Open Access

Cardiotoxicity of anthracycline‑free targeted oncological therapies in HER2‑positive breast cancer (Review)

  • Authors:
    • Jingyuan Guan
    • Mei Zhang
  • View Affiliations

  • Published online on: December 8, 2020     https://doi.org/10.3892/ol.2020.12361
  • Article Number: 100
  • Copyright: © Guan et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Anthracycline drugs are considered to be pivotal drugs in numerous chemotherapy regimens for breast cancer. However, the cardiotoxicity associated with the treatment is an important issue to be addressed. With the emergence of increasingly diverse antitumor drugs, anthracycline‑free therapies are able to reduce the cardiotoxicity caused by anthracycline drugs while ensuring that a therapeutic effect is achieved. In the present review, anthracycline‑free oncological therapy regimens for the treatment of patients with human epidermal growth factor receptor 2 (HER2)‑positive breast cancer and the associated cardiovascular toxicity are discussed, as well as some monitoring strategies. It is recommended that patients with HER2‑positive breast cancer patients should receive adjuvant chemotherapy with single or dual‑targeted therapy, with or without endocrine therapy according to the hormone receptor status determined by immunohistochemical examination. The main side effects of targeted therapy include cardiac dysfunction, hypertension and arrhythmia. According to individual risk stratification, it is recommended that patients should be periodically monitored using echocardiography, electrocardiography and serum markers, to enable the timely detection of the cardiovascular adverse reactions associated with tumor treatment, thereby preventing the morbidity and mortality caused by the cardiotoxicity of these drugs.

Introduction

Anthracycline-based chemotherapy regimens like doxorubicin and cyclophosphamide followed by docetaxel (AC-T) or AC-T + trastuzumab (AC-TH) play a prominent role in the treatment of breast cancer, and contribute to the global 5- and 10-year survival rates of 73 and 61%, respectively, for female patients (1). Unfortunately, the administration of these regimens has been limited due to the increased risk of irreversible cardiotoxicity associated with anthracyclines, with cardiac dysfunction and arrhythmia as the main adverse reactions. The long-term cardiotoxic effects of chemotherapy (2), which may lead to increased cardiovascular morbidity and mortality, account for the majority of deaths of patients with cancer from non-tumor causes (3).

For patients who have metastatic breast cancer, are older or have other underlying diseases, anthracycline drugs are not suitable because of their high toxicity. Therefore, the identification of alternatives to anthracyclines that are equally effective is important. In the phase III Breast Cancer International Research Group 006 study on patients with human epidermal growth factor receptor 2 (HER2)-positive breast cancer with axillary lymph node involvement or high-risk negative cancer, the cardiotoxicity in patients treated with docetaxel, carboplatin and trastuzumab (TCH) was lower than that in patients treated with AC-T or AC-TH. Both disease-free survival (DFS) and overall survival were significantly improved in the patients treated with TCH, the cardiotoxicity rates of which were also reduced compared with those of the other treatments; the rates of congestive heart failure (CHF) events in the TCH, AC-TH, and AC-T groups were 0.4, 2.0 and 0.7%, respectively (4,5). From other trials in which anthracycline-containing therapy was compared with anthracycline-free therapy (Table I), it is evident that the cardiotoxicity of anthracycline-free regimens is lower than that of anthracycline-containing regimens, but is not negligible.

Table I.

Clinical trials comparing anthracycline-based regimens with anthracycline-free regimens.

Table I.

Clinical trials comparing anthracycline-based regimens with anthracycline-free regimens.

First author, yearRegimenTrialPhaseNo. of patientsStudy designaCancer typePrimary endpointCardiac events(Refs.)
Slamon, 2011AnthracyclineBCIRG 006III1,073ACb4 → Tb4HER2+, invasive, high-risk, node-negative or node-positive adenocarcinomaDFSNYHA III/IV CHF, 0.7%(5)
1,074ACb4 → Tb4+H NYHA III/IV CHF, 2.0%
Non-anthracycline 1,075TCHb6 → H NYHA III/IV CHF, 0.4% (P<0.001)
Schneeweiss, 2011AnthracyclineTRYPHAENAII73 FEC+H+Pb3 → T+H+Pb3HER2+ breast cancer (primary tumor >2 cm)DFSLVEF declines ≥10% from baseline to <50%, 11.1%;   LVSD 2.8%(72)
75FECb3 → T+H+Pb3 LVEF declines ≥10% from baseline to <50%, 16.0%; LVSD, 4.0%
Non-anthracycline 77TCH+Pb6 LVEF declines ≥10% from baseline to <50%, 11.8%; LVSD, 5.4%
Van Ramshorst, 2018AnthracyclineTRAIN2III219 FEC+H+Pb3 → T+Cb+H+Pb6HER2+ breast cancerpCRSymptomatic LVSD, 1 (1%); hypertension, 5 (2%); LVEF decreased, 4 (2%)(7)
Non-anthracycline 219 T+Cb+H+Pb9 Symptomatic LVSD, 0; hypertension, 1 (<1%); LVEF decreased, 1 (<1%)

{ label (or @symbol) needed for fn[@id='tfn1-ol-0-0-12361'] } BCIRG, Breast Cancer International Research Group 006; A, doxorubicin; C, cyclophosphamide; T docetaxel; H, trastuzumab (Herceptin); FEC, 5-fluorouracil + epirubicin + cyclophosphamide; P, pertuzumab; Cb, carboplatin; HER2, human epidermal growth factor receptor 2; DFS, disease-free survival; pCR, pathological complete response; NYHA, New York Heart Association; CHF, congestive heart failure; LVEF, left ventricular ejection fraction; LVSD, left ventricular systolic dysfunction. Study design

a in this column

b n indicates n cycles.

