Skip to main content
  • Original investigation
  • Open access
  • Published:

A prospective randomized study comparing effects of empagliflozin to sitagliptin on cardiac fat accumulation, cardiac function, and cardiac metabolism in patients with early-stage type 2 diabetes: the ASSET study

Abstract

Background

While the cardioprotective benefits of sodium-glucose cotransporter-2 (SGLT2) inhibitors have been established in patients with cardiovascular disease (CVD), their advantages over other anti-diabetic drugs at earlier stages remain unclear. We compared the cardioprotective effects of empagliflozin, an SGLT2 inhibitor, with those of sitagliptin, a dipeptidyl peptidase-4 (DPP-4) inhibitor, focusing on cardiac fat accumulation, cardiac function, and cardiac metabolism in patients with early-stage type 2 diabetes mellitus (T2DM) without CVD complications.

Methods

This was a prospective, randomized, open-label, blinded-endpoint, parallel-group trial that enrolled 44 Japanese patients with T2DM. The patients were randomized for 12-week administration of empagliflozin or sitagliptin. Pericardial fat accumulation and myocardial triglyceride content were evaluated by magnetic resonance imaging and proton magnetic resonance spectroscopy, respectively. Echocardiography, 123I-β-methyl-iodophenyl pentadecanoic acid myocardial scintigraphy, and laboratory tests were performed at baseline and after the 12-week treatment period.

Results

The patients were middle-aged (50.3 ± 10.7 years, mean ± standard deviation) and overweight (body mass index 29.3 ± 4.9 kg/m2). They had a short diabetes duration (3.5 ± 3.2 years), HbA1c levels of 7.1 ± 0.8%, and preserved cardiac function (ejection fraction 73.8 ± 5.0%) with no vascular complications, except for one baseline case each of diabetic nephropathy and peripheral arterial disease. After the 12-week treatment, no differences from baseline were observed between the two groups regarding changes in pericardial, epicardial, and paracardial fat content; myocardial triglyceride content; cardiac function and mass; and cardiac fatty acid metabolism. However, considering cardiometabolic biomarkers, high-density lipoprotein cholesterol and ketone bodies, including β-hydroxybutyric acid, were significantly increased, whereas uric acid, plasma glucose, plasma insulin, and homeostasis model assessment of insulin resistance were significantly lower in the empagliflozin group than in the sitagliptin group (p < 0.05).

Conclusions

Although the effects on cardiac fat and function were not statistically different between the two groups, empagliflozin exhibited superior effects on cardiometabolic biomarkers, such as uric acid, high-density lipoprotein cholesterol, ketone bodies, and insulin sensitivity. Therefore, when considering the primary preventive strategies for CVD, early supplementation with SGLT2 inhibitors may be more beneficial than DPP-4 inhibitors, even in patients with early-stage T2DM without current CVD complications.

Clinical Trial Registration: UMIN000026340; registered on February 28, 2017. https://upload.umin.ac.jp/cgi-open-bin/icdr_e/ctr_view.cgi?recptno=R000030257

Background

Type 2 diabetes mellitus (T2DM) is a major risk factor for cardiovascular disease (CVD) [1, 2]. Various glucose-lowering agents, including sodium-glucose cotransporter-2 (SGLT2) inhibitors and dipeptidyl peptidase-4 (DPP-4) inhibitors, are commonly administered for the treatment of T2DM. The former lowers blood glucose levels by suppressing reuptake of sodium and glucose from primitive urine and has been shown to reduce CVD events in patients with/without T2DM, as well as in those at high risk of CVD or those with reduced cardiac function [3,4,5,6]. The latter lowers blood glucose levels by increasing incretin hormone levels but does not reduce CVD events [7,8,9,10]. Hence, a simple reduction in blood glucose levels is not sufficient for cardiovascular protection, and other mechanisms are involved.

It has been suggested that cardiac fat accumulation causes excessive release of proinflammatory cytokines and free fatty acids, resulting in myocardial intracellular lipotoxicity, myocardial fibrosis, and cardiac dysfunction [11,12,13,14,15,16,17]. Although both SGLT2 inhibitors and DPP-4 inhibitors reduce epicardial fat [18,19,20,21,22], no clinical studies have been performed to compare the effectiveness of these inhibitors on pericardial and epicardial fat, as well as myocardial triglyceride content. However, Lee et al. recently reported that SGLT2 inhibitors (empagliflozin and dapagliflozin) were superior to DPP-4 inhibitors for improving cardiac function after 24 months of treatment in patients with T2DM, as well as in those with reduced cardiac function and previous CVD events [23]. Moreover, regarding cardiac metabolism, it is well-known that the impaired heart switches energy sources from fatty acids to glucose [24]. Hence, disordered cardiac fatty acid metabolism could be associated with impaired myocardial lipolysis, increased myocardial triglyceride content, abnormal cardiac wall motion, and future cardiac events [25,26,27,28]. Recently, SGLT2 inhibitors were reported to induce a global change in energy substrates from glucose to lipids throughout the body [29]. However, the effects of SGLT2 inhibitors and DPP-4 inhibitors on cardiac fatty acid metabolism remain unknown.

Accumulating evidence suggests the superiority of SGLT2 inhibitors over DPP-4 inhibitors in their cardioprotective role in patients with past CVD events or high CVD risks [3,4,5,6,7,8,9,10, 23, 30,31,32,33]. However, it remains unclear whether SGLT2 inhibitors are superior to DPP-4 inhibitors in reducing cardiovascular or cardiometabolic risk factors in patients with early-stage T2DM, without CVD, and with preserved cardiac function.

In this study, we hypothesized that SGLT2 inhibitors elicit more cardioprotective effects than DPP-4 inhibitors, specifically in patients with early-stage T2DM and without CVD (including not having heart failure). Thus, considering the primary prevention of CVD, this study was performed to compare the effects of empagliflozin and sitagliptin on CVD risk factors, including pericardial and epicardial fat accumulation, myocardial triglyceride content, cardiac function, cardiac fatty acid metabolism, and metabolic biomarkers in patients with early-stage T2DM, without a history of CVD events.

Methods

Study design

This was a prospective, randomized open-label, blinded-endpoint study. The design and rationale have been reported previously [34]. This study was registered in the University Hospital Medical Information Network Clinical Trial Registry (UMIN000026340), a nonprofit organization in Japan that meets the requirements of the International Committee of Medical Journal Editors, and it was approved by the certified clinical research review board of Toho University (CRB3180016), as well as the Ethics Committee of Toho University Omori Medical Center (M16193). This study was conducted according to the Declaration of Helsinki and current legal regulations in Japan. To avoid bias in the collected data, the processes of enrollment, randomization, data management, and analysis were conducted by a third-party (Soiken, Inc., Tokyo, Japan).

