838
Views
0
CrossRef citations to date
0
Altmetric
Pharmacology

The effect of sodium-glucose co-transporter-2 inhibitors on markers of subclinical atherosclerosis

, ORCID Icon, , , &
Article: 2304667 | Received 22 Aug 2023, Accepted 19 Dec 2023, Published online: 17 Jan 2024

Abstract

Background

Despite the widespread use of classical cholesterol-lowering drugs to mitigate the adverse impacts of dyslipidaemia on atherosclerosis, many patients still face a substantial residual risk of developing atherosclerotic cardiovascular disease (CVD). This risk is partially attributed to non-traditional pathophysiological pathways. Latest evidence suggests that sodium glucose co-transporter-2 (SGLT2) inhibitors are beneficial for patients suffering from type 2 diabetes mellitus (T2DM) or established CVD by reducing morbidity and mortality. However, the underlying mechanisms of this benefit have not been clearly elucidated. It has been hypothesized that one possible mechanism could be the attenuation of subclinical atherosclerosis (SA) progression.

Aim

The objective of this narrative review is to examine the present evidence concerning the impact of SGLT2 inhibitors on markers of SA.

Results

The current evidence on the efficacy of SGLT2 on SA, endothelial function and arterial stiffness remains controversial. Findings from observational and randomized studies are quite heterogeneous; however, they converge that the antiatherosclerotic activity of SGLT2 inhibitors is not strong enough to be widely used for prevention of atherosclerosis progression in patients with or without T2DM.

Conclusions

Further research is needed to investigate the underlying mechanisms and the possible beneficial impact of SGLT2i on primary and secondary CVD prevention through attenuation of premature atherosclerosis progression.

Introduction

Atherosclerosis is a complex and multifactorial inflammatory process that commences in childhood but often remains asymptomatic for a long period of time before presenting clinically later in life [Citation1,Citation2]. Accumulating evidence suggests that atherosclerosis should be considered as an immune system-mediated process of the vascular system [Citation3]. Besides, many experimental studies have shown that atherosclerotic plaques are composed of different immune cells like lymphocytes and macrophages [Citation4].

Although classical cholesterol-lowering drugs have been widely used to lessen the negative effects of dyslipidaemia on atherosclerosis, many patients still experience a significant residual risk for developing atherosclerotic cardiovascular disease (CVD), driven by non-traditional pathophysiological pathways of atherogenesis, such as Lp(a), autoimmunity and glucometabolic mediators [Citation5]. Thus, there is a substantial and unmet need to optimize the anti-atherosclerotic therapeutic strategies in order to mitigate the atherosclerotic risk and minimize the occurrence of major adverse cardiovascular events (MACEs).

Latest evidence from landmark trials suggests that sodium glucose co-transporter-2 (SGLT2) inhibitors are beneficial for patients suffering from type 2 diabetes mellitus (T2DM) or established CVD – mainly heart failure (HF) – by reducing CVD-related morbidity and mortality [Citation6,Citation7]. Notwithstanding the significant amount of evidence in the field, the precise underlying mechanisms that underpin these advantageous outcomes are still not fully understood. Thus, the actual impact of SGLT-2 inhibitors on subclinical atherosclerosis (SA) markers remains controversial. Besides, the existing evidence is scarce regarding their effects on primary and secondary CVD prevention through attenuation of premature atherosclerosis progression in patients with or without underlying T2DM.

The detection of the exact burden of SA in the general population is practically infeasible [Citation1]. Currently, many non-invasive techniques have been proposed and incorporated into clinical practice, which can effectively contribute to the early detection of SA before its clinical manifestation [Citation1, Citation8]. These advancements in technology have enabled the clinicians to assess the cardiovascular (CV) risk in a more practical and accurate manner. Unlike the traditional approach that solely relies on CVD risk factors, the new methods also take into account the vascular damage and resulting dysfunction, which is an earlier manifestation of atherosclerosis in asymptomatic adults [Citation9]. In addition, arterial stiffness mainly assessed by pulse wave velocity (PWV) and augmentation index (AIx) has been described as target-organ damage [Citation10,Citation11]. Accumulating evidence suggests that arterial stiffness is strongly related with the incidence, and with the extent of atherosclerotic disease and thus, it should be considered as an independent risk factor of CVD-related outcomes [Citation8, Citation10–12]. The early diagnosis of premature atherosclerotic disease is of paramount significance, as it paves the way for timely interventions through primary prevention strategies [Citation1].

The aim of this narrative review is to provide a comprehensive analysis of the current evidence on the effects of SGLT2 inhibitors on markers of SA. The focus is primarily on clinical data from observational and randomized studies that assess the impact of SGLT2 inhibitors on intima-media thickness (IMT), epicardial adipose tissue (EAT) and indices of endothelial function and arterial stiffness.

SGLT2 inhibitors, mechanism of action and current indications for use

SGLT-2 inhibitors are a relatively new class of oral antidiabetic drugs that act by inhibiting renal sodium and glucose reabsorption by lowering the renal threshold for glucose absorption in the proximal tubule, thereby causing glycosuria and natriuresis [Citation13]. SGLT-2 inhibitors reduce plasma glucose concentration in patients with hyperglycaemia, but they have a trivial impact on plasma glucose concentrations in normoglycaemic individuals [Citation14]. Except for their glucose-lowering effects, they have been associated with weight loss, blood pressure reduction, and a lower risk of hypoglycaemia compared to commonly prescribed antidiabetic drugs, like insulin and sulfonylureas [Citation7, Citation14,Citation15]. In clinical practice, SGLT-2 inhibitors are used for the treatment of T2D as a second-line agent after inadequate glycaemic control using metformin, or a first-line agent for high-risk individuals with CVD, HF or chronic kidney disease (CKD) [Citation16–19]. In addition, they have recently received an indication for administration to patients with HF across the spectrum of left ventricular ejection fraction according to the recent guidelines of the American Heart Association [Citation20]. This indication was based on accumulative evidence regarding their beneficial effects on reducing HF hospitalizations and CVD mortality [Citation21], establishing the new fundamental role of SGLT2 inhibitors in the treatment of HF. In addition to the established CV benefits, SGLT-2 inhibitors have a renoprotective profile irrespectively of the severity or the aetiology of kidney disease, as well as of the diabetes status [Citation22]. However, their impact on indices of SA remains controversial.