The present review describes the current epidemiological evidence on the cardiotoxicity associated with anthracycline-free therapy, particularly targeted drugs, in patients with HER2-positive breast cancer, as well as the clinical manifestations, possible mechanisms and achievable clinical monitoring recommendations for the therapies. Since there are few retrospective clinical studies on the treatment of the cardiovascular complications associated with cancer therapy, the discussion of clinical monitoring in this review represents the opinions of the authors based on currently available information.

Epidemiology

At present, in patients with HER2-positive breast cancer, if no contraindications are present, the use of anti-HER2 targeted drugs is advocated. The National Comprehensive Cancer Network guidelines recommended that patients with HER2-positive breast cancer receive adjuvant chemotherapy with single or dual targeted therapy, with or without endocrine therapy according to hormone receptor status determined by immunohistochemical examination (6). However, evidence of cardiovascular toxicity secondary to neoadjuvant or adjuvant therapy in HER2-positive breast cancer has emerged in a trial involving HER2 blockade (7). With regard to the molecular mechanism, the activity of ErbB proteins in normal cells relies on heterodimer formation; the heterodimer formed between HER2 (also known as ErbB2) and ErbB3 induces the proliferation in breast tumor cells, and the heterodimer formed between HER2 and ErbB4 is key to the proliferation and contractility of cardiac myocytes (8). The anti-HER2 agent trastuzumab inhibits HER2-induced intracellular signaling and marks HER2-positive cells for antibody-dependent cellular cytotoxicity by binding to the extracellular structure of HER2 in cancer cells as well as cardiomyocytes (9). This leads to significantly improved DFS in women with HER2-positive breast cancer but also increases the risk of severe cardiotoxicity (10).

A retrospective study showed that 13% of patients with metastatic breast cancer who were treated with trastuzumab plus paclitaxel had cardiac dysfunction (11). In other studies, the combination of trastuzumab and pertuzumab significantly increased progression-free survival and pathological complete response (pCR) in patients with breast cancer without affecting the incidence of cardiotoxicity (1215). In the NeoSphere study (13), 45.8% of the patients treated with neoadjuvant trastuzumab and pertuzumab combined with chemotherapy achieved a pCR, compared with 29.0% of the patients treated with chemotherapy combined with single-agent trastuzumab. Among patients treated with pertuzumab, trastuzumab, and docetaxel, only 3/107 patients (3%) showed a decline of 10–15% in the left ventricular ejection fraction (LVEF) from the baseline. In a 5-year analysis of the NeoSphere study, cases of left ventricular dysfunction of grade 3 or worse were followed up, and it was found that the majority of the asymptomatic LVEF reductions of ≥10% occurred during adjuvant trastuzumab therapy (16). A 2013 meta-analysis (15) of six trials in patients with breast cancer showed that the incidence of serious cardiovascular events in patients treated with dual anti-HER2 agents was similar to that of patients treated with anti-HER2 monotherapy, and the absolute incidence of each cardiac event was relatively low (CHF, 0.88%; LVEF decline 3.1%). This result is inconsistent with that of the PERTAIN trial, a randomized, phase II trial in which left ventricular dysfunction was observed in 3 (2.4%) patients in the pertuzumab plus trastuzumab arm but none in the trastuzumab arm (17). These conflicting results may be attributed to the different mechanisms by which pertuzumab and trastuzumab act. Pertuzumab binds to the extracellular domain II of HER2, which inhibits the formation of HER2 heterodimers and consequently affects activation of the HER2 signal pathway, while trastuzumab binds to the juxtamembrane region (domain IV) of HER2, inducing receptor dimerization and activation of the cytoplasmic kinase, which subsequently leads to autophosphorylation and initiation of downstream signaling events (1820).

Trastuzumab emtansine (T-DM1) is an antibody-drug conjugate of trastuzumab that targets the microtubule inhibitor emtansine to HER2-overexpressing tumor cells. T-DM1 decreases the risk of recurrence of invasive breast cancer to a greater extent than trastuzumab alone. However, the toxicity of T-DM1 is also higher than that of trastuzumab (21). A pooled analysis of T-DM1 trials showed that T-DM1-mediated cardiotoxicity occurred early, with approximately half of the cardiac events occurring within the first 100 days of treatment, and demonstrated that older people tended to be more vulnerable, with a 5% increase of cardiac risk per 1-year increase in age (22). In the KATHERINE trial, the incidence of hypertension in the patients treated with T-DM1 was 2.0% compared with 1.2% in those treated with trastuzumab (21).

