Abstract
The global incidence of breast cancer is on the rise, a trend also observed in South Korea. However, thanks to the rapid advancements in anticancer therapies, survival rates are improving. Consequently, post-treatment health and quality of life for breast cancer survivors are emerging as significant concerns, particularly regarding treatment-related cardiotoxicity. In this review, we delve into the cardiovascular complications associated with breast cancer treatment, explore surveillance protocols for early detection and diagnosis of late complications, and discuss protective strategies against cardiotoxicity in breast cancer patients undergoing anticancer therapy, drawing from multiple guidelines.
Keywords: Anthracyclines, Breast Neoplasms, Cardiotoxicity, Echocardiography, Trastuzumab
INTRODUCTION
Globally, breast cancer ranks as the most commonly diagnosed cancer among females, with approximately 2.3 million new cases reported in 2020, and its prevalence is on the rise [1]. Fortunately, findings from a global cancer surveillance report by the CONCORD Working Group covering the years 2000 to 2014 indicate an age-standardized 5-year net survival rate of approximately 85%–90% for breast cancer in developed countries, including South Korea, reflecting an overall improvement in survival rates over time [2]. However, the risk of treatment-related cardiovascular (CV) toxicity, including heart failure (HF), among cancer survivors is becoming increasingly concerning over the long term [3,4,5]. Moreover, newer anticancer agents, such as immune checkpoint inhibitors (ICIs) and antibody-drug conjugates (ADCs), are being rapidly developed and used concomitantly or sequentially with traditional anticancer drugs known to have cardiotoxic effects; however, their combined CV toxicity has not been fully clarified. This review considers recent issues in breast cancer treatment, focusing on treatment-related CV complications, surveillance protocols for early detection and diagnosis of late complications, and protective strategies against cardiotoxicity, drawing from recent guidelines such as those from the European Society of Cardiology (ESC-2022) [6], American Society of Clinical Oncology (ASCO-2017) [7], and European Society for Medical Oncology (ESMO-2020) [8].
CV DISEASES IN PATIENTS WITH BREAST CANCER
A nationwide population-based cohort study conducted in Korea from 2002 to 2013 [9] revealed that 4% of breast cancer patients had preexisting CV diseases at the time of their cancer diagnosis, while 8% were newly diagnosed with CV diseases, primarily within 5 years following their cancer diagnosis. Additionally, both preexisting and newly diagnosed CV diseases were associated with higher mortality rates. Notably, cancer and CV diseases share genetic and molecular mechanisms in their pathophysiology, including gene mutations such as those in the TET2 gene, systemic inflammation, oxidative stress conditions, and common risk factors such as advanced age, obesity, hypertension, diabetes, hyperlipidemia, vascular diseases, and smoking [9,10,11,12]. Moreover, several anticancer therapies are known to be cardiotoxic [6]. Pediatric, adolescent, and young adult survivors of cancer are at a heightened risk of incident CV diseases compared to the general population, a risk that persists for over 30 years, particularly among those who have undergone high-dose cardiac radiation [3,5] and anthracycline (AC) therapies [3,4].
Competing mortality rates from CV causes and due to primary cancer in adult survivors of cancer may vary according to age group and cancer type. Strongman et al. [13] demonstrated that mortality due to primary cancer was higher than that from CV causes in cancer survivors aged under 60 years; however, CV mortality in elderly patients has surpassed cancer mortality over time. In their study, among breast cancer survivors, the crossover points at which CV disease mortality rates equal cancer mortality rates were 12.7 years (95% confidence interval [CI], 11.6–21.6) for those aged 65–79 years and 7.1 years (95% CI, 6.3–12.6) for those aged 80 years and above. Therefore, more careful CV monitoring is required in older survivors of cancer.
COLLABORATIVE APPROACH: MULTIDISCIPLINARY TEAM-BASED DECISION MAKING
Interrupting the use of a specific anticancer drug in patients with cancer, even if cardiotoxicity occurs, is complicated. This is especially true in scenarios where chemotherapy regimens are limited or if the chemotherapeutic agent offers a significant survival benefit compared with other agents. Consequently, it is crucial to assess the CV risks of patients prior to the initiation of anticancer treatment. Moreover, for high-risk patients, cardiac monitoring protocols should be planned for the entire duration of anticancer therapy, using a collaborative approach (Figure 1) [6,14].
Figure 1. Collaborative approach strategy for cardiotoxicity monitoring.
CV = cardiovascular; DM = diabetes mellitus; HT = hypertension; CVD = cardiovascular disease; HF = heart failure; VHD = valvular heart disease; CAD = coronary artery disease; LVEF = left ventricular ejection fraction; AC = anthracycline; RT = radiotherapy; DOX = doxorubicin; ECG = electrocardiography; TTE = transthoracic echocardiography; cTn = cardiac troponin; NP = natriuretic peptide.
*In anthracycline and anti-human epidermal growth factor 2 therapies; †If CV toxicity develops.
The hemodynamic conditions and severity of comorbid diseases in patients undergoing anticancer therapies can fluctuate due to factors, such as sympathetic stimulation from emotional or physical stress and changes in volume status, including fluid loading during chemotherapy infusion and dehydration from chemotherapy-related gastrointestinal problems. In addition, vascular endothelial growth factor-targeting drugs can cause uncontrolled or severe hypertension owing to their underlying mechanisms. Therefore, personalized patient surveillance should be maintained throughout the entire period of anticancer therapy.
MONITORING FOR ANTICANCER THERAPY-RELATED CARDIOTOXICITY
CV toxicity is typically monitored using transthoracic echocardiography (TTE), serologic cardiac biomarkers, and electrocardiography (ECG) [6,15]. If echocardiographic images are of poor quality or nondiagnostic, cardiac magnetic resonance imaging (MRI) may serve as an alternative [16]. TTE is recommended to assess the global longitudinal strain (GLS) and 3-dimensional left ventricular ejection fraction (LVEF) if feasible, in addition to estimating the 2-dimensional LVEF using the biplane Simpson method. LVEF, assessed using 2- and 3-dimensional methods, indirectly measures myocardial function by tracking volumetric changes in the left ventricle across end-diastole and end-systole (Figure 2). Conversely, GLS directly evaluates myocardial function by quantifying myocardial deformation between end-diastole and end-systole. GLS measurement is particularly useful for screening patients with subclinical cardiac dysfunction who exhibit mildly reduced or low-normal LVEF [17]. Therefore, the GLS value can serve as an early marker for cancer therapy-related cardiac dysfunction (CTRCD). Figure 3 illustrates the decrease in GLS values and the shift to an abnormal cardiac troponin (cTn) level prior to a significant decline in LVEF in a patient undergoing AC-containing chemotherapy over a period of 3 months.
Figure 2. Assessment of cardiac function on echocardiography: left ventricular ejection fraction and global longitudinal strain.
LVEF = left ventricular ejection fraction; GLS = global longitudinal strain; Ai, Bi = cylinder volumes, L = left ventricular longitudinal diameter; Ld = length at diastolic period; Ls = length at systolic period; V = volume; n = number of cylinders; EDV = end-diastolic volume; ESV = end-systolic volume.
Figure 3. Serial transthoracic echocardiography of a 60-year-old female with breast cancer undergoing doxorubicin-docetaxel chemotherapy. (A) Baseline TTE, (B) TTE 3 months later.
TTE = transthoracic echocardiography; ANT_SEPT = anteroseptal; SEPT = septal; ANT = anterior; INF = inferior; POST = posterior; LAT = lateral; GLS = global longitudinal strain; LVEF = left ventricular ejection fraction; cTn = cardiac troponin; Δ = change.
CTRCD: Definition and screening methods
CTRCD is generally defined as a decrease in GLS of ≥ 15% from baseline or a decline in LVEF of ≥ 10% to an absolute LVEF of < 50%–53%, although the cut-off values vary according to guidelines [18,19]. If CTRCD is suspected, a follow-up TTE for confirmation is recommended within 2 to 3 weeks because the measurements are load-dependent [20,21]. Additionally, GLS values are vendor-dependent; thus, follow-up TTE should be performed with the same equipment.
Serologic cardiac biomarkers, including cTn and natriuretic peptides (NPs) such as brain natriuretic peptide (BNP) and N-terminal pro-B-type natriuretic peptide (NT-proBNP), are easier to measure than TTE; thus, they are useful for screening CTRCD in patients with low- and moderate-CV risk or for monitoring CTRCD between follow-up TTE measurements. However, NP and cTn levels may be influenced by clinical characteristics, including age, sex, and comorbid conditions, such as chronic kidney disease, lung disease, atrial fibrillation (AF), and obesity [22,23,24]. Therefore, serial changes in the levels of cardiac biomarkers should be considered during anticancer therapies, in addition to their absolute levels [15].