Potential underlying mechanisms of SGLT2 inhibitors related to atherosclerosis

It is well known that inflammation plays a significant role in atherogenesis and progression of atherosclerosis [Citation3], and particularly in patients with T2DM inflammation in combination with oxidative stress, is considered as main underlying mechanisms involved in the pathogenesis of atherosclerosis [Citation23]. Indeed, many studies have demonstrated that recruitment, activation and differentiation of monocytes, polarization of macrophages, as well as activation of inflammasome and secretion of proinflammatory cytokines are strongly associated not only with the development but also with the vulnerability of the atherosclerotic plaques [Citation3, Citation24,Citation25]. Accumulating evidence from animal and human studies suggest that treatment with SGLT2 inhibitors reduces inflammatory cell infiltration in atherosclerotic plaques [Citation26–28], decreases circulating inflammatory cytokines, such as CRP, TNF-α, IL-6 and TGF-β [Citation23, Citation26,Citation27], and modulates the activity of NLRP-3 inflammasome leading to decreased release of IL-1β from macrophages [Citation29].

An early vascular change that is strongly related to atherogenesis and progression of SA is the endothelial dysfunction [Citation30]. It is well established that augmented endothelial reactivity as well as proliferation of vascular smooth muscle cells (VSMC) can provoke significant vascular damages and poor relaxation of the arteries [Citation31], particularly in patients with T2DM, due to insulin resistance, increased body weight (BW) and impaired blood glucose levels [Citation32]. The underlying pathophysiologic mechanisms that probably are involved include the elevated reactive oxygen species (ROS), the impaired synthesis of nitric oxide (NO) and imbalanced release of chemokines [Citation33,Citation34]. Evidence from animal and human studies suggest that administration of SGLT2 inhibitors potentially act beneficially on vascular endothelial reactivity [Citation35–37], and attenuate VSMC proliferation leading to reduction of oxidative stress and stability of atherosclerotic plaques [Citation38,Citation39].

In addition, as microRNAs (miRNAs) regulate majority of physiological and pathological processes in human, they can also modulate inflammatory and thrombotic pathways involved in atherogenesis. Several miRNAs have been demonstrated as diagnostic and prognostic tools for evaluating the likelihood of plaque destabilization and rupture and, consequently, the elevated risk of clinical atherosclerosis [Citation40]. In particular, decreased levels of miR-143, and increased of miR-34a are associated with endothelial dysfunction and have been associated with elevated risk of progression of atherosclerosis and plaque rupture [Citation41]. In addition, there is accumulative evidence that new anti-diabetic medications, including SGLT2 inhibitors, can modulate miRNAs expression levels and can exert beneficial effects on endothelial cells, VSMC, macrophages and platelets through reducing oxidative stress and inflammation [Citation42]. Recently, it was shown that empagliflozin downregulates miR-34a-5p in NAFLD-associated fibrosis [Citation43] and reduces miR-92 and mi-21 levels in HFpEF patients [Citation44]. Hence, more dedicated investigation is imperative to determine that miRNAs hold potential utility as biomarkers or treatment targets in the context of subclinical and clinical atherosclerosis in routine clinical practice.

Evidence from animal models and clinical studies is controversial regarding the impact of SGLT2 inhibitors on lipid metabolism (LDL, HDL and triglycerides) [Citation26,Citation27, Citation45], findings that have been confirmed by several meta-analyses [Citation46,Citation47]. Therefore, it is considered less possible that change in lipid metabolism is the main pathophysiologic pathway by which SGLT2 inhibitors exhibit their beneficial CV effects [Citation32, Citation48]. Finally, glycaemia and improved glycaemic control have been described as a putative underlying mechanism for attenuation of SA driven by newer antidiabetic drug classes [Citation49]. Despite that SGLT2 inhibitors can moderately decrease blood glucose levels through glycosuria [Citation50], many other antidiabetic drugs, including sulfonylureas, thiazolidinediones and insulin, have repeatedly failed to show CV benefit [Citation51], indicating that a glucose-independent mechanism probably plays a more important role [Citation32, Citation48].

SGLT2 inhibitors and intima-media thickness

One of the most widespread techniques used for the assessment of SA is the IMT of carotids and femoral arteries by ultrasound imaging [Citation1]. Until now, several clinical studies have investigated the impact of SGLT2 inhibitors on the progression of SA in patients with T2DM (). In their multicentre randomized control trial (RCT), Tanaka et al. found out that ipragliflozin had a neutral effect on IMT of common carotid artery (CCA-IMT) after 2 years of follow-up. Patients in the ipragliflozin arm experienced a mean change in CCA-IMT of 0.0013 mm (95% CI: −0.0155 to 0.0182), while patients in the group of non-SGLT2 inhibitors standard antidiabetic care had a mean change in CCA-IMT of 0.0015 mm (95% CI: −0.0155 to 0.0184), with the estimated difference between the two groups being −0.0001 mm (95% CI: −0.0191 to 0.0189; p = .989) [Citation52]. Similar were the findings of a multicentre, single-arm, observational cohort (FUSION) study from Japan, which showed that treatment with ipragliflozin had a non-significant effect on SA (CCA-IMT = 0.76 ± 0.16 mm at baseline vs. 0.75 ± 0.15 mm after 1 year, p = .40 in right CCA; and CCA-IMT = 0.75 ± 0.18 mm at baseline vs. 0.76 ± 0.17 mm after 1 year, p = .41 in left CCA) [Citation56]

Table 1. Summary of observational studies and RCTs assessing the effects of SGLT-2 inhibitors on indices of subclinical atherosclerosis.