With the widespread administration of trastuzumab, drug resistance has been observed in patients with metastatic breast cancer. Lapatinib, which inhibits both HER2 tyrosine kinase and the epidermal growth factor receptor (EGFR), was approved in 2007 for use in combination with capecitabine for the treatment of HER2-positive metastatic breast cancer, with EGFR serving as an alternative target to the HER2 pathway (23). Lapatinib inhibits signaling by binding to Her2-ErbB3 and EGFR complexes (24). A 2017 meta-analysis (25) of 26 lapatinib-containing trials in patients with breast cancer reported that the overall incidence of cardiovascular events was 3.0% (95% CI: 1.50–6.10%), including left ventricular dysfunction (1.7%) and LVEF decline (1.8%). Additionally, as hypertension is the most common manifestation of cardiovascular toxicity it also merits attention. The risk ratio of all-grade hypertension of vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) was reported to be 3.43 (95% CI: 2.95–3.99; P<0.00001) (15,26). Multiple reports of QT prolongation in patients treated with sunitinib, but not lapatinib, have emerged, which in some cases have been associated with sudden death (27). Tucatinib, an oral, potent, reversible HER2-specific tyrosine kinase inhibitor (TKI) has been used in combination with T-DM1 for the treatment of advanced HER2-positive metastatic breast cancer. In a phase 1b clinical trial, grade 1 heart failure (HF) occurred in 2/50 patients (4%) who received T-DM1 (3.6 mg/kg intravenous infusion every 21 days) and tucatinib (300 mg orally twice daily for 21 days), and one of the patients discontinued the study because of a consistent decline in LVEF (28). TKIs have been shown to inhibit the EGFR/Ras/Raf/MEK/ERK signaling pathway in tumor cells and cardiomyocytes, whereas the Raf/MEK/ERK cascade protects cardiomyocytes from stress-induced damage (29). A recent study revealed that ERK-dimerization inhibitory (EDI) peptide interferes with ERK dimerization and subsequently prevents ERKT188 phosphorylation (30). TUNEL assays and measurements of the mitochondrial membrane potential to assess the oxidative stress response in vitro and in vivo in mice demonstrated the non-cardiotoxicity of EDI. While the membrane potential was depolarized in the presence of all MEK inhibitors tested, EDI protected it from collapse. This intervention has also been shown to effectively inhibit cancer cell proliferation without cardiotoxicity (30). This finding provides new possibilities for the modification of TKIs to reduce or even eliminate cardiotoxicity.

Surveillance strategies

Strongly elevated risks of cardiac events have been observed following tumor therapy. Although the benefits of systemic treatments usually outweigh the risks of cardiac events, the need to support ongoing efforts to evaluate preventative strategies should be emphasized. A position paper prepared by the European Society of Cardiology (ESC) recommends that a patient's cardiac condition should be evaluated once per cycle (31). The diagnostic tools proposed for the detection of cardiotoxicity comprise electrocardiograms (ECGs), echocardiography, cardiac magnetic resonance imaging (CMRI) and cardiac biomarkers, including troponin I (TnI), high-sensitivity (hs)-TnI, brain natriuretic peptide (BNP) and N-terminal pro-BNP (NT-proBNP) (31). Based on the evaluation of risk factors for chemotherapy-related cardiotoxicity (CTRCD), there is a growing consensus that the control of relevant risk factors may enable some cardiac events to be avoided (Table II).

Table II.

Potential cardiac risk factors.

Table II.

Potential cardiac risk factors.

CategoriesRisk factors(Refs.)
Basic factorsRace(42,73)
  Caucasian
  African American
Female sex(74)
Age ≥65 years or perimenopausal(74,75)
Tobacco use(74)
ConcomitantObesity (BMI ≥30 kg/m2)(75)
diseaseDiabetes mellitus(74)
Cardiovascular disease
  Baseline LVEF, <55%(76)
  Heart failure(74)
  Hypertension(74)
  Coronary artery disease(75)
  Peripheral artery disease(75)
  Hyperlipidemia(74)
ConcomitantIrradiation of the chest(74,77)
therapyCumulative anthracycline exposure(74)

[i] BMI, body mass index; LVEF, left ventricular ejection fraction.

ECG

Diagnostic testing with ECG can be nonspecific even if the results are abnormal; however, assessment of the condition of the heart remains essential. The ESC guidelines recommended that each patient should undergo a standard 12-lead ECG examination every treatment cycle, despite studies showing that targeted drug therapy without anthracyclines reduces the risk of arrhythmias, namely tachycardia and bradycardia, and prolongs QT intervals by 40% compared with anthracyclines (31,32).

There is no evidence of arrhythmia being caused by targeted anti-HER-2 drugs such as trastuzumab and pertuzumab. However, antimicrotubule agents such as paclitaxel can cause arrhythmias, among which asymptomatic bradycardia and first-degree atrioventricular block are the most common types (33). If a patient has any previous risk factors for bradycardia, for example, there is evidence of abnormal cardiac conduction or the long-term use of negative inotropic drug, drugs that may cause arrhythmias should be used with caution and the patient's heart rate should be closely monitored during their use. ECG examination can be performed twice a week if necessary, and medication should be stopped immediately if symptoms or persistent conduction block occurs. In addition, a pacemaker may be implanted if necessary. By contrast, TKIs affect the QT interval, which is typically increased to 15 msec longer than the baseline value; the QT interval-prolonging effects of lapatinib and sunitinib are marked, with average increases of 23.4 and 22.4 msec, respectively, from baseline (34). QT interval prolongation tends to cause malignant arrhythmias, and may even lead to torsades de pointes. Adverse reactions can be graded according to the degree by which the QT interval is prolonged, and different treatments should be given accordingly (Table III). Additionally, it is suggested that electrolytes should be monitored periodically in patients who are highly vulnerable to QT interval prolongation (35).

Table III.

QTc prolongation grade and corresponding recommendations.

Table III.

QTc prolongation grade and corresponding recommendations.

QTc prolongation gradeDefinition Recommendationa
1Average QTc 450–480 msecIn patients with drug-induced LQTS, removal of the offending agent is indicated (I, A)
2Average QTc 481–500 msecIn patients with drug-induced LQTS, removal of the offending agent is indicated (I, A)
3Average QTc ≥501 msec; >60 msec change from baselineIn patients with drug-induced LQTS, removal of the offending agent is indicated (I, A)
4Torsade de pointes; polymorphic ventricular tachycardia; signs/symptoms of serious arrhythmiaIntravenous magnesium sulfate for patients who take QT-prolonging drugs and present with few episodes of torsades de pointes pacing in which the QT remains long (IIa, B); atrial or ventricular or isoproterenol (IIa, B); potassium ion repletion to 4.5–5 mmol/l (IIb, C)

a In the parentheses, the Roman numeral represents the classification of the recommendation, and the letter represents the level of evidence. National Cancer Institute's toxicity definitions for corrected QT interval prolongation (78) and specific treatment recommendations according to the American College of Cardiology/American Heart Association Task Force and the European Society of Cardiology (ACC/AHA/ESC) 2006 guidelines (79). LQTS, long QT syndrome.