Monitoring of arrhythmia in patients receiving cancer therapy
Cancer can cause arrhythmias due to chronic inflammation and metabolic changes, and both cancer and arrhythmia have common risk factors such as obesity, alcohol abuse, and smoking [25]. Moreover, various anticancer chemotherapies can trigger ECG changes and arrhythmias, either directly or secondarily through cardiomyopathies caused by ischemic origins or CTRCD [25,26].
QT prolongation occurs frequently during chemotherapy and is linked to torsade de pointes, ventricular fibrillation, and sudden cardiac death [27]. A study reported that significant corrected QT prolongation (> 450 ms) occurred in 15% of patients after the first cycle of AC chemotherapy and increased with subsequent cycles [28]. Additionally, electrolyte imbalances, including hypokalemia, and various drugs, such as antibiotics, antidepressants, and antiemetics, can contribute to QT prolongation. AF is one of the significant complications of chemotherapy [29,30]. Therefore, ECG monitoring is essential both at the start of chemotherapy and throughout its course, with Holter monitoring being considered if necessary.
AC- AND ANTI-HUMAN EPIDERMAL GROWTH FACTOR RECEPTOR 2 THERAPY-RELATED CARDIOTOXICITY
Case: ACs followed by trastuzumab
A 59-year-old woman with no prior medical history was diagnosed with human epidermal growth factor receptor 2 (HER2)-positive breast cancer (T3N1M0, stage 3A) and underwent neoadjuvant chemotherapy with doxorubicin (DOX) (cumulative dose = 300 mg/m2) and docetaxel, followed by right modified radical mastectomy. Subsequently, the patient was scheduled for a year of trastuzumab treatment. Before commencing trastuzumab, TTE revealed an LVEF of 61% with no evidence of structural heart disease. However, four months into trastuzumab treatment, the patient complained of dyspnea, and her LVEF plummeted to 29%. Following consultation with a cardiologist and discussion with her oncologist, trastuzumab treatment was halted until her LVEF improved. The patient received guideline-directed medical therapy for HF, including angiotensin-converting enzyme inhibitors, beta blockers (BBs), and spironolactone. During serial TTE follow-ups over 7 years, her LVEF fluctuated between 30%–50%, ultimately leading to the cessation of further chemotherapy.
Risk factors for AC- and HER2-targeted therapy-related cardiotoxicity
Traditional CV risk factors, including advanced age (generally ≥ 65 years, with a particularly high risk in those aged ≥ 75 to 80 years), hypertension, diabetes mellitus (DM), chronic kidney disease, current or significant past smoking history, and obesity (body mass index > 30 kg/m2), are known concurrent risk factors for AC- and trastuzumab-related cardiotoxicity [6,7,31]. Additionally, preexisting CV comorbidities, such as HF or LVEF < 50%, significant valvular heart disease, coronary artery disease or angina, and arrhythmias like AF, contribute to increased cardiotoxicity risk [6,7,31]. Prior exposure to other cardiotoxic anticancer therapies, including ACs, trastuzumab, and mediastinal or left chest radiotherapy, further increases the risk. A previous randomized controlled trial (RCT) showed that CTRCD occurred in 8% of patients with HER2-positive breast cancer receiving the AC regimen alone and in 27% of those receiving the AC regimen with trastuzumab treatment [32]. Furthermore, if the total planned cumulative dose of AC is ≥ 250 mg/m2 of DOX or its equivalent, the risk of cardiotoxicity is high, even when used alone [6]. In the present case, the patient received a cumulative dose of DOX of 300 mg/m2, followed by trastuzumab treatment. This might have been the etiology of irreversible HF despite the absence of any CV risk factors.
Genetic susceptibility to AC-related cardiotoxicity may also play a role in the development of cardiotoxicity. Recent genetic studies utilizing genomes from pediatric and adult cancer survivors have identified specific genetic variants that are susceptible to AC-related cardiotoxicity, which may explain the high inter-individual variability for CTRCD [33,34]. These variants are involved in the pathogenesis of topoisomerase 2β-mediated DNA damage, AC transport and metabolic pathways, oxidative stress generation, altered iron homeostasis, and sarcomere dysfunction [33,34,35,36,37,38,39,40,41,42,43]. In the near future, genetic testing could be explored to identify high-risk patients, in addition to evaluating clinical risk factors.
Demarcation between reversible and irreversible CTRCD
In four major trials evaluating the efficacy and safety of trastuzumab treatment, including the HERceptin Adjuvant (HERA) [44], National Surgical Adjuvant Breast and Bowel Project (NSABP) B-31 [45], North Central Cancer Treatment Group (NCCTG) N9831 [46,47], and Breast Cancer International Research Group (BCIRG) 006 [48], significant HF, classified as New York Heart Association (NYHA) grades III–IV with a decline in LVEF of ≥ 10% from baseline to a final LVEF < 50%, occurred in 0.4%–4% of patients undergoing trastuzumab treatment. Asymptomatic declines in LVEF, classified as NYHA grades I–II with the same LVEF decline criteria, occurred in 3–18% of patients undergoing trastuzumab treatment. Discontinuation of trastuzumab due to CTRCD occurred in 4.3% of patients in the HERA trial and 19% in the NCCTG N9831 trial. However, most patients experiencing CTRCD recovered within 6 months after trastuzumab discontinuation, and the long-term CV outcomes were favorable, with most CV events occurring during the treatment phase [45,49]. Conversely, a retrospective study by Yoon et al. [50] showed that persistent cardiotoxicity of ≥ 6 months was observed in 20% of patients with breast cancer undergoing trastuzumab treatment. In their study, 70% of those with persistent cardiotoxicity underwent prior AC administration (a cumulative dose of DOX = 356 ± 55 mg/m2), and TTE follow-up between the end of AC treatment and the start of trastuzumab was not mandatory. Therefore, it remains uncertain whether the cardiotoxicity is due to ACs, trastuzumab, or a combination of both.
Generally, AC-related cardiotoxicity is recognized as dose-dependent and irreversible, in contrast to the dose-independent and reversible nature of cardiotoxicity associated with HER2-targeted therapy [31,51]. Furthermore, AC-related cardiotoxicity is primarily observed within approximately 30 days following its administration but can manifest up to 6–10 years later [52,53]. Many patients with HER2-positive breast cancer undergo AC chemotherapy before trastuzumab treatment, as observed in the present case. Consequently, LV dysfunction observed during trastuzumab treatment often does not improve despite the interruption of trastuzumab and optimized management for HF. Careful CV monitoring is required when cardiotoxic chemotherapy is administered concomitantly or sequentially.
Surveillance protocols for cardiotoxicity
Surveillance protocols for cardiotoxicity based on recent guidelines are outlined in Table 1 [6,7,8]. During AC chemotherapy, TTE measurements are usually recommended at the initiation period, upon reaching a cumulative dose of ≥ 250 mg/m2 of DOX or its equivalent, and within 12 months after completing chemotherapy. TTE can be performed more frequently, every two cycles of chemotherapy, and additionally within 3 months after completing chemotherapy for high-risk patients. Serologic cardiac biomarker measurements are recommended every two cycles of chemotherapy or every cycle for high-risk patients. An ECG should be performed initially. If arrhythmia-associated symptoms occur during anticancer therapy, follow-up ECG and/or Holter monitoring should be considered. For patients undergoing HER2-targeted therapy, monitoring for CTRCD is recommended by performing TTE with cardiac biomarkers at the initiation of treatment, every 3 months during chemotherapy, and within 12 months after completing chemotherapy. Additionally, for high-risk patients, it is advised to conduct further monitoring within 3 months post-chemotherapy. The frequency of cardiac monitoring for HER2-targeted therapy has not been established based on risk stratification, as suggested by a broad consensus on expert opinions. Therefore, TTE may be performed more frequently in high-risk patients [6], whereas in low- or moderate-risk patients, it can be performed less frequently and complemented by cardiac biomarkers [14].
