Skip to main content

Use of EMPAgliflozin in the prevention of CARDiotoxicity: the EMPACARD – PILOT trial

Abstract

Background

Anthracycline-based chemotherapy represents a cornerstone treatment for a number of common cancers, including breast cancer, lymphoma, and sarcoma. However, anthracycline-induced cardiotoxicity remains a significant concern, often presenting as a decline in cardiac function which can ultimately lead to heart failure (HF) or asymptomatic left ventricular dysfunction, in up to 10–15% of patients.Sodium-glucose transport protein 2 inhibitor (SGLT2i) therapies have been demonstrated to reduce the incidence of HF in high-risk non-cancer patients. Preliminary retrospective data suggest their role in mitigating the incidence of HF during or after anthracycline treatment

Methods

The EMPACARD-PILOT trial was a prospective case‒control study involving breast cancer patients scheduled to undergo anthracycline-based chemotherapy in a 4-cycle protocol of 60 mg/m2 doxorubicin. We used the HFA/ICOS risk score to identify patients at high or very high risk of cardiotoxicity. Patients with diabetes mellitus or stable heart failure with preserved ejection fraction (HFpEF) were prescribed empagliflozin (10 mg per day), starting seven days before the administration of anthracyclines and continuing for a period of six months. Those not meeting these criteria served as controls. The primary endpoint was cancer therapy-related cardiac dysfunction (CTRCD) incidence. CTRCD was defined as either a decrease in left ventricular ejection fraction (LVEF) of at least 10% to a final value below 50% or a reduction in global longitudinal strain (GLS) of at least 15% from baseline at any point during the study. The secondary endpoints included mortality and hospitalization due to cardiovascular causes or clinical heart failure. Exploratory endpoints included increases in serum troponin and NT-proBNP levels and a decrease in the glomerular filtration rate (GFR). The safety endpoints tracked includedketoacidosis, hypoglycemia, sepsis, neutropenic fever, and urinary tract infections.

Results

During the enrollment period, 785 breast cancer patients were analysed. Of these, 107 met the inclusion criteria, and 76 subsequently provided informed consent. The study was conducted with comparable adherence rates of 81.5% in both the empagliflozin group (n = 38) and the control group (n = 38). The follow-up data from 62 patients revealed a significant reduction in the primary outcome within 6 months for the empagliflozin group compared with the control group (6.5% vs. 35.5%, p = 0.005), with a relative risk of 0.18 (95% CI: 0.04–0.75). Compared with the control treatment, treatment with empagliflozin also significantly preserved the ejection fraction at 6 months follow-up (56.8% ± 5.8% vs. 53.7% ± 6.7, p = 0.029). However, there were no significant differences between the groups in terms of NT-proBNP, cTnI, clinical heart failure, GFR, or mortality/hospitalization due to heart failure.

Conclusion

Empagliflozin is associated with reduced incidence of CTRCD in high-risk patients treated with anthracyclines. These data should serve as the foundation for a clinical trial to test whether SGLT2 inhibitors can reduce the incidence of heart failure in this patient group.

Background

Anthracycline-based chemotherapy is the standard treatment for several common cancer types, such as breast cancer, lymphoma, and sarcoma [1]. However, the use of anthracyclines is associated with the potential for the development of cardiac toxicity [2, 3]. Anthracyclines can cause cardiac toxicity, which often results in a reduction in cardiac function and can lead to heart failure (HF). HF incidence has been reported in up to 10–15% of these patients [4]. Currently, there are no established therapies to attenuate or prevent the development of anthracycline-induced cardiotoxicity (AIC) and reduce the risk of HF. Sodium‒glucose transport protein 2 inhibitor (SGLT2i) therapies can reduce the incidence of HF in high-risk patients who have received anthracyclines [5, 6]. Strong scientific plausibility and preliminary data suggest that SGLT2i may also reduce the incidence of HF in those with AIC.