Echocardiographic measurement

Echocardiography is the most important means of assessing cardiac function during cancer treatment because it is readily available and does not expose the patients to radiation. With regard to the definition of cardiotoxicity, according to guidelines and recommendations issued by the ESC, American Society of Echocardiography (ASE) and European Association of Cardiovascular Imaging, a >10% reduction in LVEF to a value below the lower limit of normal or a >15% relative reduction in global longitudinal strain (GLS) from baseline is suggestive of cardiotoxicity (31,36). Therefore, it is necessary for patients to undergo an echocardiographic assessment before starting cancer treatment. Patients with a baseline LVEF level of <50% are not suitable for chemotherapy due to their increased risk of cardiac events.

The most important indicator for the assessment of cardiac function is LVEF, which reflects the systolic function of the left ventricle, and can be evaluated using 2-dimensional (2D) and 3-dimensional (3D) echocardiographic acquisitions. The modified biplane Simpson's method is recommended by the ASE for the measurement of LVEF (37). In a study of patients undergoing chemotherapy for breast cancer, for whom comprehensive echocardiograms were performed at five time points by two investigators, 3D echocardiography (3DE) exhibited improved intra- and interobserver as well as test-retest repeatability compared with 2D methods (38), which indicates that 3D measurements can overcome the poor repeatability of echocardiographic measurements of LVEF to a certain extent. Similarly, a 2012 meta-analysis of 23 studies in which 1,638 echocardiograms were performed demonstrated that 3DE is more accurate than 2D methods for the measurement of left ventricular volume and LVEF (39).

Tissue Doppler-derived indices, including systolic (S′), early diastolic and late diastolic velocities, represent different functions of the heart. Tissue Doppler imaging (TDI) is widely recommended for evaluation of the diastolic function of the heart (40). In a study conducted by Fallah-Rad et al (41), 42 patients with breast cancer treated with adjuvant trastuzumab were evaluated using TDI. Although the peak global longitudinal and radial strain decreased, 10 patients demonstrated a reduction in lateral s' at 3 months after trastuzumab treatment, and these 10 patients all eventually developed trastuzumab-mediated cardiotoxicity. Retrospective studies have also shown that 20% of patients develop new or worsening diastolic dysfunction during breast cancer treatment (42,43). In the aforementioned studies, the diastolic dysfunction was evaluated by TDI indexes including mitral annular e'velocity (septal and lateral) and average E/e' ratio. Notably, a reduction in diastolic function was observed in patients treated with doxorubicin alone or doxorubicin followed by trastuzumab but not in those treated with trastuzumab alone (42). Early changes in diastolic function are closely associated with a subsequent reduction of ejection fraction. Therefore, the evaluation of diastolic function appears to be helpful to evaluate the prognosis of cardiac function.

Speckle-tracking echocardiography (STE) has emerged as a highly sensitive technique for the detection of myocardial dysfunction, and the increasing maturity of STE has prompted the increased use of GLS as a technique for the assessment of cardiac function (44). GLS is a reliable, effective, repeatable and less load-dependent technique for measuring left ventricular longitudinal deformation, which is measured by STE, superior to LVEF, in the diagnosis and prognosis of acute heart failure and acute myocardial infarction (45,46). Several studies have evaluated GLS in the detection of CTRCD. Laufer-Perl et al (47) evaluated 291 consecutive patients with breast cancer who received different types of cancer therapy and divided them into two groups, according to whether GLS was reduced or preserved. Observation of the patients over a median follow-up period of 2.9 months indicated that reduced GLS had the potential to identify patients with an increased risk for the development of cardiac dysfunction. Notably, the incidence and mortality rates for the patients with reduced GLS were significantly higher than those of the patients with preserved GLS. A systematic review of studies on cardiotoxicity during and after cancer chemotherapy indicated that an early decline of 10–11% in GLS can be a strong predictor of subsequent cardiotoxicity (48). Sawaya et al (49) similarly found a reduction in GLS (<19%) in all patients who developed cardiac dysfunction; although the radial and circumferential components of the strain decreased over the duration of the study, they were not predictive. Furthermore, a recent retrospective study by Santoro et al (50) highlighted the pivotal role of GLS in the prediction of cardiac outcomes after cancer therapy, suggesting that strain can be a parameter for guiding the initiation of cardioprotective strategies.

With improvements in the understanding of right ventricular (RV) function and measurement techniques, RV dysfunction secondary to chemotherapy has been identified as a factor that requires consideration. Evidence of impaired right-heart function first appeared in a single-case report by Bayer et al (51) in 2015. A 46-year-old patient with invasive ductal breast cancer developed right HF during treatment with trastuzumab after initial treatment with 5-fluorouracil + epirubicin + cyclophosphamide followed by docetaxel. The patient's heart function returned to normal after the discontinuation of the drug. Based on previous studies, it is speculated that the right heart is more sensitive to chemotherapy because its structure is thinner than that of the left heart and contains fewer myofibrils (52); therefore, its assessment may predict cardiac dysfunction that has not yet occurred in the left ventricle. For the measurement of right heart function, the guidelines from the British Society of Echocardiography are recommended; the main measurement indicators include RV ejection fraction, RV fractional area change, tricuspid annular plane systolic excursion, and RV longitudinal strain (53,54). However, further large scale studies are required to determine the significance of RV function monitoring in the prediction and prognosis of heart function in patients with breast cancer after treatment.