Table 1. Breast cancer therapy-related cardiotoxicity, monitoring protocols for early detection, and treatment strategies.
| Therapy | Drugs | Cardiotoxicity | Monitoring | Diagnosis/Treatment | ||
|---|---|---|---|---|---|---|
| ACs | Doxorubicin, epirubicin | LV dysfunction, HF | TTE: | LVEF < 50%†: | ||
| - Baseline, DOX ≥ 250 mg/m2*, 12 M post-Tx | - HF therapy: RAS-Is, BBs | |||||
| - Additionally, every 2 cycles and 3 M post-Tx in high-risk patients | - The withdrawal of the AC use‡ | |||||
| - HF symptoms (occurrence) | Consider dexrazoxane for DOX* ≥ 300 mg/m2 or liposomal ACs in high-risk patients | |||||
| Cardiac biomarkers (cTn, NPs): | ||||||
| - Baseline, every 2 cycles (every cycle in high- risk patients), DOX ≥ 250 mg/m2*, 3 M post-Tx | ||||||
| - Additionally, 12 M post-Tx in high-risk patients | ||||||
| Arrhythmia, QT prolongation | ECG, Holter monitoring (if symptomatic) | |||||
| HER2-targeted drugs | Trastuzumab, pertuzumab | LV dysfunction, HF | TTE and cardiac biomarkers (cTn, NPs): | LVEF < 40%†: | ||
| - Baseline, every 3 M, 12 M post-Tx | - HF therapy: RAS-Is, BBs | |||||
| - Additionally, 3 M post-Tx in high-risk patients | - The withdrawal of the culprit drug use until LVEF is recovered to ≥ 40%§ and symptoms are absent | |||||
| LVEF = 40%–49%: | ||||||
| - HF therapy: RAS-Is, BBs | ||||||
| - Continue chemotherapy under frequent CV monitoring if asymptomatic | ||||||
| TTE every 3–6 weeks until stable | ||||||
| Antimetabolites | 5-FU, capecitabine | Coronary vasospasm, ventricular arrythmia | Chest pain, ECG, cardiac biomarkers, TTE, Holter, coronary angiography | Nitrates, CCBs for coronary vasospasm | ||
| Consider interrupting the culprit drug use or changing the infusion regimen | ||||||
AC = anthracycline; LV = left ventricular; HF = heart failure; TTE = transthoracic echocardiography; DOX = doxorubicin; M = month; Tx = treatment; cTn = cardiac troponin; NP = natriuretic peptide; ECG = electrocardiography; LVEF = left ventricular ejection fraction; RAS-Is = renin-angiotensin system inhibitors (including renin-angiotensin-converting enzyme inhibitors and angiotensin receptor blockers); BB = beta blocker; HER2 = human epidermal growth factor 2; CV = cardiovascular; FU = fluorouracil; CCB = calcium channel blocker; ESC = European Society of Cardiology; ESMO = European Society for Medical Oncology.
*A cumulative dose of DOX or its equivalent AC, and then every 2 cycles (additionally administration of DOX of 100 mg/m2 or epirubicin of 200 mg/m2) thereafter; †If signs and symptoms of HF do not resolve and/or the LVEF remains < 40% after interrupting the culprit drug, and there are no alternative therapeutic options, reconsideration of the culprit drug’s use may be warranted; ‡If the LVEF recovers to ≥ 50% and symptoms are absent or mild, AC chemotherapy may be resumed with guideline-based HF management and close CV monitoring after a risk-benefit ratio assessment. §If the LVEF recovers to ≥ 40% (as per ESC-2022 guidelines) or ≥ 50% (as per ESMO-2020 guidelines) and symptoms are absent, HER2-targeted therapy should be considered with HF management.
If significant CTRCD with LVEF < 50% occurs during AC-containing or HER-2 targeted chemotherapy, HF management should be initiated, and it is recommended to make decisions regarding the continuation or interruption of chemotherapy through a multidisciplinary team (MDT) approach. Considering the restart of chemotherapy after the improvement of LVEF and symptoms should be done with frequent CV monitoring, following a discussion with the MDT. For asymptomatic patients experiencing mild CTRCD with LVEF ≥ 50% accompanied by a significant drop in GLS or increased cardiac biomarkers, it is recommended to continue the chemotherapy with HF management and frequent CV monitoring.
ANTIMETABOLITE-RELATED CARDIOTOXICITY
If atypical chest pain occurs during treatment with 5-fluorouracil (5-FU) or its oral prodrug capecitabine, coronary vasospasm should be initially suspected (Table 1). It should also be differentially diagnosed from ischemic heart disease, arrhythmia, or other cardiomyopathies. 5-FU-associated coronary vasospasm mainly occurs during the first administration of 5-FU but can also occur after several cycles of infusion [54]. Its incidence has been reported in the range of 1%–5% in prospective studies [55]; however, it might be underreported, as it can be overlooked due to atypical chest pain presentation. Currently, there is no evidence that prophylactic use of anti-anginal medications effectively prevents coronary vasospasm [56].
Upon the occurrence of coronary vasospasm, withdrawal or continuation of 5-FU treatment should be considered based on the risk-to-benefit ratio estimation. Some case reports have described successful rechallenge with 5-FU infusion after administering strong anti-anginal medications, including calcium channel blockers (CCBs) and nitrates, accompanied by close monitoring and/or a transition from continuous infusion to a bolus infusion or dose reduction regimen [57]. However, re-initiation of 5-FU can induce serious complications such as myocardial infarction, fatal arrhythmia, and sudden cardiac death [58,59,60].
PRIMARY AND SECONDARY PREVENTION FOR CTRCD
Renin-angiotensin system inhibitors (RAS-Is), including renin-angiotensin-converting enzyme inhibitors and angiotensin receptor blockers, and BBs
When CTRCD occurs, optimized management of HF, including RAS-Is and BBs, is recommended regardless of the presence or absence of symptoms and the severity of CTRCD [6,7,8]. However, RCT data supporting HF management in asymptomatic patients with mild CTRCD are limited [61].
Several RCTs and meta-analyses have shown that the prophylactic use of RAS-Is or BBs tends to decrease the decline in LVEF associated with CTRCD, although the statistical significance of these findings is not consistent across studies [62,63,64,65]. However, there is no conclusive evidence that its prophylactic use significantly reduces the incidence of HF. Additionally, concerns regarding increased hypotension have been raised [66,67]. Therefore, the prophylactic use of RAS-Is or BBs to prevent CTRCD can be considered for high-risk patients after careful evaluation of the risk-benefit ratio.
Separately, using RAS-Is as antihypertensive drugs can benefit patients with hypertension undergoing cardiotoxic chemotherapy, a benefit not observed with BBs. In a previous study [68], we demonstrated that RAS-Is improved clinical outcomes against DOX-related toxicity in patients with breast cancer and hypertension, compared to normotensive patients receiving DOX-containing chemotherapy. Moreover, RAS-Is and CCBs have shown more favorable CV outcomes compared to BBs and thiazide/thiazide-like diuretics in patients with hypertension and breast cancer undergoing DOX therapy [69]. Additionally, CCBs have demonstrated comparable CV outcomes to RAS-Is in patients with hypertension receiving DOX therapy. CCBs are superior to RAS-Is in lowering both central and peripheral blood pressures, reducing blood pressure variability, and decreasing arterial stiffness [70,71]. Therefore, the selection of antihypertensive medications should be tailored to patients’ clinical conditions. When dual antihypertensive therapy is necessary, the combination of CCBs and RAS-Is is recommended [6].
Statins
Several experimental studies have demonstrated that statins reduce oxidative stress and cardiac inflammation, potentially preventing AC-related cardiotoxicity [72]. Clinical studies have also shown protective effects of statins against CTRCD [73] and HF [74,75]. However, two recent major RCTs evaluating the efficacy of statins in preventing CTRCD, including the Preventing Anthracycline Cardiovascular Toxicity with Statins (PREVENT) [76] and Statins TO Prevent the Cardiotoxicity from Anthracyclines (STOP-CA) [77], showed conflicting results (Table 2). Both studies matched participants in a 1:1 ratio between the statin (atorvastatin 40 mg/day) and control groups during AC chemotherapy. The STOP-CA trial included patients with lymphoma who received a higher cumulative dose of AC in their chemotherapy regimens, whereas the PREVENT trial included patients with breast cancer and lymphoma at rates of 85% and 15%, respectively. Consequently, the median cumulative dose of ACs was 300 mg/m2 in the STOP-CA trial and 240 mg/m2 in the PREVENT trial. The outcomes, estimated by changes in LVEF following AC chemotherapy, showed a positive protective effect in the STOP-CA trial but not in the PREVENT trial. Based on these results, the prophylactic use of statins remains controversial but may be considered for high-risk patients scheduled to receive high cumulative doses of ACs [78].