On the other hand, findings from UTOPIA trial revealed that after 2 years of follow-up, both patients receiving tofogliflozin (intervention arm) and non-SGLT2 inhibitors treatment (control group) experienced a significant reduction not only in mean CCA-IMT (mean change = −0.132 mm; SE 0.007 and −0.140 mm; SE 0.006, respectively), but also in maximum left and right CCA-IMT measurements compared to baseline, without important differences in the progression of SA being observed between the two groups (mean change of CCA-IMT = 0.008 mm; 95% CI: [–0.009 to 0.025], p = .34) [Citation54]. However, in a post hoc analysis, tofogliflozin as well as non-SGLT2i hypoglycaemic treatment had a neutral effect on tissue characteristics of the carotid arterial wall as measured by mean, right and left grey-scale median (GSM-CCA) values, and these findings were robust even after adjustments for possible confounding factors like traditional CVD-related risk factors and baseline treatment [Citation53].

According to another single-centre observational cohort study from Turkey on patients with T2D who were treated with empagliflozin, a significant reduction in CCA-IMT was reported (mean change from baseline at 6 months: −1.29 mm, 95% CI: [–1.91 to −0.69], p < .001)) [Citation55]. Similar were the findings reported by Irace et al. [Citation57], who concluded that treatment with empagliflozin was associated with a significant reduction in indices of SA after 1 and 3 months (CCA-IMT = 0.793 ± 0.15 mm and 0.766 ± 0.127 mm; p < .0001 respectively compared to baseline measurements: CCA-IMT = 0.831 ± 0.156 mm). However, their non-randomized, single-arm design with small sample sizes limit the reliability and generalizability of the results, and so they should be interpreted with caution.

SGLT2i and endothelial dysfunction

Endothelial dysfunction is an early vascular change that is strongly related to atherogenesis and progression of SA. Limited research has been conducted regarding the potential influence of SGLT2i on endothelial function (), mainly through assessment of flow-mediated vasodilation (FMD) measurements in the brachial artery. According to the DEFENCE study, a multicentre parallel-group RCT from Japan, it was shown that treatment with dapagliflozin as add-on therapy to metformin was not associated with significant changes in FMD compared to control group (ΔFMD% [FMD at week 16 − FMD at baseline] = 0.85 ± 2.71 and −0.19 ± 2.51, p = .09, respectively) [Citation58]. Nevertheless, subgroup analysis revealed that dapagliflozin was associated with significant improvement of FMD among individuals with HbA1c >7.0% (p = .041) [Citation58]. A sub-analysis of the multicentre PROTECT study reached similar results, as it was shown that treatment with ipragliflozin had a neutral effect on change of FMD measurements at 24 months compared to baseline (5.2 ± 2.6% vs. 5.2 ± 2.6%, p = .98) [Citation59]. In addition, findings from another double-blind, placebo-controlled RCT involving patients suffering from T2DM and established CVD demonstrated that treatment with dapagliflozin for 12 weeks as an add-on therapy to metformin and insulin was not associated with significant improvements either in FMD or nitroglycerin-mediated dilation (NMD) values [Citation61]. Moreover, the EMBLEM trial, a multicentre, placebo-controlled RCT, which was conducted in Japan, demonstrated that treatment with empagliflozin for 6 months was not associated with significant changes in values of reactive hyperaemia peripheral arterial tonometry index (RHI) compared to control arm in 117 patients with T2DM and established CVD [Citation63].

Table 2. Summary of observational studies and RCTs assessing the effects of SGLT-2 inhibitors on indices of endothelial function.

On the other hand, according to a single-centre, parallel-group RCT from Brazil among individuals with T2DM and early atherosclerosis, treatment with dapagliflozin was associated with a significant increase in rest FMD ([+3.3 (8.2)%], compared to treatment with glibenclamide [–1.2 (7.5)%], p = .0001), indicating that SGLT2i could probably have a beneficial impact on CVD prevention among patients suffering from T2D and SA [Citation60]. Sposito et al. reached similar findings, as they reported that treatment with empagliflozin was associated with significant improvements in FMD values at 1 and 2 min (p < .05), according to the single-centre, randomized EXCEED-BHS3 Trial [Citation62]. These beneficial effects of SGLT2i on endothelial function were confirmed by two recent meta-analyses [Citation64,Citation65]. The first one pooled data from 12 RCTs and more than 18,000 patients with T2D and showed that dapagliflozin significantly improved FMD (MD = 1.22, 95% CI: [0.38–2.06]; p = .005) compared to control group [Citation65]. The other meta-analysis synthesized data from nine RCTs and two observational studies pooling data from 868 patients and it demonstrated that antidiabetic treatment with SGLT2 inhibitors was associated with significant improvements in FMD levels compared to non-SGLT2i arms (standardized MD = 0.18, 95% CI: [0.02–0.34]; p = .03) [Citation64].

SGLT2 inhibitors and arterial stiffness

It is well established that arterial stiffness plays a pivotal and independent role in the development of CVD and in clinical practice, PWV is widely used to quantify the level of arterial stiffness [Citation66]. Until now, several clinical studies have investigated the impact of SGLT2 inhibitors on arterial stiffness in patients with T2DM; however, their findings are contradictory ().

Table 3. Summary of observational studies and RCTs assessing the effects of SGLT-2 inhibitors on indices of arterial stiffness.

According to findings of a prespecified sub-analysis of UTOPIA trial, treatment with tofogliflozin was associated with significant improvements in mean, right and left brachial-ankle PWV compared to control group (–104.7 cm/s, 95% CI: [–177.0 to − 32.4]; p = .005, –109.3 cm/s, 95% CI: [–184.3 to –34.3]; p = .005 and –98.3 cm/s, 95% CI: [–172.6 to –24.1]; p = .01, respectively), even after adjusting for classical CVD RFs [Citation67]. In another double-blind, placebo-controlled RCT conducted in Germany, Bosch et al. concluded that treatment with empagliflozin was associated with significant improvements in arterial stiffness indices like central SBP (113.6 ± 12.1 vs. 118.6 ± 12.9 mmHg, p < .001), and central pulse pressure (39.1 ± 10.2 vs. 41.9 ± 10.7 mmHg, p = .027) compared to control group, indicating that empagliflozin possibly affects beneficially the vessels partially through anti-inflammatory pathways [Citation70]. In addition, a single-centre double-blind RCT conducted in Greece demonstrated that treatment with dapagliflozin was associated with significant reductions in 24 h HR-adjusted AIx and with a significant difference in change of estimated 24-h PWV (–0.16 ± 0.32 vs. 0.02 ± 0.27; p = .007) compared to placebo group [Citation71]. Similar were the findings of a prospective observational study from Spain involving 32 adults with T2DM administered dapagliflozin for 1 year. Treatment with dapagliflozin improved arterial stiffness, as it led to significant reduction in carotid-femoral PWV (–9.1 m/s, 95% CI: [8.4–10.1] vs. −9.65 m/s, 95% CI: [8.75–11.2]; p = .02) [Citation73]. Besides, one of the earlier pilot studies conducted in the field, Solini et al., demonstrated that compared to control group, treatment with SGLT2 inhibitors only for two days significantly improved both endothelial function and arterial stiffness by increasing FMD (2.8 ± 2.2 vs. 4.0 ± 2.1%, p < .05), and reducing PWV (10.1 ± 1.6 to 8.9 ± 1.6 m/s, p < .05), even after adjusting for confounding factors like BP [Citation74].