Biomarkers

Biomarkers accompanying myocardial injury identify patients at high risk of cardiotoxicity, and these patients often benefit from early treatment with, for example, angiotensin-converting enzyme inhibitors or b-blockers (31). It is expected that biomarkers may be able to predict future cardiotoxicity with high sensitivity. Cardiac troponins I and T (cTnI and cTnT) have been widely used as biomarkers of myocardial necrosis. Numerous retrospective studies have compared the concentrations of troponins before and after cancer treatment, especially in patients who developed cardiac events. In one study, an increase in cTnI following high-dose chemotherapy was found to be a strong predictor of poor cardiological outcome in patients with cancer (55). However, in another study, TnT, C-reactive protein and BNP did not change over a 2-year follow-up in patients with or without trastuzumab-mediated cardiotoxicity, while hs-cTnI proved to be a sensitive predictor of the development of cardiotoxicity (49). Therefore, hs-cTnI is a suitable biomarker for the screening of patients at high risk to determine whether more detailed tests are necessary. However, the high false-positive rate of hs-cTnI renders it unsuitable for diagnosis or for association with prognosis. In a study of 19 patients receiving chemotherapy with anthracyclines and trastuzumab, a significant increase in hs-cTnI was observed during the adjuvant trastuzumab therapy. In addition, the hs-cTNI level of patients whose left ventricular ejection fraction (≥) decreased by 5% was significantly higher compared with patients whose left ventricular ejection fraction (LVEF) decreased <5% (11.0±7.8 vs. 4.0±1.4 pg/ml; P<0.01) (56).

NT-proBNP is a hormone produced and released in response to ventricular wall stress, and has been identified as a biomarker for HF (57). Due to its value in the diagnosis of HF, a number of clinical trials have evaluated its use as a predictor of cardiotoxicity. A retrospective study of 71 patients with breast cancer who received anthracyclines demonstrated that a difference in the concentration of NT-proBNP between baseline and peak of >36% was able to predict left ventricular dysfunction (58). However, not all patients who developed cancer therapy-associated cardiotoxicity exhibited synchronous changes in NT-proBNP levels. Furthermore, no changes in NT-proBNP levels were observed in patients who received treatment only with trastuzumab regimens (41). According to currently available studies, it appears that only treatments containing anthracyclines induce changes in NT-proBNP (5962). The levels of NT-proBNP may increase rapidly within 24 h of chemotherapy and then fall back to normal levels prior to the next cycle (63). Therefore, the detection of NT-proBNP 24 h after each cycle of chemotherapy is recommended.

Soluble suppression of tumorigenicity 2 (sST2) has been indicated to be a valuable biomarker associated with cardiac remodeling in patients with HF (64). Several studies have confirmed that sST2 exhibits a positive correlation with the E/e' ratio, an echocardiographic indicator of left ventricular systolic function, in patients with breast cancer receiving chemotherapy and also those undergoing adjunctive radiotherapy (6567).

Other serum indicators have been shown to be valuable in the prediction of chemotherapeutic-associated cardiotoxicity. For example, high levels of placental growth factor and growth differentiation factor 15 were found to be associated with CTRCD in patients receiving trastuzumab therapy (61). In addition, myeloperoxidase exhibited a promising effect in the evaluation of the CTRCD risk of anthracyclines, both at baseline and throughout follow-up (68). Moreover, a study of the levels of certain microRNAs (miRNAs) in patients with breast cancer during and after anthracycline-containing chemotherapy revealed that the elevation of CHF-associated miRNAs, particularly miR-423-5p, was highly associated with the development of CTRCD (67). However, none of the aforementioned biomarkers have an appropriate balance of sensitivity and specificity, and more experiments are required to identify ideal biomarkers.

CMRI

CMRI is recommended when it is not possible to diagnose the cardiac condition with other tools or if the LVEF is on the borderline of normal (31). Compared with echocardiography, CMRI is more accurate and repeatable. It is especially suitable for the detection of diffuse myocardial fibrosis, but it is also limited by poor patient compliance (69,70). A study reviewed a cohort of adult survivors of childhood cancer who had been treated with chemotherapy, and high false-negative rates for an LVEF <50% were observed with echocardiography compared with CMRI (71). Based on these findings, comprehensive cardiac assessments should be considered for patients with an LVEF between 50 and 59% (71).

Conclusions

The emergence of targeted drugs and immunotherapy has had a major impact on cancer treatment, providing more options to eliminate the cardiotoxicity caused by chemotherapeutic drugs such as anthracyclines. However, targeted drugs such as trastuzumab have also caused reversible damage to the heart. The main manifestation is cardiac dysfunction, but cardiac arrest and acute myocardial infarction may also occur. Further coordination between oncologists and cardiologists is required to achieve a balance between tumor treatment and cardiac safety. A baseline cardiac assessment is recommended for all patients prior to chemotherapy, and surveillance strategies should be considered for individuals at high risk according to the tumor treatment strategy and the patient's previous medical history.

Although a variety of anthracycline-free treatment regimens have been proposed and used, it must be noted that most patients with breast cancer receive anthracycline-based tumor treatments. Considering the possible consequences, it is important to raise awareness of cardiotoxicity, particularly when anthracyclines are used in combination with anti-HER2 targeted drugs.