Table 2. Two randomized controlled trials evaluating the efficacy of prophylactic use of statins during anthracycline-containing chemotherapy.
| Characteristics | PREVENT | STOP-CA | |
|---|---|---|---|
| Age (yr), mean ± SD | 49 ± 12 | 50 ± 17 | |
| No. of subjects (case:control) | 139:140 | 150:150 | |
| Cancer type | |||
| Breast | 85% | 0% | |
| Lymphoma | 15% | 100% | |
| Intervention | Atorvastatin 40 mg/day | Atorvastatin 40 mg/day | |
| Median cumulative AC dose | 240 mg/m2 | 300 mg/m2 | |
| Follow-up duration | 24 months | 12 months | |
| Primary end point | ΔLVEF/initial LVEF | ΔLVEF ≥ 10% decline to < 55% | |
| Case vs. Control | 3.2% ± 0.7% vs. 3.3% ± 0.6% | 13 vs. 33 events | |
| p-value | 0.93 | 0.002 | |
PREVENT = the Preventing Anthracycline Cardiovascular Toxicity with Statins; STOP-CA = the Statins TO Prevent the Cardiotoxicity from Anthracycline; SD = standard deviation; AC = anthracycline; Δ = change; LVEF = left ventricular ejection fraction
Sodium-glucose cotransporter 2 (SGLT2) inhibitors
Experimental and clinical studies have shown that SGLT2 inhibitors inhibit intracellular glucose metabolism [79], reduce inflammatory cytokines [79,80], and improve endothelial function [81]. Furthermore, several RCTs have demonstrated that SGLT2 inhibitors improve CV outcomes in patients with HF, regardless of type 2 DM [82,83,84,85]. Recent retrospective studies have shown that SGLT2 inhibitors improve CV outcomes in patients with DM and cancer receiving AC chemotherapy [86,87,88]. In addition, the use of SGLT2 inhibitors was associated with reduced mortality compared with patients without DM and those with DM not using SGLT2 inhibitors [87]. Experimental studies involving cancer cell lines and animal models have demonstrated that SGLT2 inhibitor treatment reduces glucose uptake and mitochondrial function in cancer cells, thereby decreasing cell proliferation and clonogenic survival [89,90]. This contributes to inhibiting the development and growth of cancer cells [89,91,92]. However, whether the effects of SGLT2 inhibitors on clinical outcomes are consistent in cancer patients without DM has not been clarified. Fortunately, prospective studies, such as EMPAgliflozin in prevention of chemotherapy-related CardioToxicity (EMPACT; NCT05271162), are ongoing to evaluate the cardioprotective effects of SGLT2 inhibitors in high-risk patients undergoing AC chemotherapy.
Dexrazoxane
Meta-analyses have demonstrated that dexrazoxane significantly reduces the relative risk of HF by up to 71%–76% in patients receiving AC therapy, without significant differences in response rate, progression-free survival, and overall survival [93,94,95]. However, the use of dexrazoxane is limited to the prevention of cardiotoxicity in women with metastatic breast cancer who have received a cumulative dose of DOX of 300 mg/m2 or its equivalent and are continuing DOX therapy [94].
PRESENT AND FUTURE ANTICANCER DRUGS AND EXPECTED CARDIOTOXICITY: ICIs AND ADCs
The anticancer efficacies of recently developed ICIs and ADCs are outstanding; however, their cardiotoxic risks have not yet been fully clarified. Adverse effects of ICIs, such as pembrolizumab and atezolizumab, are associated with immune-related toxicity [96]. Cardiotoxicity from ICIs, including myocarditis, pericarditis, arrhythmia, and HF, occurs rarely, affecting approximately 0.1%–1% of cases [97,98,99]. However, the risk may increase with combination therapy and can be fulminant or fatal [97,100]. Baseline and serial ECGs and cardiac biomarker assessments are not predictive of ICI-mediated cardiotoxicity. However, changes in cardiac biomarkers, particularly cTn, are considered sensitive for early detection of cardiotoxicity. A cohort study showed that the incidence of ICI-related HF was higher in patients with previous HF or valvular heart disease [101]. Echocardiography should be considered first when cardiotoxicity is suspected. If not diagnostic, cardiac MRI and biopsy should be considered. Currently, when ICI-mediated cardiotoxicity of grade 2 (characterized by cTn elevation with mild symptoms) or higher, as defined by the Common Terminology Criteria for Adverse Events, occurs, it is recommended to discontinue ICIs and administer early high-dose corticosteroids (1–2 mg/kg/day of prednisolone) [97,98].
ADCs, including trastuzumab deruxtecan (T-DXd) and trastuzumab emtansine (T-DM1), have lower cardiotoxicity than trastuzumab. A meta-analysis on T-DXd-related cardiotoxicity reported an incidence of LVEF decline of 1.95% (95% CI, 0.65%–3.73%) and an incidence of HF of 0.26% among patients with breast cancer [102]. A retrospective pharmacovigilance study using VigiBase, the World Health Organization database for adverse drug reaction reports, indicated that trastuzumab had a higher rate of HF reports (12%) compared with ADCs (1.9% for pertuzumab, 1.7% for T-DM1, and 1.2% for T-DXd) [103]. Additionally, combination therapy involving trastuzumab with pertuzumab or other HER2 tyrosine kinase inhibitors was associated with higher odds of HF reports than the combination of trastuzumab with platinum-based chemotherapy [103]. The study also found that pertuzumab (reporting odd ratio [ROR], 0.13; 99% CI, 0.07–0.23), T-DXd (ROR, 0.06; 99% CI, 0.03–0.15), and T-DM1 (ROR, 0.10; 99% CI, 0.07–0.14) had lower odds of HF compared to trastuzumab in patients with breast cancer. However, the combination therapy of pertuzumab and T-DM1 presented higher odds of HF reporting than T-DM1 monotherapy in patients with metastatic breast cancer (ROR, 3.35; 99% CI, 2.04–5.52). Therefore, combination therapies involving trastuzumab with pertuzumab or pertuzumab with T-DM1 may require close monitoring.
LONG-TERM SURVEILLANCE FOR BREAST CANCER SURVIVORS
In a study investigating patients who underwent radiotherapy for breast cancer between the 1970s and the early 1980s, long-term CV mortality increased persistently over 15 years [104]. A population-based cohort in England demonstrated that the incidence of each CV disease increased with a cumulative DOX dose of 150 mg/m2 or with calculated heart doses of radiotherapy in patients with Hodgkin lymphoma, compared with an age- and sex-matched population, as follows: HF at an average heart dose of 3.3 Gy (range, 0.3–20.5), ischemic heart disease at an average of 7.8 Gy (range, 0.8–24.0), and valvular heart disease at an average of 12.6 Gy (range, 0.8–33.1) [105]. Therefore, long-term CV monitoring is required even after the completion of cancer therapy, particularly in patients who have undergone AC therapy or mediastinal or left chest radiotherapy.
Generally, after the completion of cancer therapy, reassessing CV risk is recommended [6]. High risk includes high-dose AC (cumulative DOX dose ≥ 250 mg/m2 or epirubicin dose ≥ 600 mg/m2), high-dose radiotherapy (mean heart dose of ≥ 15–25 Gy as per ESC-2022 or ≥ 30 Gy in ASCO-17), and the combination of lower-dose AC (DOX dose of 100–249 mg/m2) and lower-dose radiotherapy (5–15 Gy in ESC-2022 or < 30 Gy in ASCO-17), and the presence of CTRCD of more than moderate grade (LVEF < 50%) during treatment [6,7]. Among patients receiving low-dose AC or trastuzumab alone, those aged ≥ 60 years or with multiple CV risk factors (≥ 2), including smoking, hypertension, diabetes, dyslipidemia, and obesity, are also considered high risk [7]. High-risk patients are advised to undergo ECG and TTE with GLS assessment at 6–12 months and 2 years post-treatment [7,8], or at 1, 3, and 5 years post-treatment [6]. Given that CV toxicity due to mediastinal or left chest radiotherapy can manifest with a delay, it is recommended to commence CV surveillance at 5 years post-treatment, and then continue every 5 years thereafter [6,8].