Study population

The target number of patients required for registration was 44. Recruitment for the study began in April 2017 and ended in March 2019 at the Toho University Omori Medical Center. The inclusion criteria were as follows: (1) T2DM patients with proper diet and exercise therapy alone or prescribed α-glucosidase inhibitors, sulfonylureas, glinides, or combinations of these agents; (2) patients with glycated hemoglobin (HbA1c) levels of 6.0–10.0%; (3) patients aged 20–74 years; (4) patients with a body mass index of ≥ 22 kg/m2; and (5) patients who provided written informed consent. The exclusion criteria were: (1) patients with type 1 diabetes mellitus or secondary diabetes mellitus; (2) patients with renal dysfunction (estimated glomerular filtration rate < 45 mL/min/1.73 m2); (3) patients with a medical history of cerebral infarction or stroke within 12 weeks prior to giving consent for enrollment; (4) patients with a past medical history of myocardial infarction, angina pectoris, or present medical history of atrial fibrillation; (5) patients with left ventricular ejection fraction (LVEF) < 30%; (6) patients with infection; (7) patients with untreated cancer; (8) patients with collagen diseases, with the exception of well-controlled disease progression with prednisolone ≤ 5 mg/day; (9) patients with hepatic cirrhosis; (10) patients with liver failure that was virus-, autoimmune- or drug-induced; (11) patients with alcoholism; (12) pregnant or breastfeeding patients, or those planning to become pregnant during the course of the study; (13) patients allergic to empagliflozin or sitagliptin; and (14) patients with anemia (hemoglobin < 12 g/dL).

Randomization and study intervention

After consent and enrollment, baseline checkups were performed for each subject. Within 2 weeks of checkup, eligible subjects were randomly and equally assigned to the empagliflozin add-on group (empagliflozin 10 mg/day) or sitagliptin add-on group (sitagliptin 50 mg/day as the initial dose). Randomization was performed by a computer-based dynamic allocation method using the presence of sulfonylurea administration and intrahepatic lipid content assessed by proton magnetic resonance spectroscopy (1H-MRS) at baseline as the assignment factors. After treatment initiation, vital signs were assessed and urinalysis and blood tests were performed to screen for the occurrence of adverse events at 4 weeks; concurrently, the dosage of sitagliptin was increased to 100 mg/day. After the 12-week treatment period, the same assessments were conducted as those performed at baseline.

Study outcomes

The primary endpoint of this study was the change in amount of pericardial fat, accounting for the sum of epicardial and paracardial fat, as evaluated by magnetic resonance imaging (MRI). The secondary endpoints were the changes from baseline to 12 weeks in the following parameters: (1) myocardial triglyceride content measured by 1H-MRS; (2) cardiac function and mass estimated by echocardiography; (3) indicators of cardiac fatty acid metabolism assessed by iodine-123-β-methyl-iodophenyl pentadecanoic acid myocardial scintigraphy (123I-BMIPP scintigraphy); (4) blood biomarkers (see Table 4 and Additional file 1); (5) body weight and blood pressure; (6) medication compliance; and (7) incidence of adverse events (AEs).

Magnetic resonance imaging (MRI)

Pericardial fat accumulation was estimated by cine MRI with a 1.5 T whole-body MR scanner (MAGNETOM AvantoSQ1.5T B-19; Siemens, Erlangen, Germany) [35, 36]. Four-chamber cine sequences were obtained using a steady-state free precession sequence. Several cine images were used to generate a complete view of the left ventricle (LV) from the base to the apex. Scanning was performed with typical imaging parameters: repetition time, 68.4 ms; echo time, 1.48 ms; flip angle, 80 degrees; matrix, 134 × 192; field of view, 360 × 360 mm; slice thickness, 10 mm; gap, 0 mm; and calculated phases, 25. Epicardial fat and paracardial fat were estimated by a non-participating doctor using dedicated software (SYNAPSE VINCENT, Fujifilm Corporation, Tokyo, Japan). The high signal range between the myocardium and pericardium was designated as the epicardial fat area. Similarly, the high signal range outside the pericardium was considered the paracardial fat area. The pericardial fat area was calculated as the sum of epicardial and paracardial fat areas.

Proton magnetic resonance spectroscopy (1H-MRS)

The myocardial triglyceride content was measured by 1H-MRS analysis using a 1.5 T whole-body MR scanner (MAGNETOM AvantoSQ 1.5 T B-19) performed by specialists with dedicated software (Argus; Siemens), as described previously [36,37,38]. The volume of interest (VOI = 10 × 10 × 20 mm3) was manually placed on the ventricular septum of the cine images of the heart and adjusted to fit the ventricular septum of the LV. The spectra of lipid and water were acquired using point-resolved spectroscopy sequences (repetition time/echo time, 4000/30 ms). The myocardial signal was quantified as the triglyceride signal intensity at 1.4 ppm from the spectra with water suppression, and the water signals were quantified at 4.7 ppm from the spectra without water suppression.

Echocardiography

Echocardiography was performed by experienced technicians with 3.5- and 2.5-MHz transducers for two-dimensional, M-mode, and continuous-wave Doppler measurements. The percent fractional shortening of the ventricle (%FS) was calculated as follows:  %FS = {(left ventricular end-diastolic dimension-left ventricular end-systolic dimension)/left ventricular end-diastolic dimension} × 100.

Iodine-123-β-methyl-iodophenyl pentadecanoic acid myocardial scintigraphy (123I-BMIPP scintigraphy)

Cardiac fatty acid metabolism was assessed by 123I-BMIPP scintigraphy using 111 MBq of an 123I-BMIPP radiotracer (111 MBq/1.5 mg, Nihon Medi-Physics Co., Ltd., Tokyo, Japan). Early images were captured 20 min after injection of the radiotracer, and delay images were captured at 3 h, both with a dual-headed single-photon emission computed tomography (SPECT) gamma camera (Infinia H3000WT; GE Medical System, Tel Aviv, Israel). The SPECT data were acquired in step shooting mode using two detectors (180° rotation) at a matrix size of 64 × 64. A series of contiguous transaxial images with 5.89 mm thickness were reconstructed using the Butterworth filtered back-projection algorithm (order, 10; cut-off, 0.40 cycles/cm) without attenuation or correction. Regional tracer uptake was scored semi-quantitatively from 0 (normal) to 4 (severe defect) for 17 segments of the LV, and the sum of the defect scores for all segments was calculated to derive the summed rest score (SRS) [28]. The heart washout rate between 20 min and 3 h after intravenous injection of 123I-BMIPP was calculated as follows: [(count at 20 min − count at 3 h)/count at 20 min] × 100. The count per pixel data was measured for both the heart and mediastinum, and the ratio of heart-to-mediastinal uptake at 20 min and 3 h after injection of 123I-BMIPP was calculated [39].