Numerous issues remain to be resolved. For example, definitions are required for the following: The manifestations of complications associated with non-anthracycline treatments and their potential pathophysiological mechanisms; early intervention and preventive measures for tumor treatment-induced cardiotoxicity; monitoring strategies and biomarkers that are able to detect early toxic reactions; the appropriate time at which treatment should be restarted after cardiotoxicity and whether the treatment measures require adjustment; and how best to comprehensively treat patients with cancer. Cardio-oncology is a booming field that requires the cooperation of oncologists and cardiologists. The development of this discipline will improve the survival rate, quality of life and prognosis of patients.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Not applicable.

Authors' contributions

JG and MZ analyzed the literature and wrote the manuscript. Both authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

HER2

human epidermal growth factor receptor 2

DFS

disease-free survival

pCR

pathological complete response

LVEF

left ventricular ejection fraction

EGFR

epidermal growth factor receptor

T-DM1

trastuzumab emtansine

TKI

tyrosine kinase inhibitor

EDI

ERK-dimerization inhibitory

ECG

electrocardiogram

CMRI

cardiac magnetic resonance imaging

ESC

European Society of Cardiology

ASE

American Society of Echocardiography

GLS

global longitudinal strain

2D

2-dimensional

3D

3-dimensional

3DE

3D echocardiography

TDI

tissue Doppler imaging

STE

speckle-tracking echocardiography

RV

right ventricular

NT-proBNP

N-terminal pro-brain natriuretic peptide

CTRCD

chemotherapy-related cardiotoxicity

HF

heart failure

sST2

soluble suppression of tumorigenicity 2

hs-cTnI

high-sensitivity cardiac troponin I

References

1 

Maajani K, Jalali A, Alipour S, Khodadost M, Tohidinik HR and Yazdani K: The global and regional survival rate of women with breast cancer: A systematic review and meta-analysis. Clin Breast Cancer. 19:165–177. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Shan K, Lincoff AM and Young JB: Anthracycline-induced cardiotoxicity. Ann Intern Med. 125:47–58. 1996. View Article : Google Scholar : PubMed/NCBI

3 

Vejpongsa P and Yeh E: Prevention of anthracycline-induced cardiotoxicity: Challenges and opportunities. J Am Coll Cardiol. 64:938–945. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Slamon DJ, Eiermann W, Robert NJ, Giermek J, Martin M, Mackey J, Chan A, Pinter T, Valero V, Falkson C, et al: Abstract S5-04: Ten year follow-up of BCIRG-006 comparing doxorubicin plus cyclophosphamide followed by docetaxel (AC→T) with doxorubicin plus cyclophosphamide followed by docetaxel and trastuzumab (AC→TH) with docetaxel, carboplatin and trastuzumab (TCH) in HER2+early breast cancer. Cancer Res. 76 (Suppl 4):S5-04-S5-04. 2016.

5 

Slamon D, Eiermann W, Robert N, Pienkowski T, Martin M, Press M, Mackey J, Glaspy J, Chan A, Pawlicki M, et al: Adjuvant trastuzumab in HER2-positive breast cancer. N Engl J Med. 365:1273–1283. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Gradishar WJ, Anderson BO, Abraham J, Aft R, Agnese D, Allison KH, Blair SL, Burstein HJ, Dang C, Elias AD, et al: Breast cancer, version 3.2020, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 18:452–478. 2020. View Article : Google Scholar : PubMed/NCBI

7 

van Ramshorst MS, van der Voort A, van Werkhoven ED, Mandjes IA, Kemper I, Dezentjé VO, Oving IM, Honkoop AH, Tick LW, van de Wouw AJ, et al: Neoadjuvant chemotherapy with or without anthracyclines in the presence of dual HER2 blockade for HER2-positive breast cancer (TRAIN-2): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 19:1630–1640. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Wadugu B and Kuehn B: The role of neuregulin/ErbB2/ErbB4 signaling in the heart with special focus on effects on cardiomyocyte proliferation. Am J Physiol Heart Circ Physiol. 302:H2139–H2147. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Rochette L, Guenancia C, Gudjoncik A, Hachet O, Zeller M, Cottin Y and Vergely C: Anthracyclines/trastuzumab: New aspects of cardiotoxicity and molecular mechanisms. Trends Pharmacol Sci. 36:326–348. 2015. View Article : Google Scholar : PubMed/NCBI

10 

Piccart-Gebhart MJ, Procter M, Leyland-Jones B, Goldhirsch A, Untch M, Smith I, Gianni L, Baselga J, Bell R, Jackisch C, et al: Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med. 353:1659–1672. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, et al: Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 344:783–792. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Swain SM, Kim SB, Cortés J, Ro J, Semiglazov V, Campone M, Ciruelos E, Ferrero JM, Schneeweiss A, Knott A, et al: Pertuzumab, trastuzumab, and docetaxel for HER2-positive metastatic breast cancer (CLEOPATRA study): Overall survival results from a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 14:461–471. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Gianni L, Pienkowski T, Im YH, Roman L, Tseng LM, Liu MC, Lluch A, Staroslawska E, de la Haba-Rodriguez J, Im SA, et al: Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): A randomised multicentre, open-label, phase 2 trial. Lancet Oncol. 13:25–32. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Fasching PA, Hartkopf AD, Gass P, Häberle L, Akpolat-Basci L, Hein A, Volz B, Taran FA, Nabieva N, Pott B, et al: Efficacy of neoadjuvant pertuzumab in addition to chemotherapy and trastuzumab in routine clinical treatment of patients with primary breast cancer: A multicentric analysis. Breast Cancer Res Treat. 173:319–328. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Valachis A, Nearchou A, Polyzos NP and Lind P: Cardiac toxicity in breast cancer patients treated with dual HER2 blockade. Int J Cancer. 133:2245–2252. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Gianni L, Pienkowski T, Im YH, Tseng LM, Liu MC, Lluch A, Starosławska E, de la Haba-Rodriguez J, Im SA, Pedrini JL, et al: 5-year analysis of neoadjuvant pertuzumab and trastuzumab in patients with locally advanced, inflammatory, or early-stage HER2-positive breast cancer (NeoSphere): A multicentre, open-label, phase 2 randomised trial. Lancet Oncol. 17:791–800. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Rimawi M, Ferrero J, de la Haba-Rodriguez J, Poole C, De Placido S, Osborne CK, Hegg R, Easton V, Wohlfarth C and Arpino G; PERTAIN Study Group, : First-line trastuzumab plus an aromatase inhibitor, with or without pertuzumab, in human epidermal growth factor receptor 2-positive and hormone receptor-positive metastatic or locally advanced breast cancer (PERTAIN): A randomized, open-label phase II trial. J Clin Oncol. 36:2826–2835. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Hubalek M, Brantner C and Marth C: Role of pertuzumab in the treatment of HER2-positive breast cancer. Breast cancer (Dove Medical Press). 4:65–73. 2012.PubMed/NCBI