CONCLUSION
As breast cancer therapies continue to evolve and the number of cancer survivors increases, treatment-related CV toxicity has emerged as a significant concern. In particular, AC-based and/or HER2-targeted therapies may lead to CTRCD during treatment and within 5 years after completion [6]. Additionally, heart radiation due to radiotherapy can increase the risk of delayed CV disease for more than 30 years. Therefore, comprehensive CV risk assessment, vigilant monitoring for early detection, and appropriate management of CV diseases are essential before, during, and after cancer treatment. This necessitates a collaborative MDT approach that includes oncologists, cardiologists, and radiologists.
Emerging evidence regarding the cardioprotective potential of statins and SGLT2 inhibitors, as well as traditional HF management, offers promising avenues for both research and clinical practice. Moreover, newly developed regimens, including ICIs, ADCs, and HER2 tyrosine kinase inhibitors, appear to be safer than earlier chemotherapeutic agents in terms of cardiotoxicity, although their effects are not fully understood. Ultimately, our hope is for cancer survivors to live healthily and maintain a high quality of life free from CV diseases.
Footnotes
Conflict of Interest: The authors declare that they have no competing interests.
Data Availability: In accordance with the ICMJE data sharing policy, the authors have agreed to make the data available upon request.
- Conceptualization: Hwang HJ, Han SA, Sohn IS.
- Data curation: Hwang HJ.
- Formal analysis: Hwang HJ.
- Investigation: Hwang HJ.
- Methodology: Hwang HJ, Sohn IS.
- Supervision: Han SA, Sohn IS.
- Validation: Hwang HJ, Han SA.
- Visualization: Han SA, Sohn IS.
- Writing - original draft: Hwang HJ.
- Writing - review & editing: Han SA, Sohn IS.
References
- 1.Ferlay JE, Lam F, Colombet M, Mery L, Piñeros M, Znaor A, et al. Global Cancer Observatory: Cancer Today. [Accessed March 31st, 2022]. https://gco.iarc.fr/today .
- 2.Allemani C, Matsuda T, Di Carlo V, Harewood R, Matz M, Nikšić M, et al. Global surveillance of trends in cancer survival 2000-14 (CONCORD-3): analysis of individual records for 37 513 025 patients diagnosed with one of 18 cancers from 322 population-based registries in 71 countries. Lancet. 2018;391:1023–1075. doi: 10.1016/S0140-6736(17)33326-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Mulrooney DA, Yeazel MW, Kawashima T, Mertens AC, Mitby P, Stovall M, et al. Cardiac outcomes in a cohort of adult survivors of childhood and adolescent cancer: retrospective analysis of the Childhood Cancer Survivor Study cohort. BMJ. 2009;339:b4606. doi: 10.1136/bmj.b4606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Hibler E, Tanaka Y, Akhter N, Murphy K, Perlman K, Lloyd-Jones D, et al. Risk of incident heart failure among young adult cancer survivors. JACC CardioOncol. 2023;5:539–541. doi: 10.1016/j.jaccao.2023.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bhakta N, Liu Q, Yeo F, Baassiri M, Ehrhardt MJ, Srivastava DK, et al. Cumulative burden of cardiovascular morbidity in paediatric, adolescent, and young adult survivors of Hodgkin’s lymphoma: an analysis from the St Jude Lifetime Cohort Study. Lancet Oncol. 2016;17:1325–1334. doi: 10.1016/S1470-2045(16)30215-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Lyon AR, López-Fernández T, Couch LS, Asteggiano R, Aznar MC, Bergler-Klein J, et al. 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) Eur Heart J. 2022;43:4229–4361. doi: 10.1093/eurheartj/ehac244. [DOI] [PubMed] [Google Scholar]
- 7.Armenian SH, Lacchetti C, Barac A, Carver J, Constine LS, Denduluri N, et al. Prevention and monitoring of cardiac dysfunction in survivors of adult cancers: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol. 2017;35:893–911. doi: 10.1200/JCO.2016.70.5400. [DOI] [PubMed] [Google Scholar]
- 8.Curigliano G, Lenihan D, Fradley M, Ganatra S, Barac A, Blaes A, et al. Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. Ann Oncol. 2020;31:171–190. doi: 10.1016/j.annonc.2019.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Youn JC, Chung WB, Ezekowitz JA, Hong JH, Nam H, Kyoung DS, et al. Cardiovascular disease burden in adult patients with cancer: an 11-year nationwide population-based cohort study. Int J Cardiol. 2020;317:167–173. doi: 10.1016/j.ijcard.2020.04.080. [DOI] [PubMed] [Google Scholar]
- 10.Masoudkabir F, Sarrafzadegan N, Gotay C, Ignaszewski A, Krahn AD, Davis MK, et al. Cardiovascular disease and cancer: evidence for shared disease pathways and pharmacologic prevention. Atherosclerosis. 2017;263:343–351. doi: 10.1016/j.atherosclerosis.2017.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Wang Y, Wang Y, Han X, Sun J, Li C, Adhikari BK, et al. Cardio-oncology: a myriad of relationships between cardiovascular disease and cancer. Front Cardiovasc Med. 2022;9:727487. doi: 10.3389/fcvm.2022.727487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Beasley GS, Towbin JA. Acquired and modifiable cardiovascular risk factors in patients treated for cancer. J Thromb Thrombolysis. 2021;51:846–853. doi: 10.1007/s11239-020-02273-7. [DOI] [PubMed] [Google Scholar]
- 13.Strongman H, Gadd S, Matthews AA, Mansfield KE, Stanway S, Lyon AR, et al. Does cardiovascular mortality overtake cancer mortality during cancer survivorship?: an English retrospective cohort study. JACC CardioOncol. 2022;4:113–123. doi: 10.1016/j.jaccao.2022.01.102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Porter C, Azam TU, Mohananey D, Kumar R, Chu J, Lenihan D, et al. Permissive cardiotoxicity: the clinical crucible of cardio-oncology. JACC CardioOncol. 2022;4:302–312. doi: 10.1016/j.jaccao.2022.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Bracun V, Aboumsallem JP, van der Meer P, de Boer RA. Cardiac biomarkers in patients with cancer: considerations, clinical implications, and future avenues. Curr Oncol Rep. 2020;22:67. doi: 10.1007/s11912-020-00930-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Vallabhaneni S, Zhang KW, Alvarez-Cardona JA, Mitchell JD, Steen H, Woodard PK, et al. Role of cardiovascular magnetic resonance in early detection and treatment of cardiac dysfunction in oncology patients. Int J Cardiovasc Imaging. 2021;37:3003–3017. doi: 10.1007/s10554-021-02271-7. [DOI] [PubMed] [Google Scholar]
- 17.Shah AM, Solomon SD. Phenotypic and pathophysiological heterogeneity in heart failure with preserved ejection fraction. Eur Heart J. 2012;33:1716–1717. doi: 10.1093/eurheartj/ehs124. [DOI] [PubMed] [Google Scholar]
- 18.Curigliano G, Cardinale D, Suter T, Plataniotis G, de Azambuja E, Sandri MT, et al. Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO Clinical Practice Guidelines. Ann Oncol. 2012;23(Suppl 7):vii155–vii166. doi: 10.1093/annonc/mds293. [DOI] [PubMed] [Google Scholar]
- 19.Plana JC, Galderisi M, Barac A, Ewer MS, Ky B, Scherrer-Crosbie M, et al. Expert consensus for multimodality imaging evaluation of adult patients during and after cancer therapy: a report from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. Eur Heart J Cardiovasc Imaging. 2014;15:1063–1093. doi: 10.1093/ehjci/jeu192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Habibian M, Lyon AR. Monitoring the heart during cancer therapy. Eur Heart J Suppl. 2019;21:M44–M49. doi: 10.1093/eurheartj/suz230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zamorano JL, Lancellotti P, Rodriguez Muñoz D, Aboyans V, Asteggiano R, Galderisi M, et al. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines: the Task Force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC) Eur Heart J. 2016;37:2768–2801. doi: 10.1093/eurheartj/ehw211. [DOI] [PubMed] [Google Scholar]
