Abstract

Sodium-glucose co-transporter 2 (SGLT2) inhibitors have been shown to improve cardiovascular outcomes not only in patients with diabetes but also in those with heart failure, irrespective of diabetic status. However, the mechanisms underlying the cardioprotective effects of these newer anti-diabetic drugs remain to be fully elucidated. One exciting avenue that has been recently explored in both preclinical and clinical studies is the modulation of the cardiovascular autonomic nervous system. A reduction in sympathetic nervous system activity by SGLT2 inhibitors may potentially translate into a reduction in arrhythmic risk and sudden arrhythmic death, which may explain, at least partly, the cardioprotection shown in the cardiovascular outcome trials with different SGLT2 inhibitors. Although some of the data from the preclinical and clinical studies are promising, overall the findings can be contradictory. This highlights the need for more studies to address gaps in our knowledge of these novel drugs. The present review offers an in depth overview of the existing literature regarding the role of SGLT2 inhibitors in modulating cardiovascular autonomic function as one of the possible pathways of their cardioprotective effects.

Introduction

The development of sodium-glucose co-transporter 2 (SGLT2) inhibitors (SGLT2i; gliflozins) has been a fascinating story in recent therapeutics, which garnered worldwide attention due to the clinically impactful results that have been recognized since the publication of the first SGLT2 inhibitor cardiovascular outcome trial (CVOT) in 2015 (EMPA-REG OUTCOME).1 The EMPA-REG OUTCOME study was a multicentre randomized controlled trial which showed that patients with Type 2 diabetes mellitus (T2DM) at high risk for cardiovascular events who received empagliflozin had a lower rate of cardiovascular mortality, hospitalization for heart failure and all-cause mortality. Notably, the CVOTs in patients with T2DM were introduced and are designed with the specific objective to demonstrate the cardiovascular safety of any new anti-diabetic drug as mandated by both the US Food and Drug Administration (FDA) and the European Medicines Agency (EMA).2,3 Remarkably, the unexpected positive findings from EMPA-REG OUTCOME were similarly shown in subsequent CVOTs with canagliflozin and dapagliflozin (CANVAS in 20174 and DECLARE-TIMI58 in 2018,5 respectively) in patients with T2DM, suggesting a class effect which extends beyond blood glucose control. Moreover, two further SGLT2 inhibitor trials in 2019 demonstrated the benefit of these drugs in patients with chronic kidney disease and T2DM (CREDENCE6), and heart failure with reduced ejection fraction patients regardless of diabetes status (DAPA-HF7).

As SGLT2 receptors are predominantly expressed in the kidney and not the heart,8,9 it is has been shown that SGLT2 inhibitor mediated cardioprotection occurs in an off-target manner.10–13 As such, multiple mechanistic preclinical studies have been conducted, whilst clinical studies are also ongoing in an attempt to tease out the exact mechanistic pathways that may be responsible for the cardioprotection benefits observed in the aforementioned CVOTs. Indeed, the improved cardiovascular outcomes in the SGLT2 inhibitor CVOTs were driven by a reduction in hospitalization for heart failure, but the reasons behind this are unclear. An interesting observation from these trials was that the improvement in blood pressure occurred without compensatory improvement in heart rate, suggesting dampening of sympathetic nervous system (SNS) activity by SGLT2 inhibitors.1,4,5,7 This was similarly shown in experimental studies where luseogliflozin significantly decreased blood pressure without any alterations in heart rate in hypertensive rats.14 Since autonomic dysfunction (characterized by hyperactivity of the SNS) and heart failure can both occur with diabetes, and this is linked with increased mortality,15,16 it is possible that modulation of the cardiovascular autonomic function by SGLT2 inhibitors may account for the reduction in hospitalization for heart failure shown in the trials with different SGLT2 inhibitors. Furthermore, drugs that mitigate SNS activity, such as beta-blockers, have also been shown to improve outcomes in heart failure patients.17–20

This review article explores in depth the existing preclinical and clinical studies focused on the role of SGLT2 inhibitors in modulating cardiovascular autonomic function as one of the possible pathways mediating their cardioprotective effects. A summary of the key findings from the preclinical and clinical studies investigating the role of SGLT2 inhibitors in modulating cardiovascular autonomic function is also shown in Table 1.

Table 1

Summary of key findings from preclinical and clinical studies investigating the role of sodium-glucose co-transporter 2 (SGLT2) inhibitors (SGLT2i; gliflozins) in modulating cardiovascular autonomic function as one of the possible pathways of their cardioprotective effects

Preclinical studies
ReferencesDrug and experimental model(s)Main findings
Chiba et al.21Dapagliflozin, C57BL/6 miceSuppressed noradrenaline turnover, oxygen consumption and ucp1 (a thermogenesis-related gene) expression in brown adipose tissue
Yoshikawa et al.22Iprafliflozin, streptozotocin-induced diabetes Sprague-Dawley ratsImproved baroreflex sensitivity and amelioration of arterial blood pressure lability
Matthews et al.24
  • Noradrenaline, human kidney cells

  • Dapagliflozin, high-fat diet-fed mice

  • Increased SGLT2 and IL-6 expression

  • Significant glucosuria, reduced elevation of tyrosine hydroxylase and noradrenaline in the heart and kidney

Wan et al.25Empagliflozin, salt-treated obese and metabolic syndrome ratsDecreased blood pressure, normalized circadian rhythm of blood pressure and sympathetic nervous activity, increased urinary sodium excretion
Herat et al.26
  • Chemical denervation

  • Dapagliflozin, neurogenic hypertensive Schlager mouse

  • Reduced blood pressure, reduced renal SGLT2 protein expression

  • Reduced elevation of tyrosine hydroxylase and norepinephrine, protects against endothelial dysfunction, reduced inflammation

Preclinical studies
ReferencesDrug and experimental model(s)Main findings
Chiba et al.21Dapagliflozin, C57BL/6 miceSuppressed noradrenaline turnover, oxygen consumption and ucp1 (a thermogenesis-related gene) expression in brown adipose tissue
Yoshikawa et al.22Iprafliflozin, streptozotocin-induced diabetes Sprague-Dawley ratsImproved baroreflex sensitivity and amelioration of arterial blood pressure lability
Matthews et al.24
  • Noradrenaline, human kidney cells

