Infarct size, inflammatory burden, and admission hyperglycemia in diabetic patients with acute myocardial infarction treated with SGLT2-inhibitors: a multicenter international registry

Pasquale Paolisso, Luca Bergamaschi, Gaetano Santulli, Emanuele Gallinoro, Arturo Cesaro, Felice Gragnano, Celestino Sardu, Niya Mileva, Alberto Foà, Matteo Armillotta, Angelo Sansonetti, Sara Amicone, Andrea Impellizzeri, Gianni Casella, Ciro Mauro, Dobrin Vassilev, Raffaele Marfella, Paolo Calabrò, Emanuele Barbato, Carmine Pizzi, Pasquale Paolisso, Luca Bergamaschi, Gaetano Santulli, Emanuele Gallinoro, Arturo Cesaro, Felice Gragnano, Celestino Sardu, Niya Mileva, Alberto Foà, Matteo Armillotta, Angelo Sansonetti, Sara Amicone, Andrea Impellizzeri, Gianni Casella, Ciro Mauro, Dobrin Vassilev, Raffaele Marfella, Paolo Calabrò, Emanuele Barbato, Carmine Pizzi

Abstract

Background: The inflammatory response occurring in acute myocardial infarction (AMI) has been proposed as a potential pharmacological target. Sodium-glucose co-transporter 2 inhibitors (SGLT2-I) currently receive intense clinical interest in patients with and without diabetes mellitus (DM) for their pleiotropic beneficial effects. We tested the hypothesis that SGLT2-I have anti-inflammatory effects along with glucose-lowering properties. Therefore, we investigated the link between stress hyperglycemia, inflammatory burden, and infarct size in a cohort of type 2 diabetic patients presenting with AMI treated with SGLT2-I versus other oral anti-diabetic (OAD) agents.

Methods: In this multicenter international observational registry, consecutive diabetic AMI patients undergoing percutaneous coronary intervention (PCI) between 2018 and 2021 were enrolled. Based on the presence of anti-diabetic therapy at the admission, patients were divided into those receiving SGLT2-I (SGLT-I users) versus other OAD agents (non-SGLT2-I users). The following inflammatory markers were evaluated at different time points: white-blood-cell count, neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), neutrophil-to-platelet ratio (NPR), and C-reactive protein. Infarct size was assessed by echocardiography and by peak troponin levels.

Results: The study population consisted of 583 AMI patients (with or without ST-segment elevation): 98 SGLT2-I users and 485 non-SGLT-I users. Hyperglycemia at admission was less prevalent in the SGLT2-I group. Smaller infarct size was observed in patients treated with SGLT2-I compared to non-SGLT2-I group. On admission and at 24 h, inflammatory indices were significantly higher in non-SGLT2-I users compared to SGLT2-I patients, with a significant increase in neutrophil levels at 24 h. At multivariable analysis, the use of SGLT2-I was a significant predictor of reduced inflammatory response (OR 0.457, 95% CI 0.275-0.758, p = 0.002), independently of age, admission creatinine values, and admission glycemia. Conversely, peak troponin values and NSTEMI occurrence were independent predictors of a higher inflammatory status.

Conclusions: Type 2 diabetic AMI patients receiving SGLT2-I exhibited significantly reduced inflammatory response and smaller infarct size compared to those receiving other OAD agents, independently of glucose-metabolic control. Our findings are hypothesis generating and provide new insights on the cardioprotective effects of SGLT2-I in the setting of coronary artery disease.

Trial registration: Data are part of the ongoing observational registry: SGLT2-I AMI PROTECT.

Clinicaltrials: gov Identifier: NCT05261867.

Keywords: Acute myocardial infarction; Hyperglycemia; Infarct size; Inflammation; SGLT2-I.

Conflict of interest statement

The authors declare that they have no competing interests.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Box plot and Dot plot comparing the distribution of White Blood Cells (A and C) and Neutrophil-Lymphocyte Ratio (NLR) (B and D) at admission and discharge in SGLT2-I users vs. non-SGLT2-I users (blue box and red box respectively)
Fig. 2
Fig. 2
Density Plot showing the neutrophils (A and C) and lymphocytes (B and D) distribution at admission and after 24 h in SGLT2-I versus non-SGLT2-I users. Blue curve denotes non-SGLT2-I users; red curve represents patients receiving SGLT2-I. The dotted arrow shows how the peak of the neutrophil’s distribution in non SGLT2-I users moved towards higher values after 24 h
Fig. 3
Fig. 3
Correlations between neutrophils values measured at 24 h and the admission glucose levels (A) and peak troponin values (B), in the overall study population (n = 583)
Fig. 4
Fig. 4
Correlation between neutrophils at 24 h and discharge LVEF in SGLT2-I versus non-SGLT2-I users (red and blue lines respectively). An inverse correlation was found between the inflammatory burden (represented by neutrophils at 24 h) and the LVEF at discharge. Conversely, in SGLT2-I users, the higher inflammatory burden was not accompanied by a reduction in LVEF at discharge