Anthracyclines are known to increase cytokines, reduce cardiomyocyte viability, and lead to cardiomyocyte apoptosis and myocardial fibrosis, all of which contribute to their cardiotoxicity [7]. Myocardial fibrosis plays a crucial role in the progression from cardiotoxicity to clinical heart failure [7]. In patients not treated with anthracyclines, the protective benefits of SGLT2i are postulated to include a reduction in pathologic cytokines and improved cell viability, leading to decreased cardiomyocyte apoptosis and myocardial fibrosis. In animal models of anthracycline cardiotoxicity, SGLT2i prevented the increase in cytokines, as well as the reduction in cardiomyocyte viability, the increase in cardiomyocyte apoptosis, and myocardial fibrosis [8, 9].

Retrospective data support our hypothesis that SGLT2i may improve cardiac outcomes when used with anthracyclines. Several retrospective studies have shown that SGLT2i reduces heart failure hospitalizations [10] and the composite primary cardiac outcome of cardiac events, including heart failure incidence, heart failure admissions, new cardiomyopathy (defined as a > 10% decline in the ejection fraction to < 53%), and clinically significant arrhythmias [11].

Therefore, we conducted a prospective case‒control study to examine the impact of SGLT2 inhibitors on cardiac dysfunction in patients with diabetes mellitus receiving anthracycline treatment.

Methods

The EMPACARD–PILOT trial was a prospective case‒control study. We included breast cancer patients scheduled to undergo a 4-cycle protocol of doxorubicin at 60 mg/m2. The study specifically targeted a population at high or very high risk of cardiac toxicity, as determined by the HFA/ICOS risk score [12]. Patients were excluded if they had a left ventricular ejection fraction (LVEF) < 50% at baseline; prior HF hospitalization; or myocardial infarction, coronary revascularization, stroke, or transient ischaemic attack within 90 days of the start of the study. Additional exclusion criteria included previous use of SGLT2i, systolic BP ≥ 180 mmHg or diastolic BP > 100 mmHg, severe kidney dysfunction with an estimated GFR of < 20 ml/min/m2, a history of ketoacidosis, a known allergy to SGLT2i, or pregnancy. All enrolled patients provided signed informed consent.

Study intervention

After screening and providing consent, patients with diabetes mellitus or stable HFpEF were assigned to the Empagliflozin arm (10 mg per day), whereas the other patients were placed in the control group. The study drug was started 7 days before the anthracyclines were taken and continued for 6 months. Patients were evaluated at baseline, before the second and fourth cycles of chemotherapy, and 3 months post chemotherapy (6-month follow-up). Each study visit included a review of symptoms, serum biomarkers, an echocardiogram to assess LVEF and GLS, and the recording of study-related adverse events. Patients who failed to attend evaluations or did not adhere to daily medication were withdrawn from the study.

Study endpoints

The primary study endpoint, cancer therapy-related cardiac dysfunction (CTRCD), was defined as a decrease in LVEF of at least 10% from baseline to a final LVEF of less than 50% and/or a relative decrease of at least 15% in global longitudinal strain (GLS) from baseline at any time in the study. The secondary endpoint was a clinical endpoint and was a composite of death or hospitalization due to CV causes and/or clinical heart failure. Incident heart failure was defined by the European Society of Cardiology [ESC] [13]. Exploratory endpoints included an increase in serum troponin, an increase in NT-proBNP, and a decrease in the GFR. The safety endpoints were the occurrence of ketoacidosis, hypoglycemia, sepsis, neutropenic fever, or urinary tract infection.

Statistical methods

Categorical variables are presented as absolute and relative frequencies and were compared with the chi-square test or Fisher’s exact test, as appropriate. Continuous variables are presented as the means with standard deviations and were compared with Welch’s t test. Statistical significance was set at p < 0.05. All calculations were performed with R version 4.3.0. We determined that the sample size would be 100 patients in each arm.