  • Dapagliflozin, high-fat diet-fed mice

  • Increased SGLT2 and IL-6 expression

  • Significant glucosuria, reduced elevation of tyrosine hydroxylase and noradrenaline in the heart and kidney

Wan et al.25Empagliflozin, salt-treated obese and metabolic syndrome ratsDecreased blood pressure, normalized circadian rhythm of blood pressure and sympathetic nervous activity, increased urinary sodium excretion
Herat et al.26
  • Chemical denervation

  • Dapagliflozin, neurogenic hypertensive Schlager mouse

  • Reduced blood pressure, reduced renal SGLT2 protein expression

  • Reduced elevation of tyrosine hydroxylase and norepinephrine, protects against endothelial dysfunction, reduced inflammation

Clinical studies
ReferencesCohort detailsMain findings
  • EMPA-HEART CardioLink-6 Holter analysis study

  • Garg et al.30

n = 66, T2DM patients with established CAD from the EMPA-HEART CardioLink-6 study who had completed Holter monitor recordings at baseline and 6 monthsNo significant difference in HRV parameters between empagliflozin and placebo
  • EMBODY trial

  • Shimizu et al.35

n = 105, T2DM patients at 2 weeks post-AMI enrolled into a randomized double-blind study, empagliflozin vs. placeboImprovement in both cardiac sympathetic (using I-123-MIBG myocardial scintigraphy) and parasympathetic (increase in SDANN, r-MSSD and HF) activity with empagliflozin
Chen et al.40n = 158 300, Taiwanese population-based cohort study with propensity score-matching of diabetes patients on SGLT-2i vs. non-SGLT-2iLower all-cause mortality and new-onset arrhythmia in the SGLT2i (empagliflozin and dapagliflozin) group (HR 0.547 and 0.830, respectively)
  • DECLARE-TIMI 58 post hoc analysis

  • Zelniker et al.41

n = 17 160, T2DM patients from the DECLARE-TIMI 58 study5Lower risk of AF/atrial flutter (HR 0.81) in dapagliflozin group after accounting for other comorbidities and non-modifiable risk factors
  • DAPA–HF post hoc analysis

  • Curtain et al.44

N = 4744, T2DM patients from the DAPA-HF study7Lower risk serious ventricular arrhythmia, cardiac arrest or sudden death when dapagliflozin was added to conventional therapy in patients with HFrEF
Bonora et al.42Pharmacovigilance database from the FDA adverse event reporting system (2014–2019)Lower reporting of AF for SGLT2i vs. other anti-diabetic drugs (PRR 0.55)
Clinical studies
ReferencesCohort detailsMain findings
  • EMPA-HEART CardioLink-6 Holter analysis study

  • Garg et al.30

n = 66, T2DM patients with established CAD from the EMPA-HEART CardioLink-6 study who had completed Holter monitor recordings at baseline and 6 monthsNo significant difference in HRV parameters between empagliflozin and placebo
  • EMBODY trial

  • Shimizu et al.35

n = 105, T2DM patients at 2 weeks post-AMI enrolled into a randomized double-blind study, empagliflozin vs. placeboImprovement in both cardiac sympathetic (using I-123-MIBG myocardial scintigraphy) and parasympathetic (increase in SDANN, r-MSSD and HF) activity with empagliflozin
Chen et al.40n = 158 300, Taiwanese population-based cohort study with propensity score-matching of diabetes patients on SGLT-2i vs. non-SGLT-2iLower all-cause mortality and new-onset arrhythmia in the SGLT2i (empagliflozin and dapagliflozin) group (HR 0.547 and 0.830, respectively)
  • DECLARE-TIMI 58 post hoc analysis

  • Zelniker et al.41

n = 17 160, T2DM patients from the DECLARE-TIMI 58 study5Lower risk of AF/atrial flutter (HR 0.81) in dapagliflozin group after accounting for other comorbidities and non-modifiable risk factors
  • DAPA–HF post hoc analysis

  • Curtain et al.44

N = 4744, T2DM patients from the DAPA-HF study7Lower risk serious ventricular arrhythmia, cardiac arrest or sudden death when dapagliflozin was added to conventional therapy in patients with HFrEF
Bonora et al.42Pharmacovigilance database from the FDA adverse event reporting system (2014–2019)Lower reporting of AF for SGLT2i vs. other anti-diabetic drugs (PRR 0.55)

AF, atrial fibrillation; AMI, acute myocardial infarction; CAD, coronary artery disease; FDA, Food and Drug Administration; HF, high frequency; HFrEF, heart failure with reduced ejection fraction; HR, hazard ratio; HRT, heart rate turbulence; HRV, heart rate variability; I-123-MIBG, I-123-metaiodobenzylguanidine cardiac-scintigraphy; IL-6, interleukin-6; PRR, proportional reporting ratio; r-MSSD, square root of the mean of the sum of the squares of differences between adjacent RR intervals; SDANN, standard deviation of average NN intervals; SGLT-2, sodium glucose cotransporter-2; SGLT-2i, sodium glucose cotransporter-2 inhibitor; T2DM, Type 2 diabetes mellitus.