- 22.Heidenreich PA, Bozkurt B, Aguilar D, Allen LA, Byun JJ, Colvin MM, et al. 2022 AHA/ACC/HFSA Guideline for the management of heart failure: a report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation. 2022;145:e895–e1032. doi: 10.1161/CIR.0000000000001063. [DOI] [PubMed] [Google Scholar]
- 23.Mueller C, McDonald K, de Boer RA, Maisel A, Cleland JGF, Kozhuharov N, et al. Heart Failure Association of the European Society of Cardiology practical guidance on the use of natriuretic peptide concentrations. Eur J Heart Fail. 2019;21:715–731. doi: 10.1002/ejhf.1494. [DOI] [PubMed] [Google Scholar]
- 24.Suthahar N, Lau ES, Blaha MJ, Paniagua SM, Larson MG, Psaty BM, et al. Sex-specific associations of cardiovascular risk factors and biomarkers with incident heart failure. J Am Coll Cardiol. 2020;76:1455–1465. doi: 10.1016/j.jacc.2020.07.044. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Buza V, Rajagopalan B, Curtis AB. Cancer treatment-induced arrhythmias: focus on chemotherapy and targeted therapies. Circ Arrhythm Electrophysiol. 2017;10:e005443. doi: 10.1161/CIRCEP.117.005443. [DOI] [PubMed] [Google Scholar]
- 26.Agarwal MA, Sridharan A, Pimentel RC, Markowitz SM, Rosenfeld LE, Fradley MG, et al. Ventricular arrhythmia in cancer patients: mechanisms, treatment strategies and future avenues. Arrhythm Electrophysiol Rev. 2023;12:e16. doi: 10.15420/aer.2023.04. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Iwata N, Karasawa M, Omine M, Maekawa T, Suzuki T, Kawai Y. Aclarubicin-associated QTc prolongation and ventricular fibrillation. Cancer Treat Rep. 1984;68:527–529. [PubMed] [Google Scholar]
- 28.Horacek JM, Jakl M, Horackova J, Pudil R, Jebavy L, Maly J. Assessment of anthracycline-induced cardiotoxicity with electrocardiography. Exp Oncol. 2009;31:115–117. [PubMed] [Google Scholar]
- 29.Hajjar LA, Fonseca SM, Machado TI. Atrial fibrillation and cancer. Front Cardiovasc Med. 2021;8:590768. doi: 10.3389/fcvm.2021.590768. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Bao Y, Lee J, Thakur U, Ramkumar S, Marwick TH. Atrial fibrillation in cancer survivors - a systematic review and meta-analysis. Cardiooncology. 2023;9:29. doi: 10.1186/s40959-023-00180-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Barrett-Lee PJ, Dixon JM, Farrell C, Jones A, Leonard R, Murray N, et al. Expert opinion on the use of anthracyclines in patients with advanced breast cancer at cardiac risk. Ann Oncol. 2009;20:816–827. doi: 10.1093/annonc/mdn728. [DOI] [PubMed] [Google Scholar]
- 32.Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–792. doi: 10.1056/NEJM200103153441101. [DOI] [PubMed] [Google Scholar]
- 33.Al-Otaibi TK, Weitzman B, Tahir UA, Asnani A. Genetics of anthracycline-associated cardiotoxicity. Front Cardiovasc Med. 2022;9:867873. doi: 10.3389/fcvm.2022.867873. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ding Y, Du K, Niu YJ, Wang Y, Xu X. Genetic susceptibility and mechanisms underlying the pathogenesis of anthracycline-associated cardiotoxicity. Oxid Med Cell Longev. 2022;2022:5818612. doi: 10.1155/2022/5818612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Schneider BP, Shen F, Gardner L, Radovich M, Li L, Miller KD, et al. Genome-wide association study for anthracycline-induced congestive heart failure. Clin Cancer Res. 2017;23:43–51. doi: 10.1158/1078-0432.CCR-16-0908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Aminkeng F, Bhavsar AP, Visscher H, Rassekh SR, Li Y, Lee JW, et al. A coding variant in RARG confers susceptibility to anthracycline-induced cardiotoxicity in childhood cancer. Nat Genet. 2015;47:1079–1084. doi: 10.1038/ng.3374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Visscher H, Ross CJ, Rassekh SR, Sandor GS, Caron HN, van Dalen EC, et al. Validation of variants in SLC28A3 and UGT1A6 as genetic markers predictive of anthracycline-induced cardiotoxicity in children. Pediatr Blood Cancer. 2013;60:1375–1381. doi: 10.1002/pbc.24505. [DOI] [PubMed] [Google Scholar]
- 38.Visscher H, Ross CJ, Rassekh SR, Barhdadi A, Dubé MP, Al-Saloos H, et al. Pharmacogenomic prediction of anthracycline-induced cardiotoxicity in children. J Clin Oncol. 2012;30:1422–1428. doi: 10.1200/JCO.2010.34.3467. [DOI] [PubMed] [Google Scholar]
- 39.Wang X, Liu W, Sun CL, Armenian SH, Hakonarson H, Hageman L, et al. Hyaluronan synthase 3 variant and anthracycline-related cardiomyopathy: a report from the children’s oncology group. J Clin Oncol. 2014;32:647–653. doi: 10.1200/JCO.2013.50.3557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Vaitiekus D, Muckiene G, Vaitiekiene A, Sereikaite L, Inciuraite R, Insodaite R, et al. HFE gene variants’ impact on anthracycline-based chemotherapy-induced subclinical cardiotoxicity. Cardiovasc Toxicol. 2021;21:59–66. doi: 10.1007/s12012-020-09595-1. [DOI] [PubMed] [Google Scholar]
- 41.Wang X, Sun CL, Quiñones-Lombraña A, Singh P, Landier W, Hageman L, et al. CELF4 variant and anthracycline-related cardiomyopathy: a Children’s Oncology Group genome-wide association study. J Clin Oncol. 2016;34:863–870. doi: 10.1200/JCO.2015.63.4550. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Garcia-Pavia P, Kim Y, Restrepo-Cordoba MA, Lunde IG, Wakimoto H, Smith AM, et al. Genetic variants associated with cancer therapy-induced cardiomyopathy. Circulation. 2019;140:31–41. doi: 10.1161/CIRCULATIONAHA.118.037934. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Wojnowski L, Kulle B, Schirmer M, Schlüter G, Schmidt A, Rosenberger A, et al. NAD(P)H oxidase and multidrug resistance protein genetic polymorphisms are associated with doxorubicin-induced cardiotoxicity. Circulation. 2005;112:3754–3762. doi: 10.1161/CIRCULATIONAHA.105.576850. [DOI] [PubMed] [Google Scholar]
- 44.Suter TM, Procter M, van Veldhuisen DJ, Muscholl M, Bergh J, Carlomagno C, et al. Trastuzumab-associated cardiac adverse effects in the herceptin adjuvant trial. J Clin Oncol. 2007;25:3859–3865. doi: 10.1200/JCO.2006.09.1611. [DOI] [PubMed] [Google Scholar]
- 45.Romond EH, Jeong JH, Rastogi P, Swain SM, Geyer CE, Jr, Ewer MS, et al. Seven-year follow-up assessment of cardiac function in NSABP B-31, a randomized trial comparing doxorubicin and cyclophosphamide followed by paclitaxel (ACP) with ACP plus trastuzumab as adjuvant therapy for patients with node-positive, human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol. 2012;30:3792–3799. doi: 10.1200/JCO.2011.40.0010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE, Jr, Davidson NE, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 2005;353:1673–1684. doi: 10.1056/NEJMoa052122. [DOI] [PubMed] [Google Scholar]
- 47.Perez EA, Suman VJ, Davidson NE, Sledge GW, Kaufman PA, Hudis CA, et al. Cardiac safety analysis of doxorubicin and cyclophosphamide followed by paclitaxel with or without trastuzumab in the North Central Cancer Treatment Group N9831 adjuvant breast cancer trial. J Clin Oncol. 2008;26:1231–1238. doi: 10.1200/JCO.2007.13.5467. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Mackey JR, Clemons M, Côté MA, Delgado D, Dent S, Paterson A, et al. Cardiac management during adjuvant trastuzumab therapy: recommendations of the Canadian Trastuzumab Working Group. Curr Oncol. 2008;15:24–35. doi: 10.3747/co.2008.199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Cameron D, Piccart-Gebhart MJ, Gelber RD, Procter M, Goldhirsch A, de Azambuja E, et al. 11 years’ follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive early breast cancer: final analysis of the HERceptin Adjuvant (HERA) trial. Lancet. 2017;389:1195–1205. doi: 10.1016/S0140-6736(16)32616-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Yoon HJ, Kim KH, Kim HY, Park H, Cho JY, Hong YJ, et al. Impacts of non-recovery of trastuzumab-induced cardiomyopathy on clinical outcomes in patients with breast cancer. Clin Res Cardiol. 2019;108:892–900. doi: 10.1007/s00392-019-01417-x. [DOI] [PubMed] [Google Scholar]