Results

A total of 785 patients with breast cancer were analysed, with 107 meeting the inclusion criteria. Of these, 76 women consented to participate and signed the informed consent form. Thirty-eight patients were assigned to the Empagliflozin arm, and 38 were assigned to the Control arm (Fig. 1). The adherence rate was 81.5% in both arms. There were no significant differences in baseline characteristics between adherent and nonadherent patients (Supplementary Table S1). All study participants were women treated for breast cancer, with a high prevalence of cardiovascular risk factors. Most participants were at stage III, and 23.68% had previously been treated with doxorubicin, with an average mean doxorubicin dose of 310,26 mg/m2 (Table 1). A follow-up was available for 62 patients.

Fig. 1
figure 1

Screening and follow-up

Table 1 Baseline characteristics

Primary outcome

Within 6 months, the primary outcome occurred in 35.5% of the participants in the control group and in 6.5% of those in the empagliflozin group (p = 0.005, Figs. 2). Compared with the control group, the empagliflozin group presented significant preservation of LVEF (56.8% ± 5.8 versus 53.7% ± 6.7, p = 0.029) Figs 3. Compared with the control group, the empagliflozin group had a relative risk of 0.18 for the primary outcome, with a 95% CI of 0.04 to 0.75, indicating a reduced risk of decline in LVEF. Additionally, the empagliflozin group exhibited significant preservation of GLS compared with the control group (5.3% vs. 32%, p = 0.003). The only significant difference between the placebo and treatment groups in terms of basal characteristics was the use of beta-blockers (58% in the Empagliflozin group and 34% in the placebo group, p = 0.038). When adjusting for beta-blocker use, the protective effect of empagliflozin remained significant, with an OR for the primary endpoint for the placebo group of 10.29 ± 2.48 (p = 0.0055).

Fig. 2
figure 2

Primary endpoint results

Fig. 3
figure 3

Left ventricle ejection fraction over time

Secondary outcomes

There was no significant difference between the two groups in terms of clinical heart failure, death, or hospitalization due to heart failure (Table 2).

Exploratory endpoints

NT-proBNP elevation, cTnI elevation, and GFR decrease were not significantly different between the two groups (Table 2).

Table 2 Outcomes according to treatment arm

Safety outcomes

There was a low overall incidence of adverse effects, with no significant difference between the groups, as shown in Table 3.

Table 3 Adverse effects

Discussion

Heart failure and cancer are intricately linked pathologies. Research has demonstrated that the presence of heart failure in patients is a risk factor for developing cancer [14]. On the other hand, advances in cancer therapies have been associated with the emergence of several cancer-related cardiovascular diseases [15], including asymptomatic left ventricular dysfunction and heart failure.

With the advent of cardio-oncology at the beginning of this century, we focused on understanding the pathophysiology of cardiotoxicities to establish, first, early therapies for them and, above all, preventive measures to avoid them and thus ensure that patients can continue their cancer treatment (Figs 2 and 3).

In recent years, several studies have evaluated the usefulness of different drugs for preventing heart failure in patients with CRCTD [16,17,18,19,20].

Over the years, we have witnessed multiple primary prevention strategies in cardio-oncology, with conflicting results. Cardioprotection started with the use of enalapril to prevent LVEF decline in patients who developed an increase in cardiac troponin during anthracycline treatment [21].

Subsequent trials, including OVERCOME, MANTICORE, PRADA, and the study by Guglin et al., evaluated the efficacy of beta-blockers, ACEIs, and angiotensin receptor blocker 2 (ARB2) in preventing left ventricular dysfunction in cancer patients [16, 17, 22, 23]. Research has also explored the cardioprotective use of aldosterone antagonists [18, 19, 24], and various studies have investigated whether statins can prevent the occurrence of cancer therapy-related cardiac dysfunction (CTRCD) [20, 25,26,27,28].

However, the results from these studies have been mixed, possibly due to flaws in study design, endpoint definitions, and the inclusion of patients with varying levels of cardio-oncological risk, predominantly low risk.

A few years ago, a new family of drugs originally developed for the treatment of diabetes, SGLT2 inhibitors (SGLT2is), was found to be beneficial for treating heart failure. Several prospective randomized, double-blind studies have demonstrated that SGLT2i could reduce mortality or hospitalization due to heart failure and improve renal function [5, 6, 29]. Notably, the EMPEROR-Reduced and EMPEROR-Preserved trials led to the approval of empagliflozin for treating heart failure with reduced ejection fraction (HFrEF) and heart failure with preserved ejection fraction (HFpEF) [5, 6].