Table 1

Summary of key findings from preclinical and clinical studies investigating the role of sodium-glucose co-transporter 2 (SGLT2) inhibitors (SGLT2i; gliflozins) in modulating cardiovascular autonomic function as one of the possible pathways of their cardioprotective effects

Preclinical studies
ReferencesDrug and experimental model(s)Main findings
Chiba et al.21Dapagliflozin, C57BL/6 miceSuppressed noradrenaline turnover, oxygen consumption and ucp1 (a thermogenesis-related gene) expression in brown adipose tissue
Yoshikawa et al.22Iprafliflozin, streptozotocin-induced diabetes Sprague-Dawley ratsImproved baroreflex sensitivity and amelioration of arterial blood pressure lability
Matthews et al.24
  • Noradrenaline, human kidney cells

  • Dapagliflozin, high-fat diet-fed mice

  • Increased SGLT2 and IL-6 expression

  • Significant glucosuria, reduced elevation of tyrosine hydroxylase and noradrenaline in the heart and kidney

Wan et al.25Empagliflozin, salt-treated obese and metabolic syndrome ratsDecreased blood pressure, normalized circadian rhythm of blood pressure and sympathetic nervous activity, increased urinary sodium excretion
Herat et al.26
  • Chemical denervation

  • Dapagliflozin, neurogenic hypertensive Schlager mouse

  • Reduced blood pressure, reduced renal SGLT2 protein expression

  • Reduced elevation of tyrosine hydroxylase and norepinephrine, protects against endothelial dysfunction, reduced inflammation

Preclinical studies
ReferencesDrug and experimental model(s)Main findings
Chiba et al.21Dapagliflozin, C57BL/6 miceSuppressed noradrenaline turnover, oxygen consumption and ucp1 (a thermogenesis-related gene) expression in brown adipose tissue
Yoshikawa et al.22Iprafliflozin, streptozotocin-induced diabetes Sprague-Dawley ratsImproved baroreflex sensitivity and amelioration of arterial blood pressure lability
Matthews et al.24
  • Noradrenaline, human kidney cells

  • Dapagliflozin, high-fat diet-fed mice

  • Increased SGLT2 and IL-6 expression

  • Significant glucosuria, reduced elevation of tyrosine hydroxylase and noradrenaline in the heart and kidney

Wan et al.25Empagliflozin, salt-treated obese and metabolic syndrome ratsDecreased blood pressure, normalized circadian rhythm of blood pressure and sympathetic nervous activity, increased urinary sodium excretion
Herat et al.26
  • Chemical denervation

  • Dapagliflozin, neurogenic hypertensive Schlager mouse

  • Reduced blood pressure, reduced renal SGLT2 protein expression

  • Reduced elevation of tyrosine hydroxylase and norepinephrine, protects against endothelial dysfunction, reduced inflammation

Clinical studies
ReferencesCohort detailsMain findings
  • EMPA-HEART CardioLink-6 Holter analysis study

  • Garg et al.30

n = 66, T2DM patients with established CAD from the EMPA-HEART CardioLink-6 study who had completed Holter monitor recordings at baseline and 6 monthsNo significant difference in HRV parameters between empagliflozin and placebo
  • EMBODY trial

  • Shimizu et al.35

n = 105, T2DM patients at 2 weeks post-AMI enrolled into a randomized double-blind study, empagliflozin vs. placeboImprovement in both cardiac sympathetic (using I-123-MIBG myocardial scintigraphy) and parasympathetic (increase in SDANN, r-MSSD and HF) activity with empagliflozin
Chen et al.40n = 158 300, Taiwanese population-based cohort study with propensity score-matching of diabetes patients on SGLT-2i vs. non-SGLT-2iLower all-cause mortality and new-onset arrhythmia in the SGLT2i (empagliflozin and dapagliflozin) group (HR 0.547 and 0.830, respectively)
  • DECLARE-TIMI 58 post hoc analysis

  • Zelniker et al.41

n = 17 160, T2DM patients from the DECLARE-TIMI 58 study5Lower risk of AF/atrial flutter (HR 0.81) in dapagliflozin group after accounting for other comorbidities and non-modifiable risk factors
  • DAPA–HF post hoc analysis

  • Curtain et al.44

N = 4744, T2DM patients from the DAPA-HF study7Lower risk serious ventricular arrhythmia, cardiac arrest or sudden death when dapagliflozin was added to conventional therapy in patients with HFrEF
Bonora et al.42Pharmacovigilance database from the FDA adverse event reporting system (2014–2019)Lower reporting of AF for SGLT2i vs. other anti-diabetic drugs (PRR 0.55)
Clinical studies
ReferencesCohort detailsMain findings
  • EMPA-HEART CardioLink-6 Holter analysis study

  • Garg et al.30

n = 66, T2DM patients with established CAD from the EMPA-HEART CardioLink-6 study who had completed Holter monitor recordings at baseline and 6 monthsNo significant difference in HRV parameters between empagliflozin and placebo
  • EMBODY trial

  • Shimizu et al.35

n = 105, T2DM patients at 2 weeks post-AMI enrolled into a randomized double-blind study, empagliflozin vs. placeboImprovement in both cardiac sympathetic (using I-123-MIBG myocardial scintigraphy) and parasympathetic (increase in SDANN, r-MSSD and HF) activity with empagliflozin
Chen et al.40n = 158 300, Taiwanese population-based cohort study with propensity score-matching of diabetes patients on SGLT-2i vs. non-SGLT-2iLower all-cause mortality and new-onset arrhythmia in the SGLT2i (empagliflozin and dapagliflozin) group (HR 0.547 and 0.830, respectively)
  • DECLARE-TIMI 58 post hoc analysis

  • Zelniker et al.41

n = 17 160, T2DM patients from the DECLARE-TIMI 58 study5Lower risk of AF/atrial flutter (HR 0.81) in dapagliflozin group after accounting for other comorbidities and non-modifiable risk factors
  • DAPA–HF post hoc analysis

  • Curtain et al.44

N = 4744, T2DM patients from the DAPA-HF study7Lower risk serious ventricular arrhythmia, cardiac arrest or sudden death when dapagliflozin was added to conventional therapy in patients with HFrEF
Bonora et al.42Pharmacovigilance database from the FDA adverse event reporting system (2014–2019)Lower reporting of AF for SGLT2i vs. other anti-diabetic drugs (PRR 0.55)

AF, atrial fibrillation; AMI, acute myocardial infarction; CAD, coronary artery disease; FDA, Food and Drug Administration; HF, high frequency; HFrEF, heart failure with reduced ejection fraction; HR, hazard ratio; HRT, heart rate turbulence; HRV, heart rate variability; I-123-MIBG, I-123-metaiodobenzylguanidine cardiac-scintigraphy; IL-6, interleukin-6; PRR, proportional reporting ratio; r-MSSD, square root of the mean of the sum of the squares of differences between adjacent RR intervals; SDANN, standard deviation of average NN intervals; SGLT-2, sodium glucose cotransporter-2; SGLT-2i, sodium glucose cotransporter-2 inhibitor; T2DM, Type 2 diabetes mellitus.