Laboratory testing

Blood and urine samples were collected at Toho University Hospital and submitted to the central laboratory of the hospital or a private laboratory (SRL laboratory, Tokyo, Japan). All data were collected after overnight fasting. Administration of any oral hypoglycemic agents, including empagliflozin and sitagliptin, was prohibited on the sampling day. Homeostasis model assessment-insulin resistance (HOMA-IR) was calculated as follows: HOMA-IR = (plasma insulin × plasma glucose)/405.

Safety evaluation

During this study, the investigators continuously monitored any AEs through regular medical checkups. All related AEs, as well as side effects of the drug, abnormal values from the clinical tests, and unusual complaints, were reported and documented.

Sample size calculation

No previous studies have compared the effects of empagliflozin and sitagliptin on cardiac fat accumulation. In addition, while planning this study, no studies were available for estimating the effect of the SGLT2 inhibitor and DPP-4 inhibitor on pericardial fat accumulation. Therefore, since hepatic steatosis was associated with cardiac lipid accumulation and cardiac dysfunction [36, 40,41,42], we referred to previous studies that demonstrated a reduction in the intrahepatic lipid content with sitagliptin administration [43, 44]. Based on these studies, we estimated that 16 subjects per group was sufficient for this study, and assuming a dropout rate of 25%, the target number of patients for enrollment was set as 22 per group.

Statistical analysis

Analyses of the primary and secondary endpoints were performed on the full analysis set (FAS). The FAS includes all research subjects enrolled in this study and assigned to a study treatment. Subjects without primary endpoint data, or those who significantly violated the study protocol, were excluded. Safety analysis with AEs was performed on the safety analysis set, which included all subjects enrolled in this study who were administered all or part of the study treatment. We analyzed the primary endpoint using covariance models, including the baseline intrahepatic lipid content and presence of sulfonylurea medication as assignment factors. To compare categorical variables in the two groups, the Chi-square test was used for nominal variables, and a two-sample t-test was used for continuous variables, as all data were normally distributed. Statistical significance was defined as p < 0.05. All statistical analyses were performed independently by the administrative office of the ASSET study using SAS software version 9.4 (SAS Institute, Cary, NC, USA).

Results

Clinical characteristics

Figure 1 shows the flow diagram of the progress during the phases of this parallel randomized trial. A total of 127 subjects were screened and 83 were ineligible (55 subjects denied consent and 28 did not meet the inclusion criteria). Forty-four subjects were enrolled and randomized. Ultimately, 42 completed the study and were assigned to the FAS. Their baseline clinical characteristics are summarized in Table 1. Most patients were middle-aged (average, 50.3 years old) and overweight (average body mass index, 29.3 kg/m2), and their average HbA1c was 7.1%. The duration of diabetes was short (average 3.5 years), and 19 subjects were drug-naïve before enrollment. Two subjects had microvascular/macrovascular complications (one subject had diabetic nephropathy and another had peripheral arterial disease). There were no differences in any baseline clinical characteristics between the two groups. Although it was recommended to increase the sitagliptin dose to 100 mg/day during the trial, two patients remained at 50 mg/day throughout the study period at the doctor’s discretion.

Fig. 1
figure 1

Flow diagram depicting phases of the parallel randomized trial for empagliflozin and sitagliptin groups. FAS: full analysis set

Table 1 Baseline characteristics of patients in empagliflozin group and sitagliptin group

Cardiac fat accumulation

There was no difference in the change in accumulation of pericardial fat, which is composed of epicardial and paracardial fat, between the two groups (46.8 ± 182.4 vs. −33.0 ± 182.4 mm2, empagliflozin group vs. sitagliptin group, respectively, p = 0.27, Fig. 2a–c). The change in myocardial triglyceride content was also not statistically different between the groups (−0.7 ± 7.0% vs. 0.1 ± 3.2%, p = 0.64, Fig. 2d).

Fig. 2
figure 2

Change in cardiac fat accumulation compared to baseline values. a Pericardial fat accumulation, b epicardial fat accumulation, c paracardial fat accumulation, and d myocardial triglyceride content between empagliflozin and sitagliptin groups. Comparisons were performed by two-sample t-tests. n.s., not significant

Cardiac function and mass assessed by echocardiography

Table 2 shows the echocardiography parameters at baseline and after 12 weeks, as well as the change in each parameter. No significant differences were observed in the changes of each parameter between the two groups. However, LVEF and %FS were significantly decreased only in the sitagliptin group over the 12-week study period (ΔLVEF: −1.6 ± 3.0%, Δ %FS: −1.4 ± 2.7%, both p < 0.05).

Table 2 Echocardiography parameters

Parameters of cardiac fatty acid metabolism assessed by 123I-BMIPP scintigraphy

No differences were observed between the groups in any parameters of the 123I-BMIPP scintigraphy (Table 3). However, SRS was significantly decreased from baseline during the 12 weeks in the sitagliptin group (ΔSRS: −0.35 ± 0.67, p < 0.05).

Table 3 Parameters of iodine-123-β-methyl-iodophenyl pentadecanoic acid myocardial scintigraphy

Physical and metabolic parameters

Body weight was significantly reduced only in the empagliflozin group from baseline to 12 weeks, and no significant difference was observed in body weight reduction between the groups (Table 4). Additionally, HbA1c was significantly decreased to a similar extent in both groups (Table 4). Meanwhile, plasma glucose, plasma insulin, and HOMA-IR were significantly lower in the empagliflozin group than in the sitagliptin group (Table 4). High-density lipoprotein (HDL) cholesterol and apolipoprotein A-I were significantly higher in the empagliflozin group than in the sitagliptin group, whereas low-density lipoprotein cholesterol and apolipoprotein B were similar between the groups (Table 4). Uric acid was significantly decreased in the empagliflozin group compared to the sitagliptin group (Table 4). Total ketone bodies, β-hydroxybutyric acid, and acetoacetic acid were significantly higher in the empagliflozin group than in the sitagliptin group (Table 4). Although the hematocrit level was significantly increased in the empagliflozin group, no significant differences were observed between the groups (Additional file 1). Blood pressure, brain natriuretic peptide (BNP), and heart-type fatty acid-binding protein were not statistically different between the groups (Additional file 1).