In cardio-oncology, retrospective studies analysing national health databases (Oh CM et al., Gongora et al. and Abdel-Qadir et al.), which include patients with cancer and diabetes receiving anthracycline therapy, have been published [7, 10, 11]. These studies revealed that patients receiving SGLT2i had lower incidences of heart failure, heart failure hospitalizations, new cardiomyopathies, and arrhythmias [7, 10, 11].

The EMPACARD Prevention PILOT trial is a prospective open-label case‒control trial designed to evaluate the usefulness of empagliflozin in preventing CTRCD.

At the 6-month follow-up, patients treated with empagliflozin exhibited a smaller decline in global longitudinal strain (GLS) and left ventricular ejection fraction (LVEF). The only significant difference between the placebo and treatment groups in terms of basal characteristics was the use of beta-blockers (58% in the Empagliflozin group and 34% in the placebo group, p = 0.038). When adjusting for beta-blocker use, the protective effect of empagliflozin remained significant, with an OR for the primary endpoint for the placebo group of 10.29 ± 2.48 (p = 0.0055).

However, there was no statistically significant difference in the secondary endpoints. This absence of a significant difference is likely attributable to the small sample size.

Similarly, exploratory endpoints were not significantly different between the two groups. In terms of safety outcomes, there were no significant differences in the incidence of ketoacidosis, hypoglycemia, sepsis, neutropenic fever, or urinary tract infections between the groups.

The EMPACARD Prevention PILOT trial represents a promising start in investigating the role of empagliflozin as a cardioprotective strategy in patients treated with anthracyclines.

Limitations

The EMPACARD Prevention PILOT series is limited by its small sample size and relatively short follow-up period.

Future directions

Moving forward, it is imperative to conduct a prospective randomized, multicentre study to validate the findings elucidated in our trial. This approach will allow for a more comprehensive evaluation of empagliflozin’s efficacy and safety in reducing anthracycline-induced cardiotoxicity across a broader and more diverse patient population.

Conclusion

In this prospective case‒control trial, empagliflozin administered to a high-risk group were associated with a reduction in anthracycline-induced cardiac dysfunction. Furthermore, SGLT2 inhibitors have been proven safe for patients undergoing chemotherapy. These promising findings underscore the need for a clinical trial to explore whether SGLT2 inhibitors can effectively reduce the incidence of heart failure among high-risk patients receiving anthracycline treatment.

Data availability

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

AIC:

Anthracycline-induced cardiotoxicity

HF:

Heart failure

SGLT2i:

Sodium‒glucose transport protein 2 inhibitors

HFA/ICOS:

Heart failure association/international cardiOncology society

CTRCD:

Cancer therapeutic-related cardiac dysfunction

LVEF:

Left ventricular ejection fraction

GLS:

Global longitudinal strain

HfpEF:

Heart failure preserved ejection fraction

CKD:

Chronic kidney disease

GFR:

Glomerular filtration rate

CV:

Cardiovascular

References

  1. Hortobágyi GN. Anthracyclines in the treatment of Cancer. Drugs. 1997;54(Suppl 4):1–7. https://doi.org/10.2165/00003495-199700544-00003.

    Article  PubMed  Google Scholar 

  2. Raj S, Franco VI, Lipshultz SE, Anthracycline-Induced Cardiotoxicity. A review of pathophysiology, diagnosis, and treatment. Curr Treat Options Cardio Med. 2014;16:315. https://doi.org/10.1007/s11936-014-0315-4.

    Article  Google Scholar 

  3. Cardinale Daniela I, Fabiani. Cipolla Carlo Maria. Cardiotoxicity of Anthracyclines. Frontiers in Cardiovascular Medicine. Vol. 7. 2020 DOI = .https://doi.org/10.3389/fcvm.2020.00026.