Current evidence from preclinical studies

Multiple preclinical studies have sought to evaluate the cardiovascular autonomic effects of SGLT2 inhibition. In this context, Chiba et al.21 showed that acute oral administration of dapagliflozin significantly suppressed noradrenaline turnover in the brown adipose tissue of mice within 18 h, which reflects the SNS activity in adipose tissue. Interestingly, Yoshikawa et al.22 found that ipragliflozin mitigated the arterial blood pressure lability (arterial blood pressure variability which frequently impacts negatively on the T2DM pathophysiology23) in streptozotocin-induced diabetic rats. Of note, Matthews et al.24 were the first to demonstrate that oral dapagliflozin reduced SNS activity markers (e.g. tyrosine hydroxylase and noradrenaline) in the kidneys and heart of mice fed a high-fat diet (HFD). Moreover, Wan et al.25 showed that empagliflozin use in diabetic Otsuka Long Evans Tokushima (OLETF) rats improved the circadian rhythm of SNS activity (as measured by blood pressure recordings) via a sympatho-inhibitory effect during the sleeping period.

More recently, a series of elegant experiments further demonstrated the positive effects of SGLT2 inhibition on SNS activity.26 In these studies, chemical denervation of the SNS with oxidopamine (6-OHDA) in a neurogenic hypertensive non-diabetic mouse model [Schlager (BPH/2J) mouse] was shown to reduce the expression of renal SGLT2 with improvements in both metabolic and haemodynamic parameters. The authors then introduced dapagliflozin (by oral gavage every 2 days for 2 weeks) to HFD-fed Schlager mice and found that in addition to improvements in blood glucose levels and blood pressure (similar to that shown in the relevant clinical trials), SGLT2 inhibition was associated with a reduction in cardiac and renal tyrosine hydroxylase staining (a reliable marker of SNS activity) and norepinephrine content.26 In addition, SGLT2 inhibition was associated with improvement of endothelial function and reduced inflammation, documented by attenuated pro-inflammatory cytokines and increased cardiac and renal anti-inflammatory interleukin-10. In other words, the lower amounts of sympathetic activation with SGLT2 inhibitors may just be secondary to improvements in haemodynamics and metabolic stress. Collectively, the findings suggest a bidirectional relationship between SGLT2 inhibition and SNS activity.

Current evidence from clinical studies

Given the promising findings of the relevant preclinical studies, further studies aimed to evaluate the effects of SGLT2 inhibition on SNS activity in the clinical setting. Intriguingly, a relevant case report exists in the medical literature presenting the case of an 83-year-old patient with diabetic and chronic heart failure whose cardiac sympathetic nerve hyperactivity—a potentially useful parameter for evaluating the prognosis of heart failure,27 which was assessed by I-123-metaiodobenzylguanidine cardiac-scintigraphy (I-123-MIBG)—was reduced following 12 months of ipragliflozin administration, with no heart failure related re-hospitalization 2 years after the initiation of this treatment.28 More recently, two clinical studies that sought to evaluate the effects of an SGLT2 inhibitor on the cardiovascular autonomic function were published and are discussed in the following sections.

The EMPA-HEART CardioLink-6 Holter analysis study

The EMPA-HEART CardioLink-6 study was a randomized trial that compared empagliflozin vs. placebo on cardiac remodelling in patients with T2DM and established coronary artery disease (CAD), which documented that empagliflozin was associated with significantly decreased left ventricular mass at 6 months of treatment.29 From this study, Garg et al.30 extracted the collected Holter monitoring data (66 participants completed 24-h ambulatory Holter monitoring at baseline and 6 months) to conduct an exploratory analysis on the effects of empagliflozin on heart rate variability (HRV). These results were reported as the EMPA-HEART CardioLink-6 Holter analysis study which was the first clinical study to evaluate the effects of SGLT2 inhibition on cardiovascular autonomic function using HRV parameters from non-invasive Holter monitoring.30 Of note, HRV is a measure of variations in time intervals between two consecutive sinus heart beats and constitutes a surrogate marker characterizing the sympathetic and parasympathetic aspects of the autonomic nervous system. For the clinical practice, a growing body of evidence has shown that a lower HRV is associated with poorer cardiovascular outcomes, such as myocardial infarction, heart failure, sudden cardiac death, and cardiovascular mortality.31–33

Interestingly, the EMPA-HEART CardioLink-6 Holter analysis study found no significant difference in the HRV between the empagliflozin and placebo groups. Although the findings were contrary to the hypothesis that SGLT2 inhibition may have a positive impact on the SNS, as suggested by the aforementioned preclinical studies, these neutral findings may be attributed to a number of reasons. Indeed, since the study cohort consisted of subjects with stable CAD, it is plausible that more pronounced changes in the autonomic tone might be detected in patients with more advanced cardiac comorbidities, such as myocardial infarction and significant left ventricular dysfunction. Moreover, as this study presented a post hoc exploratory analysis, the analysed sample size was not powered to assess for pre-specified differences in the HRV parameters. Furthermore, although the high frequency component of HRV is largely influenced by the vagal tone, no HRV parameter is considered to be truly reflective of SNS activity, but instead the HRV parameters are felt to represent the interplay between the parasympathetic and sympathetic arms of the cardiovascular autonomic system.34 Therefore, HRV parameters may not constitute the best method to assess primary changes in the SNS due to SGLT2 inhibition. Additional modalities, such as heart rate turbulence (HRT), heart rate recovery post-exercise, or imaging of the autonomic nervous system with radiotracers (as utilized in the aforementioned case report), could provide a more holistic assessment of the cardiovascular autonomic function, but understandably this was beyond the scope of the EMPA-HEART CardioLink-6 Holter analysis study. Finally, data on the doses of beta-blockers and anti-hypertensive agents were not obtained in the context of this study, and so it was not possible to determine if this could have affected HRV parameters in the studied participants.