Table 4 Physical and metabolic parameters

Safety outcomes

The total number of AEs was significantly higher in the empagliflozin group than in the sitagliptin group (Additional file 2). This was primarily due to increased urination, a common pharmacological effect of SGLT2 inhibitors, in the empagliflozin group [5 (23.8%) vs. 0 (0.0%), p < 0.05]. No patient dropped out of the study due to AEs associated with drug administration.

Discussion

This study demonstrated that 12-week administration of empagliflozin had similar effects as sitagliptin on cardiac fat accumulation and cardiac function in patients with early-stage T2DM, without CVD complications. However, certain cardiometabolic biomarkers were significantly improved in the empagliflozin group compared to those in the sitagliptin group. These changes in cardiometabolic biomarkers by early empagliflozin supplementation may contribute to the primary prevention of CVD.

It has been shown that SGLT2 inhibitors, such as luseogliflozin, ipragliflozin, and canagliflozin significantly reduced epicardial fat accumulation in a 12-week one-arm study [18,19,20]. It was also shown that the reduction with dapagliflozin was larger than that with conventional therapy [21]. However, no changes in pericardial, epicardial, or paracardial fat accumulation following 12 weeks of empagliflozin treatment were assessed in these studies. Moreover, considering that the previous studies were one-arm or placebo-controlled trials in patients with various backgrounds, and the methods for cardiac fat evaluation varied, it is difficult to directly compare the previous results with ours. Nevertheless, patients in the current study were relatively younger (average 52.8 years) with lower HbA1c levels (average 7.1%) and shorter diabetes durations (average 3.5 years) than those reported in the previous studies (mean age range: 55–68 years; mean HbA1c range, 7.1–7.5%) [18,19,20,21]. Furthermore, patients had few microvascular and macrovascular complications in this study (only one subject had diabetic nephropathy and another had peripheral arterial disease). Therefore, these conflicting results might have originated from differences in baseline characteristics, such as the severity of T2DM or the existence and extent of vascular complications. In addition, the method employed for determination of cardiac fat accumulation differed between the current study and the previous studies. Specifically, our study used cine MRI to evaluate epicardial and pericardial fat and myocardial triglyceride content, which has been reported previously by another group [36], but the other studies used whole-heart coronary magnetic resonance angiography [18, 19], echocardiography [20, 22], or cardiac computed tomography [21] to evaluate epicardial fat content. Such differences in evaluation methods may also account for the different results in cardiac fat accumulation between the present and previous studies. Moreover, myocardial triglyceride content was unaffected despite its baseline value being higher in our subjects than in healthy Japanese subjects [37] or patients with left ventricular hypertrophy [38]. This result is consistent with that of a previous one-arm study showing that 6 months of empagliflozin treatment did not impact myocardial triglyceride content [45]. Considering that no clinical studies have assessed the effect of DPP-4 inhibitors on myocardial triglyceride content or compared the effects of SGLT2 inhibitors and DPP-4 inhibitors on cardiac lipid levels, this is the first study to show that SGLT2 inhibitors and DPP-4 inhibitors do not affect pericardial fat accumulation or myocardial triglyceride content at the early-stage of T2DM. Therefore, further studies are necessary to establish the most effective approaches for reducing pericardial and epicardial fat accumulation and myocardial triglyceride content and to clarify the impact of these reductions on CVD prevention.

Additionally, no differences were noted in the effects on cardiac function and mass between the empagliflozin and sitagliptin groups. This is consistent with a previous study showing that dapagliflozin did not alter cardiac function after 12 weeks of treatment [46]. However, other studies enrolling patients with T2DM and chronic heart failure or coronary heart disease demonstrated that 24 weeks of dapagliflozin and empagliflozin treatment improved the ratio of mitral inflow E to mitral e′ annular velocities, an indicator of diastolic function, while reducing left ventricular mass [47, 48]. Considering these previous studies, 12 weeks might have been insufficient to improve cardiac function and structure; furthermore, since cardiac function and structure were well-preserved at baseline in this study, no further improvement may have been detectable following empagliflozin treatment. In addition, a recent study reported that pericardial fat volume was associated with diastolic function even in healthy subjects with normal cardiac function [16], which supports our results showing that no reduction in pericardial fat volume or improvement in diastolic function was observed in parallel. Interestingly, although there was no difference in cardiac function between the groups, sitagliptin significantly decreased LVEF and  %FS from baseline to 12 weeks. Similarly, Mulvihill et al. demonstrated that a DPP-4 inhibitor impaired cardiac function in a rodent model [49] and Li et al. reported that DPP-4 activity was positively correlated with LV systolic function, suggesting that DPP-4 inhibition is correlated with LV systolic dysfunction in humans [50]. Furthermore, various CVD outcome trials have indicated that DPP-4 inhibitors increase the risk of hospitalizations due to heart failure [9, 51]. Unexpectedly, SRS, as assessed by 123I-BMIPP scintigraphy, which indicates myocardial fatty acid uptake and is associated with LV wall motion abnormality [27] as well as the incidence of CVD events [28, 52], showed significantly decreased values from baseline only in the sitagliptin group, although the values were not statistically different between groups. This reduction may be due to the higher baseline values in the sitagliptin group and, thus, may not have clinical implications considering that the patients in the sitagliptin group had no history of CVD events. Indeed, normal cardiac function was observed by echocardiography, which is a common and reliable diagnostic method.

In addition to direct evaluation of the heart, we measured cardiometabolic indices. Although no significant differences were observed in body weights or HbA1c levels between the groups, plasma glucose, plasma insulin, and HOMA-IR were significantly decreased from baseline in the empagliflozin group compared to the sitagliptin group values, indicating that empagliflozin can improve insulin resistance, an independent risk factor for CVD [53, 54]. In addition, empagliflozin preserved the serum levels of HDL cholesterol and apolipoprotein A-I, the latter of which is a component of HDL cholesterol, whereas it was significantly decreased in the sitagliptin group. A previous study reported that the risk of myocardial infarction was increased by approximately 25% for every 5 mg/dL decrease in the serum HDL cholesterol level [55]. The uric acid level, which positively associates with CVD risk through hypertension and vascular damage, was also significantly decreased in the empagliflozin group [56, 57]. Meanwhile, the levels of total ketone bodies, β-hydroxybutyric acid, and acetoacetic acid were markedly higher in the empagliflozin group than in the sitagliptin group. Ketone bodies serve as an energy source for the heart along with glucose and free fatty acids. Particularly, β-hydroxybutyric acid is thought to be a “super fuel” for the heart, as infusion of β-hydroxybutyric acid was shown to improve cardiac function and structural remodeling in rodent models [58, 59]. Therefore, increased ketone bodies may have a protective cardiac function in the empagliflozin group. Although no decrease was observed in BNP or heart-type fatty acid-binding protein levels, this may have been due to normally low levels at baseline, similar to that discussed above for the cardiac parameters. Supporting this, Soga et al. reported that an SGLT2 inhibitor improved BNP only in patients with BNP ≥ 100 pg/mL [60]. Taken together, these results suggest that, compared to sitagliptin, empagliflozin plays a greater role in preventing future CVD in the early stage of diabetes, without CVD complications.