  4. Larsen CM, Garcia Arango M, Dasari H et al. Association of Anthracycline With Heart Failure in Patients Treated for Breast Cancer or Lymphoma, 1985–2010. JAMA Netw Open. 2023;6(2):e2254669.https://doi.org/10.1001/jamanetworkopen.2022.54669.

  5. Milton Packer, M.D., Stefan D. Anker, M.D., Ph.D el al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N Engl J Med 2020; 383:1413-1424 .https://doi.org/10.1056/NEJMoa2022190.

  6. Stefan D. Anker, M.D., Ph.D., Javed Butler, M.D. Empagliflozin in Heart Failure with a Preserved Ejection Fraction. N Engl J Med 2021; 385:1451-1461 .https://doi.org/10.1056/NEJMoa2107038.

  7. Oh CM, Cho S, Jang JY, Kim H, Chun S, Choi M, Park S, Ko YG. Cardioprotective potential of an SGLT2 inhibitor against Doxorubicin-Induced Heart failure. Korean Circ J. 2019;49(12):1183–95. https://doi.org/10.4070/kcj.2019.0180. Epub 2019 Jul 31. PMID: 31456369; PMCID: PMC6875592.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Packer M, Butler J, Filippatos GS, Jamal W, Salsali A, Schnee J, Kimura K, Zeller C, George J, Brueckmann M, Anker SD, Zannad F and. Evaluation of the effect of sodium–glucose cotransporter 2 inhibition with empagliflozin on morbidity and mortality of patients with chronic heart failure and a reduced ejection fraction: rationale for and design of the EMPEROR-Reduced trial. Eur J Heart Fail. 2019;21:1270–8. https://doi.org/10.1002/ejhf.1536.

    Article  CAS  PubMed  Google Scholar 

  9. Tentolouris A, Vlachakis P, Tzeravini E, Eleftheriadou I, Tentolouris N. SGLT2 inhibitors: a review of their antidiabetic and cardioprotective effects. Int J Environ Res Public Health. 2019;16(16):2965. https://doi.org/10.3390/ijerph16162965. PMID: 31426529; PMCID: PMC6720282.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Abdel-Qadir H, Carrasco R, Austin P et al. The Association of Sodium-Glucose Cotransporter 2 inhibitors with Cardiovascular outcomes in Anthracycline-treated patients with Cancer. J Am Coll Cardiol CardioOnc 2023 Jun, 5 (3) 318–28https://doi.org/10.1016/j.jaccao.2023.03.011

  11. Gongora C, Drobni Z, Quinaglia Araujo Costa Silva T, et al. Sodium-glucose co-transporter-2 inhibitors and cardiac outcomes among patients treated with anthracyclines. J Am Coll Cardiol HF. 2022 Aug;10(8):559–67. https://doi.org/10.1016/j.jchf.2022.03.006.

  12. Lyon AR, Dent S, Stanway S, Earl H, Brezden-Masley C, Brez-Solal A, Tocchetti CG, Moslehi JJ, Groarke JD, Bergler-Klein J, Khoo V, Tan LL, Anker MS, von Haehling S, Maack C, Pudil R, Barac A, Thavendiranathan P, Ky B, Neilan TG, Belenkov Y, Rosen SD, Iakobishvili Z, Sverdlov AL, Hajjar LA, Macedo AVS, Manisty C, Ciardiello F, Farmakis D, de Boer RA, Skouri H, Suter TM, Cardinale D, Witteles RM, Fradley MG, Herrmann J, Cornell RF, Wechelaker A, Mauro MJ, Milojkovic D, de Lavallade H, Ruschitzka F, Coats AJS, Seferovic PM, Chioncel O, Thum T, Bauersachs J, Andres MS, Wright DJ, López-Fernández T. Plummer Eur J Heart Fail. 2020;22(11):1945–60. https://doi.org/10.1002/ejhf.1920. Epub 2020 Aug 6. PMID: 32463967; PMCID: PMC8019326.

    Article  PubMed  Google Scholar 

  13. Behnoush AH, Khalaji A, Naderi N, Ashraf H, von Haehling S. ACC/AHA/HFSA 2022 and ESC 2021 guidelines on heart failure comparison. ESC Heart Fail. 2023;10:1531–44.