The EMBODY trial

The EMBODY trial35 was a more recent prospective, multicentre, randomized, double-blind, placebo-controlled trial which evaluated patients with T2DM and acute myocardial infarction (AMI) in Japan. In this study, 105 patients were randomized at 2 weeks post-AMI to either empagliflozin or placebo, and were subsequently followed up to 24 weeks. The EMBODY trial endpoints included changes in HRV, HRT, T-wave alternans (TWA), and late potentials (LP) as measured by 24-h ambulatory Holter monitoring, whilst cardiac sympathetic activity via I-123-MIBG myocardial scintigraphy was also assessed.

Based on the results of this study, there were no significant differences in the intergroup comparison; however, there were significant improvements in the parameters reflecting both sympathetic and parasympathetic nerve activity within the empagliflozin group. In particular, the standard deviation of all 5-min mean normal RR intervals (SDANN), square root of the mean of the sum of the squares of differences between adjacent RR intervals (r-MSSD) and high frequency were increased, reflecting improved parasympathetic nerve activity. Moreover, the low-to-high frequency ratio (a marker of sympathovagal balance) was reduced which reflects improved cardiac SNS activity. Significantly improved HRT was also noted in the empagliflozin group. Cardiac SNS activity, as reflected by I-123-MIBG myocardial scintigraphy, showed improvement in both study groups, reflecting the natural recovery course of AMI. Overall, the EMBODY trial was the first randomized clinical study to demonstrate improvement in both cardiac sympathetic and parasympathetic activity by SGLT2 inhibition. The difference in the underlying cardiac comorbidity of the recruited patients with T2DM—patients with stable CAD in the EMPA-HEART Link-6 Holter analysis vs. patients with acute coronary syndrome in the EMBODY trial—may be one of the reasons that could explain the differing findings from these trials.

Current evidence for reduction in sympathetic activity and arrhythmia risk with SGLT2 inhibitors

One of the potential translational effects of SNS activity reduction caused by SGLT2 inhibition is a subsequent reduction in arrhythmias. Diabetes is associated with the development of arrhythmias such as atrial fibrillation (AF), which constitute a key risk factor for sudden cardiac death and arrhythmic deaths.36,37 Furthermore, the manifestation of arrhythmias in patients with diabetes is also associated with a worse prognosis.38 The suggested mechanisms for the increased risk of arrhythmias include myocardial fibrosis, oxidative stress and inflammation leading to cardiac electromechanical and autonomic remodelling.39 At present, there are still no SGLT2 inhibitor clinical trials or basic science studies that have investigated the drug as a pure anti-arrhythmic agent but the following observational studies attempt to address this gap in our knowledge.

Real-world data: a nationwide population-based longitudinal cohort study

As none of the CVOTs for SGLT2 inhibitors included the arrhythmic burden as a primary outcome, Chen et al.40 attempted to assess the risk of new-onset arrhythmias and all-cause mortality with SGLT2 inhibitors by utilizing real-world data from the Taiwanese National Health Insurance Research Database. In the context of this study, empagliflozin and dapagliflozin were the SGLT2 inhibitors used at that time, whilst the range of studied arrhythmias included AF and other supraventricular and ventricular arrhythmias based on the International Classification of Diseases, 9th and 10th revision, Clinical Modification (ICD-9-CM and ICD-10-CM). The study only looked at two outcome measures: all-cause mortality and new onset arrhythmia.

Interestingly, the study revealed that SGLT2 inhibitors were associated with a 17% reduction in new-onset arrhythmias [adjusted hazard ratio of 0.830 (95% confidence interval, 0.751–0.916)] and 46% reduction in all-cause mortality [adjusted hazard ratio of 0.547 (95% confidence interval, 0.482–0.621)]. In terms of the breakdown of the new-onset arrhythmias, the occurrence of AF, supraventricular arrhythmias and ventricular arrhythmias were lower in the SGLT2 inhibitor group but did not reach statistical significance [adjusted hazard ratios—AF: 0.841 (0.662–1.068), supraventricular arrhythmias 0.815 (0.570–1.167), and ventricular arrhythmias 0.797 (0.525–1.208)].

Other studies

A protective effect of SGLT2 inhibitors on AF was also reported by other recent studies. Indeed, the post hoc analysis of the DECLARE-TIMI 58 trial showed that dapagliflozin was associated with a 19% reduction in AF and atrial flutter.41 This was independent of the presence of AF/atrial flutter, atherosclerotic cardiovascular disease or history of heart failure at baseline.

In addition, Bonora et al.42 interrogated a large pharmacovigilance database (the FDA adverse event reporting system) to show that the reporting of AF was significantly lower with SGLT2 inhibitors, suggesting a protective role against the onset of AF. Taken together, the findings of these clinical studies suggest the exciting potential of SGLT2 inhibitors having clinically relevant anti-arrhythmic properties.

More recently, a meta-analysis of 34 randomized controlled trials with 63 166 patients with either T2DM or heart failure found that SGLT2 inhibitors (canagliflozin, dapagliflozin, empagliflozin, or ertogliflozin) were associated with a 19% reduced risk of incident atrial arrhythmias and 28% reduction in sudden cardiac death.43

SGLT2 inhibition may also have a positive impact in reducing the risk of developing ventricular arrhythmias. A recent post hoc analysis of the DAPA-HF trial found that dapagliflozin was associated with an improvement in composite outcome (5.9% compared with 7.4%, hazard ratio 0.79, 95% confidence interval, 0.63–0.99, P = 0.037) which consisted of the first occurrence of any serious ventricular arrhythmias, resuscitated cardiac arrest or sudden death.44

However, the association found in the post hoc analysis of DAPA-HF was not observed in a very recent meta-analysis of 19 randomised controlled trials (RCT) with 55, 590 participants (45). Sfairopoulos et al showed that SGLT2 inhibitors were not significantly associated with a lower risk of sudden cardiac death [9 RCTs, risk ratio (RR) 0.74, 95% confidence interval (CI) 0.50-1.08; P = 0.12] and ventricular arrhythmias (17 RCTs, RR 0.84, 95% CI 0.66-1.06; P = 0.14).45 However, it is worth noting that the number of sudden cardiac death and ventricular arrhythmia events were relatively few leading to wide confidence intervals and the point estimates suggested potential benefits, hence the need for further research in this area.