References

    1. Paolisso P, Foà A, Bergamaschi L, et al. Hyperglycemia, inflammatory response and infarct size in obstructive acute myocardial infarction and MINOCA. Cardiovasc Diabetol. 2021;20:33. doi: 10.1186/s12933-021-01222-9.
    1. Olsen MB, Gregersen I, Sandanger Ø, et al. Targeting the Inflammasome in Cardiovascular Disease. JACC Basic Transl Sci. 2022;7:84–98. doi: 10.1016/j.jacbts.2021.08.006.
    1. Algoet M, Janssens S, Himmelreich U, et al. ‘Myocardial ischemia-reperfusion injury and the influence of inflammation. Trends Cardiovasc Med. 2022 doi: 10.1016/j.tcm.2022.02.005.
    1. Rock KL, Kono H. The inflammatory response to cell death. Annu Rev Pathol. 2008;3:99–126. doi: 10.1146/annurev.pathmechdis.3.121806.151456.
    1. Huang S, Frangogiannis NG. Anti-inflammatory therapies in myocardial infarction: failures, hopes and challenges. Br J Pharmacol. 2018;175:1377–1400. doi: 10.1111/bph.14155.
    1. Chen C, Cong BL, Wang M, et al. Neutrophil to lymphocyte ratio as a predictor of myocardial damage and cardiac dysfunction in acute coronary syndrome patients. Integr Med Res. 2018;7:192–199. doi: 10.1016/j.imr.2018.02.006.
    1. Somaschini A, Cornara S, Demarchi A, et al. Neutrophil to platelet ratio: A novel prognostic biomarker in ST-elevation myocardial infarction patients undergoing primary percutaneous coronary intervention. Eur J Prev Cardiol. 2020;27:2338–2340. doi: 10.1177/2047487319894103.
    1. Li W, Liu Q, Tang Y. Platelet to lymphocyte ratio in the prediction of adverse outcomes after acute coronary syndrome: a meta-analysis. Sci Rep. 2017;7:40426. doi: 10.1038/srep40426.
    1. Shetelig C, Limalanathan S, Hoffmann P, et al. Association of IL-8 With Infarct Size and Clinical Outcomes in Patients With STEMI. J Am Coll Cardiol. 2018;72:187–198. doi: 10.1016/j.jacc.2018.04.053.
    1. Tøllefsen IM, Shetelig C, Seljeflot I, Eritsland J, Hoffmann P, Andersen G. High levels of interleukin-6 are associated with final infarct size and adverse clinical events in patients with STEMI. Open Heart. 2021;8:e001869. doi: 10.1136/openhrt-2021-001869.
    1. Anker SD, Butler J, Filippatos G, et al. Empagliflozin in Heart Failure with a Preserved Ejection Fraction. N Engl J Med. 2021;385:1451–1461. doi: 10.1056/NEJMoa2107038.
    1. Zinman B, Wanner C, Lachin JM, et al. Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes. N Engl J Med. 2015;373:2117–28. doi: 10.1056/NEJMoa1504720.
    1. McMurray JJV, Solomon SD, Inzucchi SE, et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N Engl J Med. 2019;381:1995–2008. doi: 10.1056/NEJMoa1911303.
    1. Andreadou I, Bell RM, Bøtker HE, Zuurbier CJ. SGLT2 inhibitors reduce infarct size in reperfused ischemic heart and improve cardiac function during ischemic episodes in preclinical models. Biochim Biophys Acta Mol Basis Dis. 2020;1866:165770. doi: 10.1016/j.bbadis.2020.165770.
    1. Salvatore T, Caturano A, Galiero R, et al. Cardiovascular Benefits from Gliflozins: Effects on Endothelial Function. Biomedicines. 2021;9:1356. doi: 10.3390/biomedicines9101356.
    1. Marfella R, D’Onofrio N, Trotta MC, et al. Sodium/glucose cotransporter 2 (SGLT2) inhibitors improve cardiac function by reducing JunD expression in human diabetic hearts. Metabolism. 2022;127:154936. doi: 10.1016/j.metabol.2021.154936.
    1. Collet JP, Thiele H, Barbato E, et al. 2020 ESC Guidelines for the management of acute coronary syndromes in patients presenting without persistent ST-segment elevation. Eur Heart J. 2021;42:1289–1367. doi: 10.1093/eurheartj/ehaa575.
    1. Ibanez B, James S, Agewall S, et al. 2017 ESC Guidelines for the management of acute myocardial infarction in patients presenting with ST-segment elevation: The Task Force for the management of acute myocardial infarction in patients presenting with ST-segment elevation of the European Society of Cardiology (ESC) Eur Heart J. 2018;39:119–177. doi: 10.1093/eurheartj/ehx393.