On the other hand, a small single-centre RCT from United Kingdom demonstrated that treatment with dapagliflozin as an add-on therapy to ramipril in T2DM patients with eGFR >60 mL/min and residual microalbuminuria was not associated with significant improvements in mean aortic PWV compared to control group (9.06 ± 1.91 m/s at baseline, 9.13 ± 2.03 m/s after 24 weeks, mean change = −0.5 m/s, 95% CI: [−1 to 2]; p = .84 in the SGLT2i group vs. 9.88 ± 2.12 m/s at baseline, 10.0 ± 1.84 m/s after 24 weeks, mean change = 0.12 m/s, 95% CI: [−0.89 to 1.13]; p = .81 in the control group) [Citation69]. According to a single-centre retrospective observational study from Korea, treatment with dapagliflozin for 6 months was not associated with significant improvements either in aorta or in extremities PWV values [Citation72]. However, subgroup analysis demonstrated that treatment with dapagliflozin was associated with significant reduction in aortic PWV (6.76 ± 1.51 m/s vs. 7.33 ± 1.48 m/s at baseline, p < .01) only among participants who lost BW [Citation72], indicating that BW loss is probably one of the main underlying mechanisms related to CVD protection of SGLT2 inhibitors administration.

A recent meta-analysis conducted by Patoulias et al. synthesizing data from 6 studies and 452 subjects showed that antidiabetic treatment with SGLT2i compared to non-SGLT2i group was associated with a non-significant reduction in PWV (MD = −0.14 m/s, 95% CI [−0.31 to 0.03], I2 = 63%, p = .11) [Citation77]. Notably, subgroup analysis including only T2DM patients (excluding HFrEF patients) demonstrated that treatment with SGLT2 inhibitors was associated with a significant reduction in PWV (MD = −0.17 m/s, 95% CI: [−0.31 to −0.04], I2 = 0%, p = .01) [Citation77]. Also, in their meta-analysis, pooling data from five studies, Wei et al. reached similar findings, as it was shown that treatment with SGLT2 inhibitors did not lead to significant changes in PWV measurements compared to controls (standardized MD: 0.11, 95% CI: [–0.15 to 0.37]; p = .4) [Citation64].

SGLT2 inhibitors and epicardial adipose tissue

It has been previously described that EAT has atherogenic properties [Citation78], as it has been associated with the progression of the SA and clinical manifestations of CVD mainly the coronary artery disease [Citation79,Citation80]. Only a few studies have investigated the possible impact of SGLT2 inhibitors on EAT (). According to a single-centre RCT conducted in Japan, treatment with dapagliflozin for 6 months was associated with significant reduction in EAT volume measured by CT and this improvement was greater in the SGLT2i arm compared to the control group (mean change from baseline in EAT volume = –16.4 ± 8.3 vs. 4.7 ± 8.8 cm3, p = .01) [Citation83]. In addition, a small observational study among 15 adults with T2D demonstrated that canagliflozin was associated with significant reductions in EAT thickness assessed by echocardiography at 3 months, as well as at 6 months compared to baseline [Citation84].

Table 4. Summary of observational studies and RCTs assessing the effects of SGLT-2 inhibitors on EAT thickness.

Similar were the findings from two small single-centre, single-arm, pilot studies conducted in Japan [Citation85,Citation86]. The first one showed that 12-week treatment with luseogliflozin was associated with significant decrease of EAT volume measured by MRI compared to baseline (117 [96–136] cm3 to 111 [88–134] cm3, p = .048) and this reduction was strongly correlated with the attenuation of systematic inflammation evaluated by CRP (r = 0.493, p = .019) [Citation85]. The second pilot study also demonstrated that treatment with ipragliflozin for 12 weeks was associated with significant decrease of EAT volume measured by MRI compared to baseline (from 102 [79–126] cm3 to 89 [66–109] cm3, p = .008) [Citation86]. Notably, according to single-centre RCT from USA, SGLT2 inhibitors show a beneficial profile also in non-diabetic patients with HFrEF [Citation81]. Indeed, treatment with empagliflozin significantly reduced EAT volume evaluated by MRI (–5.14 mL, 95% CI: [–8.36 to −1.92]) compared to control group (–0.75 mL, 95% CI: [–3.57 to 2.06]; p < .05) [Citation81]. The beneficial effect of SGLT2 inhibitors on EAT volume was confirmed by a recent meta-analysis pooling data from 8 studies and 221 patients, which demonstrated that SGLT2 inhibitors significantly reduced EAT thickness (standardized MD = −0.552, 95% CI: [−0.79 to −0.32]; I2 = 67.6%, p < .001) [Citation87].

Conclusions

Despite the advantages of SGLT2 inhibitors for CV health are well known, there have been ongoing debate regarding the impact of these agents on markers of SA, endothelial function and arterial stiffness. Although the results of observational and randomized trials are extremely diverse, they all converge to the fact that SGLT2 inhibitors’ antiatherosclerotic effect is not sufficiently potent to halt the development of atherosclerosis in individuals with or without T2DM. The underlying processes and potential positive effects of SGLT2i on primary and secondary CVD prevention by reducing early atherosclerosis development should be further investigated in the future.