Ongoing clinical trials

The EMPYREAN study is an ongoing prospective, randomized, open-label, multicentre trial of 134 patients with T2DM in Japan.46 Participants of this study are randomly allocated to receive either empagliflozin or sitagliptin (a dipeptidylpeptidase-4 inhibitor), whilst ambulatory Holter monitoring is performed at baseline and at 12 and 24 weeks of treatment. Similarly to the EMPA-HEART CardioLink-6 Holter analysis study, HRV will be analysed in the EMBODY trial with the primary endpoint being the change in the low-frequency (0.04–0.15 Hz) to high-frequency (0.15–0.4 Hz) ratio from baseline to 24 weeks.

Limitations

The autonomic nervous system is highly complex in terms of its anatomy and physiology and attempts to evaluate the effects of SGLT2 inhibitors on specific components of this sophisticated system has been challenging. Therefore, it is unsurprising that much of the conclusions drawn from the current clinical studies looking into the effects of SGLT2 inhibition on the cardiovascular autonomic nervous system modulation relied on indirect measures of autonomic function and the results have been at times, contradictory. It is also unclear if the improvement in autonomic nervous system parameters is the cause or simply an association to the improvement in cardiovascular function. In other words, the improvement in haemodynamic parameters and metabolism could either be an effect of the reduction in sympathetic activity or vice versa. In addition, there have been other mechanisms demonstrated by other research groups, which may confound the effects of autonomic nervous system modulation and reduction in arrhythmia risk observed in the clinical studies. For example, dapagliflozin (in animal studies) has been shown to reduce myofibroblast infiltration and myocardial fibrosis (which can lead to a reduction in arrhythmic substrate) via the activation of known mediator of cardioprotection, the janus kinase—signal transducer and activator of transcription 3 (JAK-STAT3) pathway.47 It is also worth noting that unlike the SGLT2 inhibitor CVOTs, the clinical studies that attempted to evaluate the reduction in arrhythmia risk by SGLT2 inhibitors as described in this article were not prospective or randomized and not designed to look at the reduction in arrhythmia onset, burden or arrhythmic death. Unlike recent SGLT2 inhibitor trials like the DAPA-HF,7 EMPEROR-Reduced,48 and EMPEROR-Preserved49 which evaluated a specific heart failure patient population, similarly designed clinical trials are needed in a cardiac arrhythmia patient population.

Future pre-clinical and clinical studies should therefore aim to address the aforementioned limitations when designing their studies.

Future perspectives and conclusions

Although the exact mechanism(s) behind the cardioprotection noted with treatment with the different SGLT2 inhibitors have yet to be established, both the preclinical and clinical studies discussed in this review highlight the potential role of SGLT2 inhibitors in modulating cardiovascular autonomic function. Additional studies from bench to bedside are clearly required to understand how SGLT2 inhibitors exert their off-target effects on the autonomic nervous system, and how this modulation translates into improved cardiovascular outcomes. In terms of the potential of SGLT2 inhibitors to be repurposed as anti-arrhythmic agents, there is still a great deficiency in basic science and clinical studies to support this. In conclusion, more pre-clinical and clinical studies are required to address this gap in our knowledge on the role of SGLT2 inhibitors as modulators of cardiac autonomic function.

Acknowledgements

All authors have made an important scientific contribution to the study and have assisted with the drafting or revising of the manuscript, in accordance with the definition of an author as stated by the International Committee of Medical Journal Editors (ICMJE) at http://www.icmje.org/recommendations. We confirm that neither the manuscript nor any part of it has been published or is under consideration for publication elsewhere and that there are no conflicts of interests or funding to report. The guarantor for this article is Professor Faizel Osman.

Conflict of interest: none declared.

References

1

Zinman
B
,
Wanner
C
,
Lachin
JM
,
Fitchett
D
,
Bluhmki
E
,
Hantel
S
et al. ;
EMPA-REG OUTCOME Investigators
.
Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes
.
N Engl J Med
2015
;
373
:
2117
28
.

2

FDA
.
Guidance for Industry Diabetes Mellitus—Evaluating Cardiovascular Risk in New Antidiabetic Therapies to Treat Type 2 Diabetes
.
Silver Spring, MD
:
FDA
.
2008
.

3

EMA
.
Guideline on Clinical Investigation of Medicinal Products in the Treatment or Prevention of Diabetes Mellitus
.
London, UK
:
EMA
.
2012
.

4

Neal
B
,
Perkovic
V
,
Mahaffey
KW
,
de Zeeuw
D
,
Fulcher
G
,
Erondu
N
et al.
Canagliflozin and cardiovascular and renal events in type 2 diabetes
.
N Engl J Med
2017
;
377
:
644
57
.

5

Wiviott
S
,
Raz
I
,
Bonaca
M
,
Mosenzon
O
,
Kato
E
,
Cahn
A
et al.
Dapagliflozin and cardiovascular outcomes in type 2 diabetes
.
N Engl J Med
2019
;
380
:
347
57
.

6

Perkovic
V
,
Jardine
MJ
,
Neal
B
,
Bompoint
S
,
Heerspink
HJL
,
Charytan
DM
et al.
Canagliflozin and renal outcomes in type 2 diabetes and nephropathy
.
N Engl J Med
2019
;
380
:
2295
306
.

7

McMurray
JJV
,
Solomon
SD
,
Inzucchi
SE
,
Køber
L
,
Kosiborod
MN
,
Martinez
FA
et al.
Dapagliflozin in patients with heart failure and reduced ejection fraction
.
N Engl J Med
2019
;
381
:
1995
2008
.

8

Wright
EM
,
Loo
DDF
,
Hirayama
BA
,
Human
C.
Biology of human sodium glucose transporters
.
Physiol Rev
2011
;
91
:
733
94
.