Previously, we reported that linagliptin, a DPP-4 inhibitor, and dapagliflozin both protect endothelial function in patients with early-stage T2DM [61, 62]. In addition, we reported that dapagliflozin was more effective than sitagliptin for lowering HbA1c levels, reducing body weight, and avoiding hypoglycemia in early-stage T2DM, which may lead to CVD prevention [63]. However, the current study is the first to compare the cardiovascular and cardiometabolic effects of empagliflozin and sitagliptin in a randomized controlled trial in patients with early-stage diabetes with no complications of CVD and with preserved cardiac function. Although the direct effects on cardiac lipid accumulation and cardiac function did not differ between the two groups, empagliflozin was superior to sitagliptin for cardiometabolic parameters, such as uric acid, HDL cholesterol, ketone bodies, and insulin sensitivity following only 12 weeks of treatment, which was consistent with a previous report [64]. Further studies are needed to clarify how these results affect long-term cardioprotection; however, early administration of SGLT2 inhibitors would be beneficial for the primary prevention of CVD, as well as secondary prevention.

This study had the following limitations. First, the small sample size and short observation period may partially explain the lack of significant differences in cardiac parameters. Second, patients had a short history of diabetes and no CVD, which may have made it difficult to detect significant differences in cardiac parameters between the groups. Finally, while metformin is highly recommended as the first-line drug in Europe and the United States [65], the use of metformin was avoided in this study to eliminate its effects on insulin sensitivity and cardiac protection in patients who were overweight [66]. Interestingly, it was reported that metformin may moderate CVD outcomes with DPP-4 inhibitor use [67] but SGLT2 inhibitors provide cardioprotective effects regardless of concomitant metformin use [68]. In this regard, this study is the first to compare the cardioprotective effects directly between SGLT2 inhibitors and DPP-4 inhibitors without metformin use in patients with early-stage diabetes without CVD that includes heart failure. Overall, although we comprehensively compared the cardioprotective effects of SGLT2 inhibitors and DPP-4 inhibitors in a randomized controlled trial, clinical studies of a large sample size with subjects of various ethnicities are warranted to confirm our results.

Conclusions

No significant differences were observed in the effects on cardiac fat accumulation, cardiac function, and cardiac fatty acid metabolism between the empagliflozin and sitagliptin groups after 12 weeks of treatment. However, regarding cardiometabolic biomarkers, empagliflozin significantly decreased serum uric acid and increased HDL cholesterol, ketone bodies, and insulin sensitivity compared to the corresponding sitagliptin values. Therefore, early supplementation with SGLT2 inhibitors may be preferable to DPP-4 inhibitors to provide early cardiac protection and primary prevention of CVD in patients with early-stage T2DM and preserved cardiac function.

Availability of data and materials

The datasets analyzed during the current study are available from the corresponding author upon reasonable request.

Abbreviations

AE:

Adverse event

BNP:

Brain natriuretic peptide

CVD:

Cardiovascular disease

DPP-4:

Dipeptidyl peptidase-4

FAS:

Full analysis set

HbA1c:

Glycated hemoglobin

HDL:

High-density lipoprotein

HOMA-IR:

Homeostasis model assessment-insulin resistance

LV:

Left ventricular

LVEF:

Left ventricle ejection fraction

MRI:

Magnetic resonance imaging

SGLT2:

Sodium-glucose cotransporter-2

SPECT:

Single-photon emission computed tomography

SRS:

Summed rest score

T2DM:

Type 2 diabetes mellitus

1H-MRS:

Proton magnetic resonance spectroscopy

123I-BMIPP scintigraphy:

Iodine-123-β-methyl-iodophenyl pentadecanoic acid myocardial scintigraphy

%FS:

Percent fractional shortening

References

  1. Kannel WB, McGee DL. Diabetes and glucose tolerance as risk factors for cardiovascular disease: the Framingham study. Diabetes Care. 1979;2:120–6.

    Article  CAS  PubMed  Google Scholar 

  2. Haffner SM, Lehto S, Rönnemaa T, Pyörälä K, Laakso M. Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med. 1998;339:229–34.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  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.

    Article  CAS  PubMed  Google Scholar 

  5. 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.

    Article  CAS  PubMed  Google Scholar 

  6. 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.

    Article  CAS  PubMed  Google Scholar 

  7. Green JB, Bethel MA, Armstrong PW, Buse JB, Engel SS, Garg J, et al. Effect of sitagliptin on cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2015;373:232–42.

    Article  CAS  PubMed  Google Scholar 

  8. White WB, Cannon CP, Heller SR, Nissen SE, Bergenstal RM, Bakris GL, et al. Alogliptin after acute coronary syndrome in patients with type 2 diabetes. N Engl J Med. 2013;369:1327–35.

    Article  CAS  PubMed  Google Scholar 

  9. Scirica BM, Bhatt DL, Braunwald E, Steg PG, Davidson J, Hirshberg B, et al. Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus. N Engl J Med. 2013;369:1317–26.

    Article  CAS  PubMed  Google Scholar 

  10. Rosenstock J, Perkovic V, Johansen OE, Cooper ME, Kahn SE, Marx N, et al. Effect of Linagliptin vs placebo on major cardiovascular events in adults with type 2 diabetes and high cardiovascular and renal risk: The CARMELINA randomized clinical trial. JAMA. 2019;321:69–79.

    Article  CAS  PubMed  Google Scholar 

  11. Rosito GA, Massaro JM, Hoffmann U, Ruberg FL, Mahabadi AA, Vasan RS, et al. Pericardial fat, visceral abdominal fat, cardiovascular disease risk factors, and vascular calcification in a community-based sample: the Framingham Heart Study. Circulation. 2008;117:605–13.