- 51.Ewer MS, Lippman SM. Type II chemotherapy-related cardiac dysfunction: time to recognize a new entity. J Clin Oncol. 2005;23:2900–2902. doi: 10.1200/JCO.2005.05.827. [DOI] [PubMed] [Google Scholar]
- 52.Swain SM, Whaley FS, Ewer MS. Congestive heart failure in patients treated with doxorubicin: a retrospective analysis of three trials. Cancer. 2003;97:2869–2879. doi: 10.1002/cncr.11407. [DOI] [PubMed] [Google Scholar]
- 53.Chatterjee K, Zhang J, Honbo N, Karliner JS. Doxorubicin cardiomyopathy. Cardiology. 2010;115:155–162. doi: 10.1159/000265166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zafar A, Drobni ZD, Mosarla R, Alvi RM, Lei M, Lou UY, et al. The incidence, risk factors, and outcomes with 5-fluorouracil-associated coronary vasospasm. JACC CardioOncol. 2021;3:101–109. doi: 10.1016/j.jaccao.2020.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Sara JD, Kaur J, Khodadadi R, Rehman M, Lobo R, Chakrabarti S, et al. 5-fluorouracil and cardiotoxicity: a review. Ther Adv Med Oncol. 2018;10:1758835918780140. doi: 10.1177/1758835918780140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Eskilsson J, Albertsson M. Failure of preventing 5-fluorouracil cardiotoxicity by prophylactic treatment with verapamil. Acta Oncol. 1990;29:1001–1003. doi: 10.3109/02841869009091790. [DOI] [PubMed] [Google Scholar]
- 57.Clasen SC, Ky B, O’Quinn R, Giantonio B, Teitelbaum U, Carver JR. Fluoropyrimidine-induced cardiac toxicity: challenging the current paradigm. J Gastrointest Oncol. 2017;8:970–979. doi: 10.21037/jgo.2017.09.07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Saif MW, Shah MM, Shah AR. Fluoropyrimidine-associated cardiotoxicity: revisited. Expert Opin Drug Saf. 2009;8:191–202. doi: 10.1517/14740330902733961. [DOI] [PubMed] [Google Scholar]
- 59.Südhoff T, Enderle MD, Pahlke M, Petz C, Teschendorf C, Graeven U, et al. 5-Fluorouracil induces arterial vasocontractions. Ann Oncol. 2004;15:661–664. doi: 10.1093/annonc/mdh150. [DOI] [PubMed] [Google Scholar]
- 60.Chong JH, Ghosh AK. Coronary artery vasospasm induced by 5-fluorouracil: proposed mechanisms, existing management options and future directions. Interv Cardiol. 2019;14:89–94. doi: 10.15420/icr.2019.12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Khouri MG, Douglas PS, Mackey JR, Martin M, Scott JM, Scherrer-Crosbie M, et al. Cancer therapy-induced cardiac toxicity in early breast cancer: addressing the unresolved issues. Circulation. 2012;126:2749–2763. doi: 10.1161/CIRCULATIONAHA.112.100560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Vaduganathan M, Hirji SA, Qamar A, Bajaj N, Gupta A, Zaha V, et al. Efficacy of neurohormonal therapies in preventing cardiotoxicity in patients with cancer undergoing chemotherapy. JACC CardioOncol. 2019;1:54–65. doi: 10.1016/j.jaccao.2019.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Vuong JT, Stein-Merlob AF, Cheng RK, Yang EH. Novel therapeutics for anthracycline induced cardiotoxicity. Front Cardiovasc Med. 2022;9:863314. doi: 10.3389/fcvm.2022.863314. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Brown SA, Okwuosa TM, Barac A, Volgman AS. The role of angiotensin-converting enzyme inhibitors and β-blockers in primary prevention of cardiac dysfunction in breast cancer patients. J Am Heart Assoc. 2020;9:e015327. doi: 10.1161/JAHA.119.015327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lewinter C, Nielsen TH, Edfors LR, Linde C, Bland JM, LeWinter M, et al. A systematic review and meta-analysis of beta-blockers and renin-angiotensin system inhibitors for preventing left ventricular dysfunction due to anthracyclines or trastuzumab in patients with breast cancer. Eur Heart J. 2022;43:2562–2569. doi: 10.1093/eurheartj/ehab843. [DOI] [PubMed] [Google Scholar]
- 66.Alizadehasl A, Ghadimi N, Kaveh S, Maleki M, Ghavamzadeh A, Noohi F, et al. Prevention of anthracycline-induced cardiotoxicity: a systematic review and network meta-analysis. Int J Clin Pharm. 2021;43:25–34. doi: 10.1007/s11096-020-01146-6. [DOI] [PubMed] [Google Scholar]
- 67.Fang K, Zhang Y, Liu W, He C. Effects of angiotensin-converting enzyme inhibitor/angiotensin receptor blocker use on cancer therapy-related cardiac dysfunction: a meta-analysis of randomized controlled trials. Heart Fail Rev. 2021;26:101–109. doi: 10.1007/s10741-019-09906-x. [DOI] [PubMed] [Google Scholar]
- 68.Hwang HJ, Lee TG. Impact on clinical outcomes of renin-angiotensin system inhibitors against doxorubicin-related toxicity in patients with breast cancer and hypertension: a nationwide cohort study in South Korea. PLoS One. 2023;18:e0294649. doi: 10.1371/journal.pone.0294649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Hwang HJ, Han SA. Differential cardiovascular outcomes of each antihypertensive drug class in patients with hypertension and breast cancer undergoing doxorubicin-containing chemotherapy. J Breast Cancer. 2023;26:492–503. doi: 10.4048/jbc.2023.26.e34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Williams B, Mancia G, Spiering W, Agabiti Rosei E, Azizi M, Burnier M, et al. 2018 ESC/ESH Guidelines for the management of arterial hypertension. Eur Heart J. 2018;39:3021–3104. doi: 10.1093/eurheartj/ehy339. [DOI] [PubMed] [Google Scholar]
- 71.Mehlum MH, Liestøl K, Kjeldsen SE, Wyller TB, Julius S, Rothwell PM, et al. Blood pressure-lowering profiles and clinical effects of angiotensin receptor blockers versus calcium channel blockers. Hypertension. 2020;75:1584–1592. doi: 10.1161/HYPERTENSIONAHA.119.14443. [DOI] [PubMed] [Google Scholar]
- 72.Riad A, Bien S, Westermann D, Becher PM, Loya K, Landmesser U, et al. Pretreatment with statin attenuates the cardiotoxicity of doxorubicin in mice. Cancer Res. 2009;69:695–699. doi: 10.1158/0008-5472.CAN-08-3076. [DOI] [PubMed] [Google Scholar]
- 73.Acar Z, Kale A, Turgut M, Demircan S, Durna K, Demir S, et al. Efficiency of atorvastatin in the protection of anthracycline-induced cardiomyopathy. J Am Coll Cardiol. 2011;58:988–989. doi: 10.1016/j.jacc.2011.05.025. [DOI] [PubMed] [Google Scholar]
- 74.Seicean S, Seicean A, Plana JC, Budd GT, Marwick TH. Effect of statin therapy on the risk for incident heart failure in patients with breast cancer receiving anthracycline chemotherapy: an observational clinical cohort study. J Am Coll Cardiol. 2012;60:2384–2390. doi: 10.1016/j.jacc.2012.07.067. [DOI] [PubMed] [Google Scholar]
- 75.Abdel-Qadir H, Bobrowski D, Zhou L, Austin PC, Calvillo-Argüelles O, Amir E, 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;10:e018393. doi: 10.1161/JAHA.119.018393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Hundley WG, D’Agostino R, Jr, Crotts T, Craver K, Hackney MH, Jordan JH, et al. Statins and left ventricular ejection fraction following doxorubicin treatment. J NEJM Evid. 2022;1:10.1056/evidoa2200097. doi: 10.1056/evidoa2200097. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Neilan TG, Quinaglia T, Onoue T, Mahmood SS, Drobni ZD, Gilman HK, et al. Atorvastatin for anthracycline-associated cardiac dysfunction: the STOP-CA randomized clinical trial. JAMA. 2023;330:528–536. doi: 10.1001/jama.2023.11887. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Pedicino D, Patrono C. Adverse effects of anthracyclines: does atorvastatin STOP-CArdiotoxicity? Eur Heart J. 2023;44:4506–4507. doi: 10.1093/eurheartj/ehad611. [DOI] [PubMed] [Google Scholar]