9

Ghezzi
C
,
Loo
DDF
,
Wright
EM.
Physiology of renal glucose handling via SGLT1, SGLT2 and GLUT2
.
Diabetologia
2018
;
61
:
2087
97
.

10

Lee
MMY
,
Petrie
MC
,
McMurray
JJV
,
Sattar
N.
How do SGLT2 (sodium-glucose cotransporter 2) inhibitors and GLP-1 (glucagon-like peptide-1) receptor agonists reduce cardiovascular outcomes? Completed and ongoing mechanistic trials
.
Arterioscler Thromb Vasc Biol
2020
;
40
:
506
22
.

11

Lim
VG
,
Bell
RM
,
Arjun
S
,
Kolatsi-Joannou
M
,
Long
DA
,
Yellon
DM.
SGLT2 inhibitor, canagliflozin, attenuates myocardial infarction in the diabetic and nondiabetic heart
.
JACC Basic Transl Sci
2019
;
4
:
15
26
.

12

De Pascalis
A
,
Cianciolo
G
,
Capelli
I
,
Brunori
G
,
La Manna
G.
SGLT2 inhibitors, sodium and off-target effects: an overview
.
J Nephrol
2021
;
34
:
673
80
.

13

Chung
YJ
,
Park
KC
,
Tokar
S
,
Eykyn
TR
,
Fuller
W
,
Pavlovic
D
et al.
Off-target effects of sodium-glucose co-transporter 2 blockers: empagliflozin does not inhibit Na+/H+ exchanger-1 or lower [Na+]i in the heart
.
Cardiovasc Res
2021
;
117
:
2794
2806
.

14

Rahman
A
,
Fujisawa
Y
,
Nakano
D
,
Hitomi
H
,
Nishiyama
A.
Effect of a selective SGLT2 inhibitor, luseogliflozin, on circadian rhythm of sympathetic nervous function and locomotor activities in metabolic syndrome rats
.
Clin Exp Pharmacol Physiol
2017
;
44
:
522
5
.

15

Vinik
AI
,
Ziegler
D.
Diabetic cardiovascular autonomic neuropathy
.
Circulation
2007
;
115
:
387
97
.

16

Kannel
WB
,
Kannel
C
,
Paffenbarger
RS
,
Cupples
LA.
Heart rate and cardiovascular mortality: the Framingham study
.
Am Heart J
1987
;
113
:
1489
94
.

17

Packer
M
,
Coats
AJS
,
Fowler
MB
,
Katus
HA
,
Krum
H
,
Mohacsi
P
et al.
Effect of carvedilol on survival in severe chronic heart failure
.
N Engl J Med
2001
;
344
:
1651
8
.

18

Dargie
HJ
,
Lechat
P.
The Cardiac Insufficiency Bisoprolol Study II (CIBIS-II): a randomised trial
.
Lancet
1999
;
353
:
9
13
.

19

Hjalmarson
A
,
Goldstein
S
,
Fagerberg
B
,
Wedel
H
,
Waagstein
F
,
Kjekshus
J
et al.
Effect of metoprolol CR/XL in chronic heart failure: metoprolol CR/XL Randomised Intervention Trial in Congestive Heart Failure (MERIT-HF)
.
Lancet
1999
;
353
:
2001
7
.

20

Packer
M
,
Bristow
MR
,
Cohn
JN
,
Colucci
WS
,
Fowler
MB
,
Gilbert
EM
et al.
The effect of carvedilol on morbidity and mortality in patients with chronic heart failure
.
N Engl J Med
1996
;
334
:
1349
55
.

21

Chiba
Y
,
Yamada
T
,
Tsukita
S
,
Takahashi
K
,
Munakata
Y
,
Shirai
Y
et al.
Dapagliflozin, a sodium-glucose co-transporter 2 inhibitor, acutely reduces energy expenditure in BAT via neural signals in mice
.
PLoS One
2016
;
11
:
e0150756
.

22

Yoshikawa
T
,
Kishi
T
,
Shinohara
K
,
Takesue
K
,
Shibata
R
,
Sonoda
N
et al.
Arterial pressure lability is improved by sodium-glucose cotransporter 2 inhibitor in streptozotocin-induced diabetic rats
.
Hypertens Res
2017
;
40
:
646
51
.

23

Tanaka
A
,
Node
K.
Amelioration of arterial pressure lability: an unmissable target for diabetes management
.
Hypertens Res
2017
;
40
:
629
31
.

24

Matthews
VB
,
Elliot
RH
,
Rudnicka
C
,
Hricova
J
,
Herat
L
,
Schlaich
MP.
Role of the sympathetic nervous system in regulation of the sodium glucose cotransporter 2
.
J Hypertens
2017
;
35
:
2059
68
.

25

Wan
N
,
Rahman
A
,
Hitomi
H
,
Nishiyama
A.
The effects of sodium-glucose cotransporter 2 inhibitors on sympathetic nervous activity
.
Front Endocrinol (Lausanne)
2018
;
9
:
421
.

26

Herat
LY
,
Magno
AL
,
Rudnicka
C
,
Hricova
J
,
Carnagarin
R
,
Ward
NC
et al.
SGLT2 inhibitor–induced sympathoinhibition: a novel mechanism for cardiorenal protection
.
JACC Basic Transl Sci
2020
;
5
:
169
79
.

27

Narula
J
,
Gerson
M
,
Thomas
GS
,
Cerqueira
MD
,
Jacobson
AF.
123I-MIBG imaging for prediction of mortality and potentially fatal events in heart failure: the ADMIRE-HFX study
.
J Nucl Med
2015
;
56
:
1011
31
.

28

Kiuchi
S
,
Hisatake
S
,
Kabuki
T
,
Fujii
T
,
Oka
T
,
Dobashi
S
et al.
Long-term use of ipragliflozin improved cardiac sympathetic nerve activity in a patient with heart failure: a case report
.
Drug Discov Ther
2018
;
12
:
51
4
.