    Article  PubMed  Google Scholar 

  12. Ding J, Hsu FC, Harris TB, Liu Y, Kritchevsky SB, Szklo M, et al. The association of pericardial fat with incident coronary heart disease: the Multi-Ethnic Study of Atherosclerosis (MESA). Am J Clin Nutr. 2009;90:499–504.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Hirata Y, Tabata M, Kurobe H, Motoki T, Akaike M, Nishio C, et al. Coronary atherosclerosis is associated with macrophage polarization in epicardial adipose tissue. J Am Coll Cardiol. 2011;58:248–55.

    Article  CAS  PubMed  Google Scholar 

  14. Shimabukuro M, Hirata Y, Tabata M, Dagvasumberel M, Sato H, Kurobe H, et al. Epicardial adipose tissue volume and adipocytokine imbalance are strongly linked to human coronary atherosclerosis. Arterioscler Thromb Vasc Biol. 2013;33:1077–84.

    Article  CAS  PubMed  Google Scholar 

  15. Maurovich-Horvat P, Kallianos K, Engel LC, Szymonifka J, Schlett CL, Koenig W, et al. Relationship of thoracic fat depots with coronary atherosclerosis and circulating inflammatory biomarkers. Obesity. 2015;23:1178–84.

    Article  CAS  PubMed  Google Scholar 

  16. de Wit-Verheggen VHW, Altintas S, Spee RJM, Mihl C, van Kuijk SMJ, Wildberger JE, et al. Pericardial fat and its influence on cardiac diastolic function. Cardiovasc Diabetol. 2020;19:129.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. McGavock JM, Lingvay I, Zib I, Tillery T, Salas N, Unger R, et al. Cardiac steatosis in diabetes mellitus: a 1H-magnetic resonance spectroscopy study. Circulation. 2007;116:1170–5.

    Article  PubMed  Google Scholar 

  18. Bouchi R, Terashima M, Sasahara Y, Asakawa M, Fukuda T, Takeuchi T, et al. Luseogliflozin reduces epicardial fat accumulation in patients with type 2 diabetes: a pilot study. Cardiovasc Diabetol. 2017;16:32.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Fukuda T, Bouchi R, Terashima M, Sasahara Y, Asakawa M, Takeuchi T, et al. Ipragliflozin reduces epicardial fat accumulation in non-obese type 2 diabetic patients with visceral obesity: a pilot study. Diabetes Ther. 2017;8:851–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Yagi S, Hirata Y, Ise T, Kusunose K, Yamada H, Fukuda D, et al. Canagliflozin reduces epicardial fat in patients with type 2 diabetes mellitus. Diabetol Metab Syndr. 2017;9:78.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Sato T, Aizawa Y, Yuasa S, Kishi S, Fuse K, Fujita S, et al. The effect of dapagliflozin treatment on epicardial adipose tissue volume. Cardiovasc Diabetol. 2018;17:6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lima-Martinez MM, Paoli M, Rodney M, Balladares N, Contreras M, D’Marco L, et al. Effect of sitagliptin on epicardial fat thickness in subjects with type 2 diabetes and obesity: a pilot study. Endocrine. 2016;51:448–55.

    Article  CAS  PubMed  Google Scholar 

  23. Lee SJ, Lee KH, Oh HG, Seo HJ, Jeong SJ, Kim CH. Effect of sodium-glucose cotransporter-2 inhibitors versus dipeptidyl peptidase 4 inhibitors on cardiovascular function in patients with type 2 diabetes mellitus and coronary artery disease. J Obes Metab Syndr. 2019;28:254–61.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Tadamura E, Tamaki N, Kudoh T, Hattori N, Konishi J. BMIPP compared with PET metabolism. Int J Card Imaging. 1999;15:61–9.

    Article  CAS  PubMed  Google Scholar 

  25. Knapp FF Jr, Ambrose KR, Goodman MM. New radioiodinated methyl-branched fatty acids for cardiac studies. Eur J Nucl Med. 1986;12(Suppl):S39–44.

    Article  CAS  PubMed  Google Scholar 

  26. Li M, Hirano KI, Ikeda Y, Higashi M, Hashimoto C, Zhang B, et al. Triglyceride deposit cardiomyovasculopathy: a rare cardiovascular disorder. Orphanet J Rare Dis. 2019;14:134.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Tateno M, Tamaki N, Yukihiro M, Kudoh T, Hattori N, Tadamura E, et al. Assessment of fatty acid uptake in ischemic heart disease without myocardial infarction. J Nucl Med. 1996;37:1981–5.

    CAS  PubMed  Google Scholar 

  28. Matsuki T, Tamaki N, Nakata T, Doi A, Takahashi H, Iwata M, et al. Prognostic value of fatty acid imaging in patients with angina pectoris without prior myocardial infarction: comparison with stress thallium imaging. Eur J Nucl Med Mol Imaging. 2004;31:1585–91.

    Article  PubMed  Google Scholar 

  29. Ferrannini E, Baldi S, Frascerra S, Astiarraga B, Heise T, Bizzotto R, et al. Shift to fatty substrate utilization in response to sodium-glucose cotransporter 2 inhibition in subjects without diabetes and patients with type 2 diabetes. Diabetes. 2016;65:1190–5.

    Article  CAS  PubMed  Google Scholar 

  30. Carbone S, Canada JM, Billingsley HE, Kadariya D, Dixon DL, Trankle CR, et al. Effects of empagliflozin on cardiorespiratory fitness and significant interaction of loop diuretics. Diabetes Obes Metab. 2018;20:2014–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Carbone S, Billingsley HE, Canada JM, Bressi E, Rotelli B, Kadariya D, et al. The effects of canagliflozin compared to sitagliptin on cardiorespiratory fitness in type 2 diabetes mellitus and heart failure with reduced ejection fraction: the CANA-HF study. Diabetes Metab Res Rev. 2020;36:e3335.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Sezai A, Sekino H, Unosawa S, Taoka M, Osaka S, Tanaka M. Canagliflozin for Japanese patients with chronic heart failure and type II diabetes. Cardiovasc Diabetol. 2019;18:76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Kitazawa T, Seino H, Ohashi H, Inazawa T, Inoue M, Ai M, et al. Comparison of tofogliflozin versus glimepiride as the third oral agent added to metformin plus a dipeptidyl peptidase-4 inhibitor in Japanese patients with type 2 diabetes: a randomized, 24-week, open-label, controlled trial (STOP-OB). Diabetes Obes Metab. 2020;22:1659–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Shigiyama F, Hiruma S, Hisatake S, Shiraga N, Ikeda T, Hirose T, et al. Rationale, Design for the ASSET study: a prospective randomized study comparing empagliflozin’s effect to sitagliptin on cardiac fat accumulation/function in patients with type 2 diabetes. Diabetes Ther. 2019;10:1509–21.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Sicari R, Sironi AM, Petz R, Frassi F, Chubuchny V, De Marchi D, et al. Pericardial rather than epicardial fat is a cardiometabolic risk marker: an MRI vs echo study. J Am Soc Echocardiogr. 2011;24:1156–62.