    1. Lang RM, Badano LP, Mor-Avi V, et al. Recommendations for cardiac chamber quantification by echocardiography in adults: an update from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. Eur Heart J Cardiovasc Imaging. 2015;16:233–70. doi: 10.1093/ehjci/jev014.
    1. 2 Classification and Diagnosis of Diabetes: Standards of Medical Care in Diabetes-2019. Diabetes Care. 2019;42:S13-s28.
    1. Varzideh F, Kansakar U, Santulli G. SGLT2 inhibitors in cardiovascular medicine. Eur Heart J Cardiovasc Pharmacother. 2021;7:e67-e68. doi: 10.1093/ehjcvp/pvab039.
    1. Lopaschuk GD, Verma S. Mechanisms of Cardiovascular Benefits of Sodium Glucose Co-Transporter 2 (SGLT2) Inhibitors: A State-of-the-Art Review. JACC Basic Transl Sci. 2020;5:632–644. doi: 10.1016/j.jacbts.2020.02.004.
    1. Cowie MR, Fisher M. SGLT2 inhibitors: mechanisms of cardiovascular benefit beyond glycaemic control. Nat Rev Cardiol. 2020;17:761–772. doi: 10.1038/s41569-020-0406-8.
    1. Palmiero G, Cesaro A, Vetrano E, et al. Impact of SGLT2 Inhibitors on Heart Failure: From Pathophysiology to Clinical Effects. Int J Mol Sci. 2021;22:5863. doi: 10.3390/ijms22115863.
    1. D’Onofrio N, Sardu C, Trotta MC, et al. Sodium-glucose co-transporter2 expression and inflammatory activity in diabetic atherosclerotic plaques: Effects of sodium-glucose co-transporter2 inhibitor treatment. Mol Metab. 2021;54:101337. doi: 10.1016/j.molmet.2021.101337.
    1. Buse JB, Wexler DJ, Tsapas A, et al. 2019 update to: Management of hyperglycaemia in type 2 diabetes, 2018. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD) Diabetologia. 2020;63:221–228. doi: 10.1007/s00125-019-05039-w.
    1. Grubić Rotkvić P, Cigrovski Berković M, Bulj N, Rotkvić L. Minireview: are SGLT2 inhibitors heart savers in diabetes? Heart Fail Rev. 2020;25:899–905. doi: 10.1007/s10741-019-09849-3.
    1. Marfella R, Siniscalchi M, Esposito K, et al. Effects of stress hyperglycemia on acute myocardial infarction: role of inflammatory immune process in functional cardiac outcome. Diabetes Care. 2003;26:3129–35. doi: 10.2337/diacare.26.11.3129.
    1. Esper RJ, Nordaby RA. Cardiovascular events, diabetes and guidelines: the virtue of simplicity. Cardiovasc Diabetol. 2019;18:42. doi: 10.1186/s12933-019-0844-y.
    1. Sattar N. Revisiting the links between glycaemia, diabetes and cardiovascular disease. Diabetologia. 2013;56:686–95. doi: 10.1007/s00125-012-2817-5.
    1. Cannon CP, Perkovic V, Agarwal R, et al. Evaluating the effects of canagliflozin on cardiovascular and renal events in patients with type 2 diabetes mellitus and chronic kidney disease according to baseline HbA1c, including those with HbA1c < 7%: results from the CREDENCE trial. Circulation. 2020;141:407–410. doi: 10.1161/CIRCULATIONAHA.119.044359.
    1. Connelly KA, Zhang Y, Visram A, et al. Empagliflozin improves diastolic function in a nondiabetic rodent model of heart failure with preserved ejection fraction. JACC Basic Transl Sci. 2019;4:27–37. doi: 10.1016/j.jacbts.2018.11.010.
    1. Paolisso P, Foà A, Bergamaschi L, et al. Impact of admission hyperglycemia on short and long-term prognosis in acute myocardial infarction: MINOCA versus MIOCA. Cardiovasc Diabetol. 2021;20:192. doi: 10.1186/s12933-021-01384-6.
    1. Iannantuoni F, de Marañon A M, Diaz-Morales N, et al. The SGLT2 Inhibitor empagliflozin ameliorates the inflammatory profile in type 2 diabetic patients and promotes an antioxidant response in Leukocytes. J Clin Med. 2019;8:1814. doi: 10.3390/jcm8111814.
    1. Heerspink HJL, Perco P, Mulder S, et al. Canagliflozin reduces inflammation and fibrosis biomarkers: a potential mechanism of action for beneficial effects of SGLT2 inhibitors in diabetic kidney disease. Diabetologia. 2019;62:1154–1166. doi: 10.1007/s00125-019-4859-4.