29

Verma
S
,
Mazer
CD
,
Yan
AT
,
Mason
T
,
Garg
V
,
Teoh
H
et al.
Effect of empagliflozin on left ventricular mass in patients with type 2 diabetes mellitus and coronary artery disease: the EMPA-HEART CardioLink-6 randomized clinical trial
.
Circulation
2019
;
140
:
1693
702
.

30

Garg
V
,
Verma
S
,
Connelly
KA
,
Yan
AT
,
Sikand
A
,
Garg
A
et al.
Does empagliflozin modulate the autonomic nervous system among individuals with type 2 diabetes and coronary artery disease? The EMPA-HEART CardioLink-6 Holter analysis
.
Metabol Open
2020
;
7
:
100039
.

31

Maheshwari
A
,
Norby
FL
,
Soliman
EZ
,
Adabag
S
,
Whitse
EA
,
Alonso
A
et al.
Low heart rate variability in a 2-minute electrocardiogram recording is associated with an increased risk of sudden cardiac death in the general population: the atherosclerosis risk in communities study
.
PLoS One
2016
;
11
:
e0161648
.

32

Dekker
JM
,
Crow
RS
,
Folsom
AR
,
Hannan
PJ
,
Liao
D
,
Swenne
CA
et al.
Low heart rate variability in a 2-minute rhythm strip predicts risk of coronary heart disease and mortality from several causes: the ARIC study
.
Circulation
2000
;
102
:
1239
44
.

33

Vaseghi
M
,
Shivkumar
K.
The role of the autonomic nervous system in sudden cardiac death
.
Prog Cardiovasc Dis
2008
;
50
:
404
19
.

34

Heart rate variability: standards of measurement, physiological interpretation and clinical use
.
Task Force of the European Society of Cardiology and the North American Society of Pacing and Electrophysiology
.
Circulation
1996
;
93
:
1043
65
.

35

Shimizu
W
,
Kubota
Y
,
Hoshika
Y
,
Mozawa
K
,
Tara
S
,
Tokita
Y
et al. ;
EMBODY Trial Investigators
.
Effects of empagliflozin versus placebo on cardiac sympathetic activity in acute myocardial infarction patients with type 2 diabetes mellitus: the EMBODY trial
.
Cardiovasc Diabetol
2020
;
19
:
148
.

36

Bell
DSH
,
Goncalves
E.
Atrial fibrillation and type 2 diabetes: prevalence, etiology, pathophysiology and effect of anti-diabetic therapies
.
Diabetes Obes Metab
2019
;
21
:
210
7
.

37

Weidner
K
,
Behnes
M
,
Schupp
T
,
Rusnak
J
,
Reiser
L
,
Bollow
A
et al.
Type 2 diabetes is independently associated with all-cause mortality secondary to ventricular tachyarrhythmias
.
Cardiovasc Diabetol
2018
;
17
:
125
.

38

Chen-Scarabelli
C
,
Scarabelli
TM.
Suboptimal glycemic control, independently of QT interval duration, is associated with increased risk of ventricular arrhythmias in a high-risk population
.
Pacing Clin Electrophysiol
2006
;
29
:
9
14
.

39

Wang
A
,
Green
JB
,
Halperin
JL
,
Piccini
JP.
Atrial fibrillation and diabetes mellitus: JACC review topic of the week
.
J Am Coll Cardiol
2019
;
74
:
1107
15
.

40

Chen
HY
,
Huang
JY
,
Siao
WZ
,
Jong
GP.
The association between SGLT2 inhibitors and new-onset arrhythmias: a nationwide population-based longitudinal cohort study
.
Cardiovasc Diabetol
2020
;
19
:
73
.

41

Zelniker
TA
,
Bonaca
MP
,
Furtado
RHM
,
Mosenzon
O
,
Kuder
JF
,
Murphy
SA
et al.
Effect of dapagliflozin on atrial fibrillation in patients with type 2 diabetes mellitus: insights from the DECLARE-TIMI 58 Trial
.
Circulation
2020
;
141
:
1227
34
.

42

Bonora
BM
,
Raschi
E
,
Avogaro
A
,
Fadini
GP.
SGLT-2 inhibitors and atrial fibrillation in the Food and Drug Administration adverse event reporting system
.
Cardiovasc Diabetol
2021
;
20
:
39
.

43

Fernandes
GC
,
Fernandes
A
,
Cardoso
R
,
Penalver
J
,
Knijnik
L
,
Mitrani
RD
et al.
Association of SGLT2 inhibitors with arrhythmias and sudden cardiac death in patients with type 2 diabetes or heart failure: a meta-analysis of 34 randomized controlled trials
.
Heart Rhythm
2021
;
21
:
S1547
5271
.

44

Curtain
JP
,
Docherty
KF
,
Jhund
PS
,
Petrie
MC
,
Inzucchi
SE
,
Kober
L
et al.
Effect of dapagliflozin on ventricular arrhythmias, resuscitated cardiac arrest, or sudden death in DAPA-HF
.
Eur Heart J
2021
;
42
:
3727
38
.

45

Sfairopoulos
D
,
Zhang
N
,
Wang
Y
,
Chen
Z
,
Letsas
KP
,
Tse
G
et al.
Association between sodium-glucose cotransporter-2 inhibitors and risk of sudden cardiac death or ventricular arrhythmias: a meta-analysis of randomized controlled trials
.
Europace
2022
;
24
:
20
30
.

46

Motoki
H
,
Masuda
I
,
Yasuno
S
,
Oba
K
,
Shoin
W
,
Usami
S
et al.
Rationale and design of the EMPYREAN study
.
ESC Heart Fail
2020
;
7
:
3134
41
.

47

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
.

48

Packer
M
,
Anker
SD
,
Butler
J
,
Filippatos
G
,
Pocock
SJ
,
Carson
P
et al.
Cardiovascular and renal outcomes with empagliflozin in heart failure
.
N Engl J Med
2020
;
383
:
1413
24
.

49

Anker
SD
,
Butler
J
,
Filippatos
G
,
Ferreira
JP
,
Bocchi
E
,
Bohm
M
et al.
Empagliflozin in heart failure with a preserved ejection fraction
.
N Engl J Med
2021
;
385
:
1451
61
.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)