19 

Cho HS, Mason K, Ramyar KX, Stanley AM, Gabelli SB, Denney DW Jr and Leahy DJ: Structure of the extracellular region of HER2 alone and in complex with the Herceptin Fab. Nature. 421:756–760. 2003. View Article : Google Scholar : PubMed/NCBI

20 

Franklin MC, Carey KD, Vajdos FF, Leahy DJ, de Vos AM and Sliwkowski MX: Insights into ErbB signaling from the structure of the ErbB2-pertuzumab complex. Cancer Cell. 5:317–328. 2004. View Article : Google Scholar : PubMed/NCBI

21 

von Minckwitz G, Huang CS, Mano MS, Loibl S, Mamounas EP, Untch M, Wolmark N, Rastogi P, Schneeweiss A, Redondo A, et al: Trastuzumab emtansine for residual invasive HER2-positive breast cancer. N Engl J Med. 380:617–628. 2019. View Article : Google Scholar : PubMed/NCBI

22 

Ponde N, Ameye L, Lambertini M, Paesmans M, Piccart M and de Azambuja E: Trastuzumab emtansine (T-DM1)-associated cardiotoxicity: Pooled analysis in advanced HER2-positive breast cancer. Eur J Cancer. 126:65–73. 2020. View Article : Google Scholar : PubMed/NCBI

23 

Geyer CE, Forster J, Lindquist D, Chan S, Romieu CG, Pienkowski T, Jagiello-Gruszfeld A, Crown J, Chan A, Kaufman B, et al: Lapatinib plus capecitabine for HER2-positive advanced breast cancer. N Engl J Med. 355:2733–2743. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Hsu W, Huang C, Yen C, Cheng A and Hsieh P: The HER2 inhibitor lapatinib potentiates doxorubicin-induced cardiotoxicity through iNOS signaling. Theranostics. 8:3176–3188. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Choi HD and Chang MJ: Cardiac toxicities of lapatinib in patients with breast cancer and other HER2-positive cancers: A meta-analysis. Breast Cancer Res Treat. 166:927–936. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Li J and Gu J: Cardiovascular toxicities with vascular endothelial growth factor receptor tyrosine kinase inhibitors in cancer patients: A meta-analysis of 77 randomized controlled trials. Clin. Drug Investg. 38:1109–1123. 2018. View Article : Google Scholar

27 

Ghatalia P, Je Y, Kaymakcalan MD, Sonpavde G and Choueiri TK: QTc interval prolongation with vascular endothelial growth factor receptor tyrosine kinase inhibitors. Br J Cancer. 112:296–305. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Borges VF, Ferrario C, Aucoin N, Falkson C, Khan Q, Krop I, Welch S, Conlin A, Chaves J, Bedard PL, et al: Tucatinib combined with ado-trastuzumab emtansine in advanced ERBB2/HER2-positive metastatic breast cancer A phase 1b clinical trial. JAMA Oncol. 4:1214–1220. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Force T, Krause D and Van Etten R: Molecular mechanisms of cardiotoxicity of tyrosine kinase inhibition. Nat Rev Cancer. 7:332–344. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Tomasovic A, Brand T, Schanbacher C, Kramer S, Hümmert MW, Godoy P, Schmidt-Heck W, Nordbeck P, Ludwig J, Homann S, et al: Interference with ERK-dimerization at the nucleocytosolic interface targets pathological ERK1/2 signaling without cardiotoxic Side-effects. Nat Commun. 11:17332020. View Article : Google Scholar : PubMed/NCBI

31 

Zamorano JL, Lancellotti P, Rodriguez Muñoz D, Aboyans V, Asteggiano R, Galderisi M, Habib G, Lenihan DJ, Lip GYH, Lyon AR, 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. 19:9–42. 2017. View Article : Google Scholar

32 

Nickel AC, Patel A, Saba NF, Leon AR, El-Chami MF and Merchant FM: Incidence of cancer treatment-induced arrhythmia associated with novel targeted chemotherapeutic agents. J The Am Heart Assoc. 7:e0101012018. View Article : Google Scholar

33 

Tamargo J, Caballero R and Delpon E: Cancer chemotherapy and cardiac arrhythmias: A review. Drug Safety. 38:129–152. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Herrmann J: Adverse cardiac effects of cancer therapies: Cardiotoxicity and arrhythmia. Nat Rev Cardiol. 17:474–502. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Curigliano G, Cardinale D, Suter T, Plataniotis G, de Azambuja E, Sandri MT, Criscitiello C, Goldhirsch A, Cipolla C and Roila F; ESMO Guidelines Working Group, : Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO Clinical Practice Guidelines. Ann Oncol. 23 (Suppl 7):S155–S166. 2012. View Article : Google Scholar