Author contributions

Panagiotis Stachteas: conception and design, analysis and interpretation of the data, drafting and revising of the paper; Paschalis Karakasis: drafting and revising of the paper; Dimitrios Patoulias: conception and design, drafting and revising of the paper; Francesco Clemenza: critical revision of the paper; Nikolaos Fragakis: revising of the paper critically for intellectual content; Manfredi Rizzo: conception and design, revising of the paper critically for intellectual content. All authors have read and approved the final version of the manuscript to be published. All authors agree to be accountable for all aspects of the work.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Data availability statement

This is a review article analysing secondary data, so data sharing is not applicable.

Additional information

Funding

The author(s) reported there is no funding associated with the work featured in this article.

References

  • Gatto L, Prati F. Subclinical atherosclerosis: how and when to treat it? Eur Heart J Suppl. 2020;22(Suppl. E):1–13. doi: 10.1093/eurheartj/suaa068.
  • Lefkou E, Fragakis N, Ioannidou E, et al. Increased levels of proinflammatory cytokines in children with family history of coronary artery disease. Clin Cardiol. 2010;33(4):E6–E10.
  • Libby P, Ridker PM, Hansson GK, et al. Inflammation in atherosclerosis. J Am Coll Cardiol. 2009;54(23):2129–2138. doi: 10.1016/j.jacc.2009.09.009.
  • Shoenfeld Y, Sherer Y, Harats D. Atherosclerosis as an infectious, inflammatory and autoimmune disease. Trends Immunol. 2001;22(6):293–295. doi: 10.1016/s1471-4906(01)01922-6.
  • Sampson UK, Fazio S, Linton MF. Residual cardiovascular risk despite optimal LDL cholesterol reduction with statins: the evidence, etiology, and therapeutic challenges. Curr Atheroscler Rep. 2012;14(1):1–10. doi: 10.1007/s11883-011-0219-7.
  • Patoulias D, Fragakis N, Rizzo M. The therapeutic role of SGLT-2 inhibitors in acute heart failure: from pathophysiologic mechanisms to clinical evidence with pooled analysis of relevant studies across safety and efficacy endpoints of interest. Life. 2022;12(12):2062. doi: 10.3390/life12122062.
  • Heerspink HJL, Perkins BA, Fitchett DH, et al. Sodium glucose cotransporter 2 inhibitors in the treatment of diabetes mellitus. Circulation. 2016;134(10):752–772. doi: 10.1161/CIRCULATIONAHA.116.021887.
  • Vlachopoulos C, Xaplanteris P, Aboyans V, et al. The role of vascular biomarkers for primary and secondary prevention. A position paper from the European Society of Cardiology Working Group on peripheral circulation. Atherosclerosis. 2015;241(2):507–532. doi: 10.1016/j.atherosclerosis.2015.05.007.
  • Williams B, Mancia G, Spiering W, et al. 2018 ESC/ESH guidelines for the management of arterial hypertension. Eur Heart J. 2018;39(33):3021–3104. doi: 10.1093/eurheartj/ehy339.
  • Mitchell GF, Hwang S-J, Vasan RS, et al. Arterial stiffness and cardiovascular events. Circulation. 2010;121(4):505–511. doi: 10.1161/CIRCULATIONAHA.109.886655.
  • Laurent S, Boutouyrie P, Asmar R, et al. Aortic stiffness is an independent predictor of all-cause and cardiovascular mortality in hypertensive patients. Hypertension. 2001;37(5):1236–1241. doi: 10.1161/01.hyp.37.5.1236.
  • Vlachopoulos C, Aznaouridis K, Stefanadis C. Prediction of cardiovascular events and all-cause mortality with arterial stiffness. J Am Coll Cardiol. 2010;55(13):1318–1327. doi: 10.1016/j.jacc.2009.10.061.
  • Giglio RV, Patti AM, Rizvi AA, et al. Advances in the pharmacological management of diabetic nephropathy: a 2022 international update. Biomedicines. 2023;11(2):291. doi: 10.3390/biomedicines11020291.
  • Zaccardi F, Webb DR, Htike ZZ, et al. Efficacy and safety of sodium-glucose co-transporter-2 inhibitors in type 2 diabetes mellitus: systematic review and network meta-analysis. Diabetes Obes Metab. 2016;18(8):783–794. doi: 10.1111/dom.12670.
  • Hsia DS, Grove O, Cefalu WT. An update on sodium-glucose co-transporter-2 inhibitors for the treatment of diabetes mellitus. Curr Opin Endocrinol Diabetes Obes. 2016;24(1);73–79.
  • Vardeny O, Vaduganathan M. Practical guide to prescribing sodium-glucose cotransporter 2 inhibitors for cardiologists. JACC Heart Fail. 2019;7(2):169–172. doi: 10.1016/j.jchf.2018.11.013.
  • Cosentino F, Grant PJ, Aboyans V, et al. 2019 ESC guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the EASD. Eur Heart J. 2020;41(2):255–323. doi: 10.1093/eurheartj/ehz486.
  • Davies MJ, Aroda VR, Collins BS, et al. Management of hyperglycemia in type 2 diabetes, 2022. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetes Care. 2022;45(11):2753–2786. doi: 10.2337/dci22-0034.
  • Buse JB, Wexler DJ, Tsapas A, 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;43(2):487–493. doi: 10.2337/dci19-0066.
  • Heidenreich PA, Bozkurt B, Aguilar D, et al. AHA/ACC/HFSA guideline for the management of heart failure: executive summary. J Am Coll Cardiol. 2022;79(17):1757–1780. doi: 10.1016/j.jacc.2021.12.011.
  • Vaduganathan M, Docherty KF, Claggett BL, et al. SGLT-2 inhibitors in patients with heart failure: a comprehensive meta-analysis of five randomised controlled trials. Lancet. 2022;400(10354):757–767. doi: 10.1016/S0140-6736(22)01429-5.