    Article  PubMed  Google Scholar 

  36. Graner M, Siren R, Nyman K, Lundbom J, Hakkarainen A, Pentikainen MO, et al. Cardiac steatosis associates with visceral obesity in nondiabetic obese men. J Clin Endocrinol Metab. 2013;98:1189–97.

    Article  CAS  PubMed  Google Scholar 

  37. Sai E, Shimada K, Yokoyama T, Sato S, Nishizaki Y, Miyazaki T, et al. Evaluation of myocardial triglyceride accumulation assessed on 1H-magnetic resonance spectroscopy in apparently healthy Japanese subjects. Intern Med. 2015;54:367–73.

    Article  CAS  PubMed  Google Scholar 

  38. Sai E, Shimada K, Yokoyama T, Hiki M, Sato S, Hamasaki N, et al. Myocardial triglyceride content in patients with left ventricular hypertrophy: comparison between hypertensive heart disease and hypertrophic cardiomyopathy. Heart Vessels. 2017;32:166–74.

    Article  PubMed  Google Scholar 

  39. Biswas SK, Sarai M, Toyama H, Yamada A, Harigaya H, Naruse H, et al. Role of 123I-BMIPP and serum B-type natriuretic peptide for the evaluation of patients with heart failure. Singapore Med J. 2012;53:398–402.

    PubMed  Google Scholar 

  40. Rijzewijk LJ, van der Meer RW, Smit JW, Diamant M, Bax JJ, Hammer S, et al. Myocardial steatosis is an independent predictor of diastolic dysfunction in type 2 diabetes mellitus. J Am Coll Cardiol. 2008;52:1793–9.

    Article  PubMed  Google Scholar 

  41. Levelt E, Pavlides M, Banerjee R, Mahmod M, Kelly C, Sellwood J, et al. Ectopic and visceral fat deposition in lean and obese patients with type 2 diabetes. J Am Coll Cardiol. 2016;68:53–63.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Lautamäki R, Borra R, Iozzo P, Komu M, Lehtimäki T, Salmi M, et al. Liver steatosis coexists with myocardial insulin resistance and coronary dysfunction in patients with type 2 diabetes. Am J Physiol Endocrinol Metab. 2006;291:E282–90.

    Article  PubMed  CAS  Google Scholar 

  43. Watanabe T, Tamura Y, Kakehi S, Funayama T, Gastaldelli A, Takeno K, et al. Effects of sitagliptin on ectopic fat contents and glucose metabolism in type 2 diabetic patients with fatty liver: a pilot study. J Diabetes Investig. 2015;6:164–72.

    Article  CAS  PubMed  Google Scholar 

  44. Cui J, Philo L, Nguyen P, Hofflich H, Hernandez C, Bettencourt R, et al. Sitagliptin vs placebo for non-alcoholic fatty liver disease: a randomized controlled trial. J Hepatol. 2016;65:369–76.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Hsu JC, Wang CY, Su MM, Lin LY, Yang WS. Effect of empagliflozin on cardiac function, adiposity, and diffuse fibrosis in patients with type 2 diabetes mellitus. Sci Rep. 2019;9:15348.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Bonora BM, Vigili de Kreutzenberg S, Avogaro A, Fadini GP. Effects of the SGLT2 inhibitor dapagliflozin on cardiac function evaluated by impedance cardiography in patients with type 2 diabetes. Secondary analysis of a randomized placebo-controlled trial. Cardiovasc Diabetol. 2019;18:106.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Tanaka H, Soga F, Tatsumi K, Mochizuki Y, Sano H, Toki H, et al. Positive effect of dapagliflozin on left ventricular longitudinal function for type 2 diabetic mellitus patients with chronic heart failure. Cardiovasc Diabetol. 2020;19:6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. 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.

    Article  PubMed  Google Scholar 

  49. Mulvihill EE, Varin EM, Ussher JR, Campbell JE, Bang KW, Abdullah T, et al. Inhibition of dipeptidyl peptidase-4 impairs ventricular function and promotes cardiac fibrosis in high fat-fed diabetic mice. Diabetes. 2016;65:742–54.

    Article  CAS  PubMed  Google Scholar 

  50. Li JW, Chen YD, Liu YQ, Wang JD, Chen WR, Zhang YQ, et al. Plasma dipeptidyl-peptidase-4 activity is associated with left ventricular systolic function in patients with ST-segment elevation myocardial infarction. Sci Rep. 2017;7:6097.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Kim KJ, Choi J, Lee J, Bae JH, An JH, Kim HY, et al. Dipeptidyl peptidase-4 inhibitor compared with sulfonylurea in combination with metformin: cardiovascular and renal outcomes in a propensity-matched cohort study. Cardiovasc Diabetol. 2019;18:28.

    Article  PubMed  PubMed Central  Google Scholar 

  52. Hashimoto H, Nakanishi R, Mizumura S, Hashimoto Y, Okamura Y, Kiuchi S, et al. Prognostic value of 123I-BMIPP SPECT in patients with nonischemic heart failure with preserved ejection fraction. J Nucl Med. 2018;59:259–65.

    Article  CAS  PubMed  Google Scholar 

  53. Després JP, Lamarche B, Mauriège P, Cantin B, Dagenais GR, Moorjani S, et al. Hyperinsulinemia as an independent risk factor for ischemic heart disease. N Engl J Med. 1996;334:952–7.

    Article  PubMed  Google Scholar 

  54. Patel TP, Rawal K, Bagchi AK, Akolkar G, Bernardes N, Dias DDS, et al. Insulin resistance: an additional risk factor in the pathogenesis of cardiovascular disease in type 2 diabetes. Heart Fail Rev. 2016;21:11–23.