    Article  PubMed  Google Scholar 

  14. Vikash Jaiswal SP, Ang V, Agrawal M, Hameed MRA, Saleeb A, Jaiswal M, Shah NM, Lao. Jia Ee Chia, Kusum Paudel, Alessia Gimelli, Jerome Zacks, Association between heart failure and the incidence of cancer: a systematic review and meta-analysis. Eur Heart J Open. September 2023;3(5):oead073. https://doi.org/10.1093/ehjopen/oead073.

  15. Alexander R, Lyon T, López-Fernández LS, Couch R, Asteggiano MC, Aznar J, Bergler-Klein G, Boriani D, Cardinale R, Cordoba B, Cosyns DJ, Cutter E, de Azambuja, Rudolf A, de Boer SF, Dent D, Farmakis SA, Gevaert DA, Gorog J, Herrmann D, Lenihan J, Moslehi B, Moura, Sonja S, Salinger R, Stephens, Thomas M, Suter S, Szmit J, Tamargo P, Thavendiranathan, Carlo G, Tocchetti P, van der Meer, Helena JH, van der Pal, November, ESC Scientific Document Group., 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS): Developed by the task force on cardio-oncology of the European Society of Cardiology (ESC), European Heart Journal, 2022;43(41):4229-4361. https://doi.org/10.1093/eurheartj/ehac244

  16. Geeta Gulati SL, Heck AH, Ree P, Hoffmann J, Schulz-Menger MW, Fagerland TH, Storås. Tor-Arne Hagve, Helge Røsjø, Kjetil Steine, Jürgen Geisler, Torbjørn Omland, Prevention of cardiac dysfunction during adjuvant breast cancer therapy (PRADA): a 2 × 2 factorial, randomized, placebo-controlled, double-blind clinical trial of candesartan and metoprolol, 2016;37(21):1671-1680.European Heart Journal, Volume 37, Issue 21, 1 June 2016, Pages 1671–1680.

  17. Guglin M, Krischer J, Tamura R, Fink A, Bello-Matricaria L, McCaskill-Stevens W, Munster PN. Randomized Trial of Lisinopril Versus Carvedilol to Prevent Trastuzumab Cardiotoxicity in patients with breast Cancer. J Am Coll Cardiol. 2019;73(22):2859–68. PMID: 31171092; PMCID: PMC6557296.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Mir A, Badi Y, Bugazia S, Nourelden AZ, Fathallah AH, Ragab KM, Alsillak M, Elsayed SM, Hagrass AI, Bawek S, Kalot M, Brumberger ZL. Efficacy and safety of cardioprotective drugs in chemotherapy-induced cardiotoxicity: an updated systematic review & network meta-analysis. Cardiooncology. 2023;9(1):10. https://doi.org/10.1186/s40959-023-00159-0. PMID: 36804940; PMCID: PMC9938608.

    Article  PubMed  PubMed Central  Google Scholar 

  19. Akpek M, Ozdogru I, Sahin O, Inanc M, Dogan A, Yazici C, Berk V, Karaca H, Kalay N, Oguzhan A, Ergin A. Protective effects of spironolactone against anthracycline-induced cardiomyopathy. Eur J Heart Fail. 2015;17:81–9.

    Article  CAS  PubMed  Google Scholar 

  20. Neilan TG, Quinaglia T, Onoue T, et al. Atorvastatin for Anthracycline-Associated Cardiac Dysfunction: the STOP-CA Randomized Clinical Trial. JAMA. 2023;330(6):528–36. https://doi.org/10.1001/jama.2023.11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Cardinale D, Iacopo F, Cipolla CM. Cardiotoxicity of anthracyclines. Front Cardiovasc Med. 2020;7:26. 10.3389.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Pituskin E, Mackey JR, Koshman S, Jassal D, Pitz M, Haykowsky MJ, Pagano JJ, Chow K, Thompson RB, Vos LJ, Ghosh S, Oudit GY, Ezekowitz JA, Paterson DI. Multidisciplinary Approach to Novel therapies in Cardio-Oncology Research (MANTICORE 101-Breast): a Randomized Trial for the Prevention of Trastuzumab-Associated Cardiotoxicity. J Clin Oncol. 2017;35(8):870–7. https://doi.org/10.1200/JCO.2016.68.7830.