  • Baigent C, Emberson JR, Haynes R, et al. Impact of diabetes on the effects of sodium glucose co-transporter-2 inhibitors on kidney outcomes: collaborative meta-analysis of large placebo-controlled trials. Lancet. 2022;400(10365):1788–1801. doi: 10.1016/S0140-6736(22)02074-8.
  • Kang Y, Zhan F, He M, et al. Anti-inflammatory effects of sodium-glucose co-transporter 2 inhibitors on atherosclerosis. Vascul Pharmacol. 2020;133–134:106779. doi: 10.1016/j.vph.2020.106779.
  • Idzkowska E, Eljaszewicz A, Miklasz P, et al. The role of different monocyte subsets in the pathogenesis of atherosclerosis and acute coronary syndromes. Scand J Immunol. 2015;82(3):163–173. doi: 10.1111/sji.12314.
  • Baldrighi M, Mallat Z, Li X. NLRP3 inflammasome pathways in atherosclerosis. Atherosclerosis. 2017;267:127–138. doi: 10.1016/j.atherosclerosis.2017.10.027.
  • Han JH, Oh TJ, Lee G, et al. The beneficial effects of empagliflozin, an SGLT2 inhibitor, on atherosclerosis in ApoE −/− mice fed a Western diet. Diabetologia. 2017;60(2):364–376. doi: 10.1007/s00125-016-4158-2.
  • Nakatsu Y, Kokubo H, Bumdelger B, et al. The SGLT2 inhibitor luseogliflozin rapidly normalizes aortic mRNA levels of inflammation-related but not lipid-metabolism-related genes and suppresses atherosclerosis in diabetic ApoE KO mice. Int J Mol Sci. 2017;18(8):1704. doi: 10.3390/ijms18081704.
  • Nasiri-Ansari Ν, Dimitriadis GK, Agrogiannis G, et al. Canagliflozin attenuates the progression of atherosclerosis and inflammation process in APOE knockout mice. Cardiovasc Diabetol. 2018;17(1):106. doi: 10.1186/s12933-018-0749-1.
  • Kim SR, Lee S-G, Kim SH, et al. SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease. Nat Commun. 2020;11(1):2127. doi: 10.1038/s41467-020-15983-6.
  • Davignon J, Ganz P. Role of endothelial dysfunction in atherosclerosis. Circulation. 2004;109(23 Suppl. 1):III27–III32. doi: 10.1161/01.CIR.0000131515.03336.f8.
  • Zhang L, Gong D, Li S, et al. Meta-analysis of the effects of statin therapy on endothelial function in patients with diabetes mellitus. Atherosclerosis. 2012;223(1):78–85. doi: 10.1016/j.atherosclerosis.2012.01.031.
  • Barraclough JY, Patel S, Yu J, et al. The role of sodium glucose cotransporter-2 inhibitors in atherosclerotic cardiovascular disease: a narrative review of potential mechanisms. Cells. 2021;10(10):2699. doi: 10.3390/cells10102699.
  • Durante W, Behnammanesh G, Peyton KJ. Effects of sodium-glucose co-transporter 2 inhibitors on vascular cell function and arterial remodeling. Int J Mol Sci. 2021;22(16):8786. doi: 10.3390/ijms22168786.
  • Dhananjayan R, Koundinya KSS, Malati T, et al. Endothelial dysfunction in type 2 diabetes mellitus. Indian J Clin Biochem. 2016;31(4):372–379. doi: 10.1007/s12291-015-0516-y.
  • Gaspari T, Spizzo I, Liu H, et al. Dapagliflozin attenuates human vascular endothelial cell activation and induces vasorelaxation: a potential mechanism for inhibition of atherogenesis. Diab Vasc Dis Res. 2018;15(1):64–73. doi: 10.1177/1479164117733626.
  • El-Daly M, Pulakazhi Venu VK, Saifeddine M, et al. Hyperglycaemic impairment of PAR2-mediated vasodilation: prevention by inhibition of aortic endothelial sodium-glucose-co-transporter-2 and minimizing oxidative stress. Vascul Pharmacol. 2018;109:56–71. doi: 10.1016/j.vph.2018.06.006.
  • Adingupu DD, Göpel SO, Grönros J, et al. SGLT2 inhibition with empagliflozin improves coronary microvascular function and cardiac contractility in prediabetic ob/ob−/− mice. Cardiovasc Diabetol. 2019;18(1):16. doi: 10.1186/s12933-019-0820-6.
  • Sukhanov S, Higashi Y, Yoshida T, et al. The SGLT2 inhibitor empagliflozin attenuates interleukin-17A-induced human aortic smooth muscle cell proliferation and migration by targeting TRAF3IP2/ROS/NLRP3/caspase-1-dependent IL-1β and IL-18 secretion. Cell Signal. 2021;77:109825. doi: 10.1016/j.cellsig.2020.109825.
  • Li H, Shin SE, Seo MS, et al. The anti-diabetic drug dapagliflozin induces vasodilation via activation of PKG and Kv channels. Life Sci. 2018;197:46–55. doi: 10.1016/j.lfs.2018.01.032.
  • Badacz R, Przewłocki T, Legutko J, et al. microRNAs associated with carotid plaque development and vulnerability: the clinician’s perspective. Int J Mol Sci. 2022;23(24):15645. doi: 10.3390/ijms232415645.
  • Kabłak-Ziembicka A, Badacz R, Przewłocki T. Clinical application of serum microRNAs in atherosclerotic coronary artery disease. J Clin Med. 2022;11(22):6849. doi: 10.3390/jcm11226849.
  • Pignatelli P, Baratta F, Buzzetti R, et al. The sodium-glucose co-transporter-2 (SGLT2) inhibitors reduce platelet activation and thrombus formation by lowering NOX2-Related oxidative stress: a pilot study. Antioxidants. 2022;11(10):1878. doi: 10.3390/antiox11101878.
  • Shen Y, Cheng L, Xu M, et al. SGLT2 inhibitor empagliflozin downregulates miRNA-34a-5p and targets GREM2 to inactivate hepatic stellate cells and ameliorate non-alcoholic fatty liver disease-associated fibrosis. Metabolism. 2023;146:155657. doi: 10.1016/j.metabol.2023.155657.