- 79.Xu C, Wang W, Zhong J, Lei F, Xu N, Zhang Y, et al. Canagliflozin exerts anti-inflammatory effects by inhibiting intracellular glucose metabolism and promoting autophagy in immune cells. Biochem Pharmacol. 2018;152:45–59. doi: 10.1016/j.bcp.2018.03.013. [DOI] [PubMed] [Google Scholar]
- 80.Sardu C, Gatta G, Pieretti G, Onofrio N, Balestrieri ML, Scisciola L, et al. SGLT2 breast expression could affect the cardiovascular performance in pre-menopausal women with fatty vs. non fatty breast via over-inflammation and sirtuins’ down regulation. Eur J Intern Med. 2023;113:57–68. doi: 10.1016/j.ejim.2023.04.012. [DOI] [PubMed] [Google Scholar]
- 81.Salvatore T, Caturano A, Galiero R, Di Martino A, Albanese G, Vetrano E, et al. Cardiovascular benefits from gliflozins: effects on endothelial function. Biomedicines. 2021;9:1356. doi: 10.3390/biomedicines9101356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Wiviott SD, Raz I, Bonaca MP, Mosenzon O, Kato ET, Cahn A, et al. Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2019;380:347–357. doi: 10.1056/NEJMoa1812389. [DOI] [PubMed] [Google Scholar]
- 83.McMurray JJV, Solomon SD, Inzucchi SE, Køber L, Kosiborod MN, Martinez FA, et al. Dapagliflozin in patients with heart failure and reduced ejection fraction. N Engl J Med. 2019;381:1995–2008. doi: 10.1056/NEJMoa1911303. [DOI] [PubMed] [Google Scholar]
- 84.Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, et al. Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373:2117–2128. doi: 10.1056/NEJMoa1504720. [DOI] [PubMed] [Google Scholar]
- 85.Packer M, Anker SD, Butler J, Filippatos G, Pocock SJ, Carson P, et al. Cardiovascular and renal outcomes with empagliflozin in heart failure. N Engl J Med. 2020;383:1413–1424. doi: 10.1056/NEJMoa2022190. [DOI] [PubMed] [Google Scholar]
- 86.Gongora CA, Drobni ZD, Quinaglia Araujo Costa Silva T, Zafar A, Gong J, Zlotoff DA, et al. Sodium-glucose co-transporter-2 inhibitors and cardiac outcomes among patients treated with anthracyclines. JACC Heart Fail. 2022;10:559–567. doi: 10.1016/j.jchf.2022.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Hwang HJ, Kim M, Jun JE, Yon DK. Sodium-glucose cotransporter-2 inhibitors improve clinical outcomes in patients with type 2 diabetes mellitus undergoing anthracycline-containing chemotherapy: an emulated target trial using nationwide cohort data in South Korea. Sci Rep. 2023;13:21756. doi: 10.1038/s41598-023-48678-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Abdel-Qadir H, Carrasco R, Austin PC, Chen Y, Zhou L, Fang J, et al. The association of sodium-glucose cotransporter 2 inhibitors with cardiovascular outcomes in anthracycline-treated patients with cancer. JACC CardioOncol. 2023;5:318–328. doi: 10.1016/j.jaccao.2023.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Zhou J, Zhu J, Yu SJ, Ma HL, Chen J, Ding XF, et al. Sodium-glucose co-transporter-2 (SGLT-2) inhibition reduces glucose uptake to induce breast cancer cell growth arrest through AMPK/mTOR pathway. Biomed Pharmacother. 2020;132:110821. doi: 10.1016/j.biopha.2020.110821. [DOI] [PubMed] [Google Scholar]
- 90.Villani LA, Smith BK, Marcinko K, Ford RJ, Broadfield LA, Green AE, et al. The diabetes medication canagliflozin reduces cancer cell proliferation by inhibiting mitochondrial complex-I supported respiration. Mol Metab. 2016;5:1048–1056. doi: 10.1016/j.molmet.2016.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Shiba K, Tsuchiya K, Komiya C, Miyachi Y, Mori K, Shimazu N, et al. Canagliflozin, an SGLT2 inhibitor, attenuates the development of hepatocellular carcinoma in a mouse model of human NASH. Sci Rep. 2018;8:2362. doi: 10.1038/s41598-018-19658-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Nasiri AR, Rodrigues MR, Li Z, Leitner BP, Perry RJ. SGLT2 inhibition slows tumor growth in mice by reversing hyperinsulinemia. Cancer Metab. 2019;7:10. doi: 10.1186/s40170-019-0203-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.van Dalen EC, Caron HN, Dickinson HO, Kremer LC. Cardioprotective interventions for cancer patients receiving anthracyclines. Cochrane Database Syst Rev. 2011;2011:CD003917. doi: 10.1002/14651858.CD003917.pub3. [DOI] [PubMed] [Google Scholar]
- 94.Cvetković RS, Scott LJ. Dexrazoxane : a review of its use for cardioprotection during anthracycline chemotherapy. Drugs. 2005;65:1005–1024. doi: 10.2165/00003495-200565070-00008. [DOI] [PubMed] [Google Scholar]
- 95.Seymour L, Bramwell V, Moran LA The Provincial Systemic Treatment Disease Site Group. Use of dexrazoxane as a cardioprotectant in patients receiving doxorubicin or epirubicin chemotherapy for the treatment of cancer. Cancer Prev Control. 1999;3:145–159. [PubMed] [Google Scholar]
- 96.Nunes Filho P, Albuquerque C, Pilon Capella M, Debiasi M. Immune checkpoint inhibitors in breast cancer: a narrative review. Oncol Ther. 2023;11:171–183. doi: 10.1007/s40487-023-00224-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Schneider BJ, Naidoo J, Santomasso BD, Lacchetti C, Adkins S, Anadkat M, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: ASCO guideline update. J Clin Oncol. 2021;39:4073–4126. doi: 10.1200/JCO.21.01440. [DOI] [PubMed] [Google Scholar]
- 98.Tan S, Day D, Nicholls SJ, Segelov E. Immune checkpoint inhibitor therapy in oncology: current uses and future directions: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol. 2022;4:579–597. doi: 10.1016/j.jaccao.2022.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Dolladille C, Akroun J, Morice PM, Dompmartin A, Ezine E, Sassier M, et al. Cardiovascular immunotoxicities associated with immune checkpoint inhibitors: a safety meta-analysis. Eur Heart J. 2021;42:4964–4977. doi: 10.1093/eurheartj/ehab618. [DOI] [PubMed] [Google Scholar]
- 100.Johnson DB, Balko JM, Compton ML, Chalkias S, Gorham J, Xu Y, et al. Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med. 2016;375:1749–1755. doi: 10.1056/NEJMoa1609214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Laenens D, Yu Y, Santens B, Jacobs J, Beuselinck B, Bechter O, et al. Incidence of cardiovascular events in patients treated with immune checkpoint inhibitors. J Clin Oncol. 2022;40:3430–3438. doi: 10.1200/JCO.21.01808. [DOI] [PubMed] [Google Scholar]
- 102.Soares LR, Vilbert M, Rosa VD, Oliveira JL, Deus MM, Freitas-Junior R. Incidence of interstitial lung disease and cardiotoxicity with trastuzumab deruxtecan in breast cancer patients: a systematic review and single-arm meta-analysis. ESMO Open. 2023;8:101613. doi: 10.1016/j.esmoop.2023.101613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Waliany S, Caswell-Jin J, Riaz F, Myall N, Zhu H, Witteles RM, et al. Pharmacovigilance analysis of heart failure associated with anti-HER2 monotherapies and combination regimens for cancer. JACC CardioOncol. 2023;5:85–98. doi: 10.1016/j.jaccao.2022.09.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Darby SC, McGale P, Taylor CW, Peto R. Long-term mortality from heart disease and lung cancer after radiotherapy for early breast cancer: prospective cohort study of about 300,000 women in US SEER cancer registries. Lancet Oncol. 2005;6:557–565. doi: 10.1016/S1470-2045(05)70251-5. [DOI] [PubMed] [Google Scholar]
- 105.Ramroth J, Shakir R, Darby SC, Cutter DJ, Kuan V. Cardiovascular disease incidence rates: a study using routinely collected health data. Cardiooncology. 2023;9:41. doi: 10.1186/s40959-023-00189-8. [DOI] [PMC free article] [PubMed] [Google Scholar]



