes Modern College of Pharmacy Nigdi Pune 44

Photo
Sachin Amrutkar
Co-author

Pes Modern College of Pharmacy Nigdi Pune 44

Photo
Swaraj Dhande
Co-author

Pes Modern College of Pharmacy Nigdi Pune 44

Photo
Shreyas Kurhe
Co-author

Pes Modern College of Pharmacy Nigdi Pune 44

Rushikesh Bhosle*, Darshil Ingale, Shreyas Kurhe, Swaraj Dhande, Sachin Amrutkar, Molecular Mechanisms of SGLT2 Inhibitors Bridging Glucose Lowering and Cardiac Protection: A Comprehensive Review, Int. J. Sci. R. Tech., 2025, 2 (9), 234-246. https://doi.org/10.5281/zenodo.17190566

More related articles
Transdermal Drug Delivery Systems in Diabetes Mana...
Amin Shaikh, Pratiksha Ravankole, Priyanka Konde, Fiza Mulani, ...
Floristic Diversity of Kansari Mavli Sacred Grove ...
Kainisha Gamit, Anusha Maitreya, Himanshu Pandya , Hitesh Solanki...
A Review on Sacred Groves of South Gujarat, India...
Kainisha Gamit, Anusha Maitreya, Himanshu Pandya , Hitesh Solanki...
Related Articles
Land Accumulation and Concentration for the Development of High-Tech Agriculture...
Nguyen Mau Dung, Nguyen Duc Minh Tu, Nguyen Phuong Phuong, ...
Modern Therapeutic Approach towards Schizophrenia: A Comprehensive Review of Con...
Deep Jyoti Shah, Mahesh Kumar Yadav, Pintu Kumar, Love Mahato, Kush Mahato, Dulari Majhi, Aditya Raj...
Transdermal Drug Delivery Systems in Diabetes Management: A Review...
Amin Shaikh, Pratiksha Ravankole, Priyanka Konde, Fiza Mulani, ...
More related articles
Transdermal Drug Delivery Systems in Diabetes Management: A Review...
Amin Shaikh, Pratiksha Ravankole, Priyanka Konde, Fiza Mulani, ...
Floristic Diversity of Kansari Mavli Sacred Grove in Songadh Forest Tapi Distric...
Kainisha Gamit, Anusha Maitreya, Himanshu Pandya , Hitesh Solanki , ...
A Review on Sacred Groves of South Gujarat, India...
Kainisha Gamit, Anusha Maitreya, Himanshu Pandya , Hitesh Solanki , ...
Transdermal Drug Delivery Systems in Diabetes Management: A Review...
Amin Shaikh, Pratiksha Ravankole, Priyanka Konde, Fiza Mulani, ...
Floristic Diversity of Kansari Mavli Sacred Grove in Songadh Forest Tapi Distric...
Kainisha Gamit, Anusha Maitreya, Himanshu Pandya , Hitesh Solanki , ...
A Review on Sacred Groves of South Gujarat, India...
Kainisha Gamit, Anusha Maitreya, Himanshu Pandya , Hitesh Solanki , ...