  • Mone P, Lombardi A, Kansakar U, et al. Empagliflozin improves the microRNA signature of endothelial dysfunction in patients with heart failure with preserved ejection fraction and diabetes. J Pharmacol Exp Ther. 2023;384(1):116–122. doi: 10.1124/jpet.121.001251.
  • Hattori S. Anti-inflammatory effects of empagliflozin in patients with type 2 diabetes and insulin resistance. Diabetol Metab Syndr. 2018;10(1):93. doi: 10.1186/s13098-018-0395-5.
  • Storgaard H, Gluud LL, Bennett C, et al. Benefits and harms of sodium-glucose co-transporter 2 inhibitors in patients with type 2 diabetes: a systematic review and meta-analysis. PLOS One. 2016;11(11):e0166125. doi: 10.1371/journal.pone.0166125.
  • Wang Y, Xu L, Yuan L, et al. Sodium-glucose co-transporter-2 inhibitors suppress atrial natriuretic peptide secretion in patients with newly diagnosed type 2 diabetes. Diabet Med. 2016;33(12):1732–1736. doi: 10.1111/dme.13107.
  • Pahud de Mortanges A, Salvador DJr., Laimer M, et al. The role of SGLT2 inhibitors in atherosclerosis: a narrative mini-review. Front Pharmacol. 2021;12:751214. doi: 10.3389/fphar.2021.751214.
  • Sheahan KH, Wahlberg EA, Gilbert MP. An overview of GLP-1 agonists and recent cardiovascular outcomes trials. Postgrad Med J. 2020;96(1133):156–161. doi: 10.1136/postgradmedj-2019-137186.
  • Idris I, Donnelly R. Sodium-glucose co-transporter-2 inhibitors: an emerging new class of oral antidiabetic drug. Diabetes Obes Metab. 2009;11(2):79–88. doi: 10.1111/j.1463-1326.2008.00982.x.
  • Low Wang CC, Hess CN, Hiatt WR, et al. Clinical update: cardiovascular disease in diabetes mellitus. Circulation. 2016;133(24):2459–2502. doi: 10.1161/CIRCULATIONAHA.116.022194.
  • Tanaka A, Sata M, Okada Y, et al. Effect of ipragliflozin on carotid intima-media thickness in patients with type 2 diabetes: a multicenter, randomized, controlled trial. Eur Heart J Cardiovasc Pharmacother. 2023;9(2):165–172. doi: 10.1093/ehjcvp/pvac059.
  • Katakami N, Mita T, Maeda N, et al. Evaluation of the effect of tofogliflozin on the tissue characteristics of the carotid wall—a sub-analysis of the UTOPIA trial. Cardiovasc Diabetol. 2022;21(1):19. doi: 10.1186/s12933-022-01451-6.
  • Katakami N, Mita T, Yoshii H, et al. Tofogliflozin does not delay progression of carotid atherosclerosis in patients with type 2 diabetes: a prospective, randomized, open-label, parallel-group comparative study. Cardiovasc Diabetol. 2020;22(1):143. doi: 10.1186/s12933-020-01079-4.
  • Ardahanli İ. Empagliflozin may regresses carotid intima-media thickness and epicardial adipose tissue volume in patients with type 2 diabetes mellitus. Lokman Hekim Health Sci. 2021;1(3):74–80. doi: 10.14744/lhhs.2021.80001.
  • Nomiyama T, Shimono D, Horikawa T, et al. Efficacy and safety of sodium-glucose cotransporter 2 inhibitor ipragliflozin on glycemic control and cardiovascular parameters in Japanese patients with type 2 diabetes mellitus; Fukuoka Study of Ipragliflozin (FUSION). Endocr J. 2018;65(8):859–867. doi: 10.1507/endocrj.EJ18-0022.
  • Irace C, Casciaro F, Scavelli FB, et al. Empagliflozin influences blood viscosity and wall shear stress in subjects with type 2 diabetes mellitus compared with incretin-based therapy. Cardiovasc Diabetol. 2018;17(1):52. doi: 10.1186/s12933-018-0695-y.
  • Shigiyama F, Kumashiro N, Miyagi M, 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(1):84. doi: 10.1186/s12933-017-0564-0.
  • Kishimoto S, Higashi Y, Imai T, et al. Lack of impact of ipragliflozin on endothelial function in patients with type 2 diabetes: sub-analysis of the PROTECT study. Cardiovasc Diabetol. 2023;22(1):119. doi: 10.1186/s12933-023-01856-x.
  • Sposito AC, Breder I, Soares AAS, et al. Dapagliflozin effect on endothelial dysfunction in diabetic patients with atherosclerotic disease: a randomized active-controlled trial. Cardiovasc Diabetol. 2021;20(1):74. doi: 10.1186/s12933-021-01264-z.
  • Zainordin NA, Hatta SFWM, Mohamed Shah FZ, et al. Effects of dapagliflozin on endothelial dysfunction in type 2 diabetes with established ischemic heart disease (EDIFIED). J Endocr Soc. 2020;4(1):bvz017.
  • Sposito AC, Breder I, Barreto J, et al. Evolocumab on top of empagliflozin improves endothelial function of individuals with diabetes: randomized active-controlled trial. Cardiovasc Diabetol. 2022;21(1):147. doi: 10.1186/s12933-022-01584-8.
  • Tanaka A, Shimabukuro M, Machii N, et al. Effect of empagliflozin on endothelial function in patients with type 2 diabetes and cardiovascular disease: results from the multicenter, randomized, placebo-controlled, double-blind EMBLEM trial. Diabetes Care. 2019;42(10):e159–e161. doi: 10.2337/dc19-1177.
  • Wei R, Wang W, Pan Q, et al. Effects of SGLT-2 inhibitors on vascular endothelial function and arterial stiffness in subjects with type 2 diabetes: a systematic review and meta-analysis of randomized controlled trials. Front Endocrinol. 2022;13:826604. doi: 10.3389/fendo.2022.826604.
  • Wu Q-L, Zheng T, Li S-Z, et al. Effects of dapagliflozin in the progression of atherosclerosis in patients with type 2 diabetes: a meta-analysis of randomized controlled trials. Diabetol Metab Syndr. 2022;14(1):41. doi: 10.1186/s13098-022-00810-3.