    Article  CAS  PubMed  Google Scholar 

  55. Castelli WP. Cardiovascular disease and multifactorial risk: challenge of the 1980s. Am Heart J. 1983;106:1191–200.

    Article  CAS  PubMed  Google Scholar 

  56. Feig DI, Kang DH, Johnson RJ. Uric acid and cardiovascular risk. N Engl J Med. 2008;359:1811–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Odden MC, Amadu AR, Smit E, Lo L, Peralta CA. Uric acid levels, kidney function, and cardiovascular mortality in US adults: National Health and Nutrition Examination Survey (NHANES) 1988-1994 and 1999-2002. Am J Kidney Dis. 2014;64:550–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Horton JL, Davidson MT, Kurishima C, Vega RB, Powers JC, Matsuura TR, et al. The failing heart utilizes 3-hydroxybutyrate as a metabolic stress defense. JCI Insight. 2019;4:e124079.

    Article  PubMed Central  Google Scholar 

  59. Gormsen LC, Svart M, Thomsen HH, Søndergaard E, Vendelbo MH, Christensen N, et al. Ketone body infusion with 3-hydroxybutyrate reduces myocardial glucose uptake and increases blood flow in humans: a positron emission tomography study. J Am Heart Assoc. 2017;6:e005066.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Soga F, Tanaka H, Tatsumi K, Mochizuki Y, Sano H, Toki H, et al. Impact of dapagliflozin on left ventricular diastolic function of patients with type 2 diabetic mellitus with chronic heart failure. Cardiovasc Diabetol. 2018;17:132.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Shigiyama F, Kumashiro N, Miyagi M, Iga R, Kobayashi Y, Kanda E, et al. Linagliptin improves endothelial function in patients with type 2 diabetes: a randomized study of linagliptin effectiveness on endothelial function. J Diabetes Investig. 2017;8:330–40.

    Article  CAS  PubMed  Google Scholar 

  62. Shigiyama F, Kumashiro N, Miyagi M, Ikehara K, Kanda E, Uchino H, et al. Effectiveness of dapagliflozin on vascular endothelial function and glycemic control in patients with early-stage type 2 diabetes mellitus: DEFENCE study. Cardiovasc Diabetol. 2017;16:84.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Fuchigami A, Shigiyama F, Kitazawa T, Okada Y, Ichijo T, Higa M, et al. Efficacy of dapagliflozin versus sitagliptin on cardiometabolic risk factors in Japanese patients with type 2 diabetes: a prospective, randomized study (DIVERSITY-CVR). Cardiovasc Diabetol. 2020;19:1.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Shao SC, Chang KC, Lin SJ, Chien RN, Hung MJ, Chan YY, et al. Favorable pleiotropic effects of sodium glucose cotransporter 2 inhibitors: head-to-head comparisons with dipeptidyl peptidase-4 inhibitors in type 2 diabetes patients. Cardiovasc Diabetol. 2020;19:17.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Buse JB, Wexler DJ, Tsapas A, Rossing P, Mingrone G, Mathieu C, et al. 2019 Update to: management of hyperglycemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care. 2020;2020(43):487–93.

    Article  CAS  Google Scholar 

  66. Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year follow-up of intensive glucose control in type 2 diabetes. N Engl J Med. 2008;359:1577–89.

    Article  CAS  PubMed  Google Scholar 

  67. Crowley MJ, Williams JW Jr, Kosinski AS, D’Alessio DA, Buse JB. Metformin use may moderate the effect of DPP-4 inhibitors on cardiovascular outcomes. Diabetes Care. 2017;40:1787–9.

    Article  PubMed  Google Scholar 

  68. Neuen BL, Arnott C, Perkovic V, Figtree G, de Zeeuw D, Fulcher G, et al. Sodium-glucose co-transporter-2 inhibitors with and without metformin: a meta-analysis of cardiovascular, kidney and mortality outcomes. Diabetes Obes Metab. 2021;23:382–90.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank all participants and staff of this prospective study. We thank Hirokazu Yamada and Soiken Holdings, Inc., Tokyo, Japan for their excellent assistance with the statistical analysis. We would like to thank Editage (www.editage.jp) for English language editing.

Funding

This study was funded by the Boehringer Ingelheim & Eli Lilly and Company Diabetes Alliance. Both companies had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

NK designed the study. SHiruma , FS, and NK conducted the study. SHiruma , FS, and NK wrote and edited the manuscript. TH contributed to the discussion. SHisatake, SM, and TI managed cardiac examinations such as echocardiography and 123I-BMIPP scintigraphy. NS and MH managed image examination such as cardiac MRI and 1H-MRS. NK is the guarantor of this work and, as such, had full access to all study data and takes responsibility for the integrity of the data and accuracy of data analysis. All named authors meet the International Committee of Medical Journal Editors criteria for authorship for this article, take responsibility for the integrity of the work, and have given their approval for this version to be published. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Naoki Kumashiro.

Ethics declarations

Ethics approval and consent to participate

All investigators involved in this study followed the “World Medical Association Declaration of Helsinki” (2013 revision), and “Ethical Guidelines for Medical and Health Research Involving Human Subjects” (December 22, 2014, Ministry of Education, Culture, Sports, Science and Technology/Ministry of Health, Labor and Welfare), and other laws and regulations. Written informed consent was obtained from all participants.

Consent for publication

Not applicable.

Competing interests

NK received research funds from Nippon Boehringer Ingelheim Co., Ltd., and lecture fees from Takeda Pharmaceutical Company Ltd.; Ono Pharmaceutical Co., Ltd.; Mitsubishi Tanabe Pharma Corporation; and Nippon Boehringer Ingelheim Co., Ltd. TH received research funds from AstraZeneca K.K., Mitsubishi Tanabe Pharma Corporation, and Novo Nordisk Pharma Ltd., and lecture fees from Sanofi K.K.; Eli Lilly Japan K.K.; Novo Nordisk Pharma Ltd.; Sumitomo Dainippon Pharma Co., Ltd.; Ono Pharmaceutical Co., Ltd.; AstraZeneca K.K.; Mitsubishi Tanabe Pharma Corporation; and Kowa Company, Limited. All funding agencies had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript. SH1, FS, SH2, SM, NS, MH, and TI declare that they have no conflicts of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Additional file 1.

Additional patient parameters (physical and biochemical parameters that showed no significant differences between the empagliflozin and sitagliptin groups).

Additional file 2.

Adverse events (list of observed adverse events).

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hiruma, S., Shigiyama, F., Hisatake, S. et al. A prospective randomized study comparing effects of empagliflozin to sitagliptin on cardiac fat accumulation, cardiac function, and cardiac metabolism in patients with early-stage type 2 diabetes: the ASSET study. Cardiovasc Diabetol 20, 32 (2021). https://doi.org/10.1186/s12933-021-01228-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12933-021-01228-3

Keywords