    1. Leng W, Wu M, Pan H, et al. The SGLT2 inhibitor dapagliflozin attenuates the activity of ROS-NLRP3 inflammasome axis in steatohepatitis with diabetes mellitus. Ann Transl Med. 2019;7:429. doi: 10.21037/atm.2019.09.03.
    1. Sardu C, Massetti M, Testa N, et al. Effects of Sodium-Glucose Transporter 2 Inhibitors (SGLT2-I) in patients with ischemic heart disease (IHD) treated by coronary artery bypass grafting via MiECC: inflammatory burden, and clinical outcomes at 5 years of follow-up. Front Pharmacol. 2021;12:777083. doi: 10.3389/fphar.2021.777083.
    1. Ye Y, Jia X, Bajaj M, Birnbaum Y. Dapagliflozin Attenuates Na(+)/H(+) Exchanger-1 in Cardiofibroblasts via AMPK Activation. Cardiovasc Drugs Ther. 2018;32:553–558. doi: 10.1007/s10557-018-6837-3.
    1. Byrne NJ, Matsumura N, Maayah ZH, et al. Empagliflozin blunts worsening cardiac dysfunction associated with reduced NLRP3 (Nucleotide-Binding Domain-Like Receptor Protein 3) inflammasome activation in heart failure. Circ Heart Fail. 2020;13:e006277. doi: 10.1161/CIRCHEARTFAILURE.119.006277.
    1. Yu YW, Que JQ, Liu S, et al. Sodium-Glucose Co-transporter-2 inhibitor of dapagliflozin attenuates myocardial ischemia/reperfusion injury by limiting NLRP3 inflammasome activation and modulating autophagy. Front Cardiovasc Med. 2021;8:768214. doi: 10.3389/fcvm.2021.768214.
    1. Youm YH, Nguyen KY, Grant RW, et al. The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. 2015;21:263–9. doi: 10.1038/nm.3804.
    1. Zelniker TA, Braunwald E. Mechanisms of Cardiorenal Effects of Sodium-Glucose Cotransporter 2 Inhibitors: JACC State-of-the-Art Review. J Am Coll Cardiol. 2020;75:422–434. doi: 10.1016/j.jacc.2019.11.031.
    1. Kim SR, Lee SG, Kim SH, et al. SGLT2 inhibition modulates NLRP3 inflammasome activity via ketones and insulin in diabetes with cardiovascular disease. Nat Commun. 2020;11:2127. doi: 10.1038/s41467-020-15983-6.
    1. De Rosa M, Gambardella J, Shu J, Santulli G. Dietary fat is a key determinant in balancing mitochondrial dynamics in heart failure: a novel mechanism underlying the obesity paradox. Cardiovasc Res. 2018;114:925–927. doi: 10.1093/cvr/cvy074.
    1. Glatz JFC, Nabben M, Young ME, Schulze PC, Taegtmeyer H, Luiken J. Re-balancing cellular energy substrate metabolism to mend the failing heart. Biochim Biophys Acta Mol Basis Dis. 2020;1866:165579. doi: 10.1016/j.bbadis.2019.165579.
    1. Jankauskas SS, Kansakar U, Varzideh F, et al. Heart failure in diabetes. Metabolism. 2021;125:154910. doi: 10.1016/j.metabol.2021.154910.
    1. Sowton AP, Griffin JL, Murray AJ. Metabolic Profiling of the Diabetic Heart: Toward a Richer Picture. Front Physiol. 2019;10:639. doi: 10.3389/fphys.2019.00639.
    1. Ferrannini E, Baldi S, Frascerra S, 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. doi: 10.2337/db15-1356.
    1. Abdurrachim D, Manders E, Nicolay K, Mayoux E, Prompers JJ. Single dose of empagliflozin increases in vivo cardiac energy status in diabetic db/db mice. Cardiovasc Res. 2018;114:1843–1844.
    1. Shimizu W, Kubota Y, Hoshika Y, et al. Effects of empagliflozin versus placebo on cardiac sympathetic activity in acute myocardial infarction patients with type 2 diabetes mellitus: the EMBODY trial. Cardiovasc Diabetol. 2020;19:148. doi: 10.1186/s12933-020-01127-z.
    1. Rossello X, Bueno H, Pocock SJ, et al. Predictors of all-cause mortality and ischemic events within and beyond 1 year after an acute coronary syndrome: Results from the EPICOR registry. Clin Cardiol. 2019;42:111–119. doi: 10.1002/clc.23116.
    1. Pocock SJ, Huo Y, Van de Werf F, et al. Predicting two-year mortality from discharge after acute coronary syndrome: An internationally-based risk score. Eur Heart J Acute Cardiovasc Care. 2019;8:727–737. doi: 10.1177/2048872617719638.

Source: PubMed

3
Subscribe