  • Patoulias D, Papadopoulos C, Zografou I, et al. Effect of empagliflozin and dapagliflozin on ambulatory arterial stiffness in patients with type 2 diabetes mellitus and cardiovascular co-morbidities: a prospective, observational study. Medicina. 2022;58(9):1167. doi: 10.3390/medicina58091167.
  • Katakami N, Mita T, Yoshii H, et al. Effect of tofogliflozin on arterial stiffness in patients with type 2 diabetes: prespecified sub-analysis of the prospective, randomized, open-label, parallel-group comparative UTOPIA trial. Cardiovasc Diabetol. 2021;20(1):4. doi: 10.1186/s12933-020-01206-1.
  • Kourtidou C, Rafailidis V, Varouktsi G, et al. Effects of sodium-glucose co-transporter-2 inhibitors on markers of vascular damage. J Pers Med. 2023;13(3):536. doi: 10.3390/jpm13030536.
  • Karalliedde J, Fountoulakis N, Stathi D, et al. Does dapagliflozin influence arterial stiffness and levels of circulating anti-aging hormone soluble Klotho in people with type 2 diabetes and kidney disease? Results of a randomized parallel group clinical trial. Front Cardiovasc Med. 2022;9:992327. doi: 10.3389/fcvm.2022.992327.
  • Bosch A, Ott C, Jung S, et al. How does empagliflozin improve arterial stiffness in patients with type 2 diabetes mellitus? Sub analysis of a clinical trial. Cardiovasc Diabetol. 2019;18(1):44. doi: 10.1186/s12933-019-0839-8.
  • Papadopoulou E, Loutradis C, Tzatzagou G, et al. Dapagliflozin decreases ambulatory central blood pressure and pulse wave velocity in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled clinical trial. J Hypertens. 2021;39(4):749–758. doi: 10.1097/HJH.0000000000002690.
  • Hong J-Y, Park K-Y, Kim J-D, et al. Effects of 6 months of dapagliflozin treatment on metabolic profile and endothelial cell dysfunction for obese type 2 diabetes mellitus patients without atherosclerotic cardiovascular disease. J Obes Metab Syndr. 2020;29(3):215–221. doi: 10.7570/jomes20040.
  • Hidalgo Santiago JC, Maraver Delgado J, Cayón Blanco M, et al. Arterial en pacientes con diabetes mellitus tipo 2. Med Clin. 2020;154(5):171–174. doi: 10.1016/j.medcli.2019.05.028.
  • Solini A, Giannini L, Seghieri M, et al. Dapagliflozin acutely improves endothelial dysfunction, reduces aortic stiffness and renal resistive index in type 2 diabetic patients: a pilot study. Cardiovasc Diabetol. 2017;16(1):138. doi: 10.1186/s12933-017-0621-8.
  • Striepe K, Jumar A, Ott C, et al. Effects of the selective sodium-glucose cotransporter 2 inhibitor empagliflozin on vascular function and central hemodynamics in patients with type 2 diabetes mellitus. Circulation. 2017;136(12):1167–1169. doi: 10.1161/CIRCULATIONAHA.117.029529.
  • Bechlioulis A, Markozannes G, Chionidi I, et al. The effect of SGLT2 inhibitors, GLP1 agonists, and their sequential combination on cardiometabolic parameters: a randomized, prospective, intervention study. J Diabetes Complications. 2023;37(4):108436. doi: 10.1016/j.jdiacomp.2023.108436.
  • Patoulias D, Papadopoulos C, Kassimis G, et al. Effect of sodium-glucose co-transporter-2 inhibitors on arterial stiffness: a systematic review and meta-analysis of randomized controlled trials. Vasc Med. 2022;27(5):433–439. doi: 10.1177/1358863X221101653.
  • Sacks HS, Fain JN. Human epicardial adipose tissue: a review. Am Heart J. 2007;153(6):907–917. doi: 10.1016/j.ahj.2007.03.019.
  • Mahabadi AA, Lehmann N, Kälsch H, et al. Association of epicardial adipose tissue with progression of coronary artery calcification Is more pronounced in the early phase of atherosclerosis. JACC Cardiovasc Imaging. 2014;7(9):909–916. doi: 10.1016/j.jcmg.2014.07.002.
  • Rosito GA, Massaro JM, Hoffmann U, et al. Pericardial fat, visceral abdominal fat, cardiovascular disease risk factors, and vascular calcification in a community-based sample. Circulation. 2008;117(5):605–613. doi: 10.1161/CIRCULATIONAHA.107.743062.
  • Requena-Ibáñez JA, Santos-Gallego CG, Rodriguez-Cordero A, et al. Mechanistic insights of empagliflozin in nondiabetic patients with HFrEF. JACC Heart Fail. 2021;9(8):578–589. doi: 10.1016/j.jchf.2021.04.014.
  • 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. 2021;20(1):32. doi: 10.1186/s12933-021-01228-3.
  • Sato T, Aizawa Y, Yuasa S, et al. The effect of dapagliflozin treatment on epicardial adipose tissue volume. Cardiovasc Diabetol. 2018;17(1):6. doi: 10.1186/s12933-017-0658-8.
  • Yagi S, Hirata Y, Ise T, et al. Canagliflozin reduces epicardial fat in patients with type 2 diabetes mellitus. Diabetol Metab Syndr. 2017;9(1):78. doi: 10.1186/s13098-017-0275-4.
  • Bouchi R, Terashima M, Sasahara Y, et al. Luseogliflozin reduces epicardial fat accumulation in patients with type 2 diabetes: a pilot study. Cardiovasc Diabetol. 2017;16(1):32. doi: 10.1186/s12933-017-0516-8.
  • Fukuda T, Bouchi R, Terashima M, et al. Ipragliflozin reduces epicardial fat accumulation in non-obese type 2 diabetic patients with visceral obesity: a pilot study. Diabetes Ther. 2017;8(4):851–861. doi: 10.1007/s13300-017-0279-y.
  • Myasoedova VA, Parisi V, Moschetta D, et al. Efficacy of cardiometabolic drugs in reduction of epicardial adipose tissue: a systematic review and meta-analysis. Cardiovasc Diabetol. 2023;22(1):23. doi: 10.1186/s12933-023-01738-2.