    Article  CAS  PubMed  Google Scholar 

  23. Bosch X, Rovira M, Sitges M, Domènech A, Ortiz-Pérez JT, de Caralt TM, Morales-Ruiz M, Perea RJ, Monzó M, Esteve J. Enalapril and carvedilol for preventing chemotherapy-induced left ventricular systolic dysfunction in patients with malignant hemopathies: the OVERCOME trial (preventiOn of left ventricular dysfunction with Enalapril and caRvedilol in patients submitted to intensive ChemOtherapy for the treatment of malignant hEmopathies). J Am Coll Cardiol. 2013;61(23):2355–62.

    Article  CAS  PubMed  Google Scholar 

  24. Moey MYY, Liles DK, Carabello B. Concomitant use of renin-angiotensin-aldosterone system inhibitors prevent trastuzumab-induced cardiotoxicity in HER2 + breast cancer patients: an institutional retrospective study. Cardio-Oncology. 2019;5:9. https://doi.org/10.1186/s40959-019-0043-8.

    Article  PubMed  PubMed Central  Google Scholar 

  25. Obasi M, Abovich A, Vo JB, Gao Y, Papatheodorou SI, Nohria A, Asnani A, Partridge AH. Statins to mitigate cardiotoxicity in cancer patients treated with anthracyclines and/or trastuzumab: a systematic review and meta-analysis. Cancer Causes Control. 2021;32(12):1395–405. https://doi.org/10.1007/s10552-021-01487-1.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Calvillo-Argüelles O, Abdel-Qadir H, Michalowska M, et al. Cardioprotective effect of statins in patients with HER2-positive breast cancer receiving trastuzumab therapy. Can J Cardiol. 2019;35:153–9.

    Article  PubMed  Google Scholar 

  27. Abdel-Qadir H, Bobrowski D, Zhou L et al. Statin exposure and risk of heart failure after anthracycline- or trastuzumab-based chemotherapy for early breast cancer: a propensity score–matched cohort study. J Am Heart Assoc 2021;Jan 19.

  28. Thavendiranathan P, Houbois C, Marwick TH et al. Statins to prevent early cardiac dysfunction in cancer patients at increased cardiotoxicity risk receiving anthracyclines. Eur Heart J Cardiovasc Pharmacother 2023;Apr 29.

  29. McMurray JJV, Solomon SD, Inzucchi SE, Køber L, Kosiborod MN, Martinez FA, Ponikowski P, Sabatine MS, Anand IS, Bělohlávek J, Böhm M, Chiang CE, Chopra VK, de Boer RA, Desai AS, Diez M, Drozdz J, Dukát A, Ge J, Howlett JG, Katova T, Kitakaze M, Ljungman CEA, Merkely B, Nicolau JC, O’Meara E, Petrie MC, Vinh PN, Schou M, Tereshchenko S, Verma S, Held C, DeMets DL, Docherty KF, Jhund PS, Bengtsson O, Sjöstrand M, Langkilde AM. DAPA-HF Trial committees and investigators. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med. 2019;381(21):1995–2008. https://doi.org/10.1056/NEJMoa1911303.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

None.

Funding

None.

Author information

Authors and Affiliations

Authors

Contributions

AD y VG conducted the study, wrote and reviewed the document. DC y TLF wrote and reviewed the document.

Corresponding author

Correspondence to Andrés J. Daniele.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, 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 you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. 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-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Daniele, A.J., Gregorietti, V., Costa, D. et al. Use of EMPAgliflozin in the prevention of CARDiotoxicity: the EMPACARD – PILOT trial. Cardio-Oncology 10, 58 (2024). https://doi.org/10.1186/s40959-024-00260-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40959-024-00260-y