36 

Lang RM, Badano LP, Mor-Avi V, Afilalo J, Armstrong A, Ernande L, Flachskampf FA, Foster E, Goldstein SA, Kuznetsova T, et al: Recommendations for cardiac chamber quantification by echocardiography in adults: An update from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. Eur Heart J Cardiovasc Imaging. 16:233–271. 2015. View Article : Google Scholar : PubMed/NCBI

37 

Mitchell C, Rahko PS, Blauwet LA, Canaday B, Finstuen JA, Foster MC, Horton K, Ogunyankin KO, Palma RA and Velazquez EJ: Guidelines for performing a comprehensive transthoracic echocardiographic examination in Adults: Recommendations from the American society of echocardiography. J Am Soc Echocardiogr. 32:1–64. 2019. View Article : Google Scholar : PubMed/NCBI

38 

Thavendiranathan P, Grant AD, Negishi T, Plana JC, Popovic ZB and Marwick TH: Reproducibility of echocardiographic techniques for sequential assessment of left ventricular ejection fraction and volumes application to patients undergoing cancer chemotherapy. J Am Coll Cardiol. 61:77–84. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Dorosz JL, Lezotte DC, Weitzenkamp DA, Allen LA and Salcedo EE: Performance of 3-dimensional echocardiography in measuring left ventricular volumes and ejection fraction: A systematic review and meta-analysis. J Am Coll Cardiol. 59:1799–1808. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Nagueh SF, Appleton CP, Gillebert TC, Marino PN, Oh JK, Smiseth OA, Waggoner AD, Flachskampf FA, Pellikka PA and Evangelisa A: Recommendations for the evaluation of left ventricular diastolic function by echocardiography. J Am Soc Echocardiogr. 22:107–133. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Fallah-Rad N, Walker JR, Wassef A, Lytwyn M, Bohonis S, Fang T, Tian G, Kirkpatrick ID, Singal PK, Krahn M, et al: The utility of cardiac biomarkers, tissue velocity and strain imaging, and cardiac magnetic resonance imaging in predicting early left ventricular dysfunction in patients with human epidermal growth factor receptor II-positive breast cancer treated with adjuvant trastuzumab therapy. J Am Coll Cardiol. 57:2263–2270. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Upshaw JN, Finkelman B, Hubbard RA, Smith AM, Narayan HK, Arndt L, Domchek S, DeMichele A, Fox K, Shah P, et al: Comprehensive assessment of changes in left ventricular diastolic function with contemporary breast cancer therapy. JACC Cardiovasc Imaging. 13:198–210. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Timoteo AT, Moura Branco L, Filipe F, Galrinho A, Rio P, Portugal G, Oliveira S and Ferreira RC: Cardiotoxicity in breast cancer treatment: What about left ventricular diastolic function and left atrial function? Echocardiography. 36:1806–1813. 2019. View Article : Google Scholar : PubMed/NCBI

44 

Wang B, Yu Y, Zhang Y, Hao X, Zhao H, Yang S, Sun Q and Wang Y: Speckle tracking echocardiography in the early detection and prediction of anthracycline cardiotoxicity in diffuse large B-cell lymphoma treated with (R)-CHOP regimen. Echocardiography. 37:421–428. 2020. View Article : Google Scholar : PubMed/NCBI

45 

Park J, Park J, Park J and Cho G: Global longitudinal strain to predict mortality in patients with acute heart failure. J Am Coll Cardiol. 71:1947–1957. 2018. View Article : Google Scholar : PubMed/NCBI

46 

Ersbøll M, Valeur N, Mogensen UM, Andersen MJ, Møller JE, Velazquez EJ, Hassager C, Søgaard P and Køber L: Prediction of all-cause mortality and heart failure admissions from global left ventricular longitudinal strain in patients with acute myocardial infarction and preserved left ventricular ejection fraction. J Am Coll Cardiol. 61:2365–2373. 2013. View Article : Google Scholar : PubMed/NCBI

47 

Laufer-Perl M, Arnold JH, Mor L, Amrami N, Derakhshesh M, Moshkovits Y, Sadeh B, Arbel Y, Topilsky Y and Rozenbaum Z: The association of reduced global longitudinal strain with cancer therapy-related cardiac dysfunction among patients receiving cancer therapy. Clin Res Cardiol. 109:255–262. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Thavendiranathan P, Poulin F, Lim KD, Plana JC, Woo A and Marwick TH: Use of myocardial strain imaging by echocardiography for the early detection of cardiotoxicity in patients during and after cancer chemotherapy: A sys. J Am Coll Cardiol. 63:2751–2768. 2014. View Article : Google Scholar : PubMed/NCBI

49 

Sawaya H, Sebag IA, Plana JC, Januzzi JL, Ky B, Tan TC, Cohen V, Banchs J, Carver JR, Wiegers SE, et al: Assessment of Echocardiography and biomarkers for the extended prediction of cardiotoxicity in patients treated with anthracyclines, Taxanes, and trastuzumab. Circ Cardiovasc Imaging. 5:596–603. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Santoro C, Esposito R, Lembo M, Sorrentino R, De Santo I, Luciano F, Casciano O, Giuliano M, De Placido S, Trimarco B, et al: Strain-oriented strategy for guiding cardioprotection initiation of breast cancer patients experiencing cardiac dysfunction. Eur Heart J Cardiovasc Imaging. 20:1345–1352. 2019. View Article : Google Scholar : PubMed/NCBI

51 

Bayar N, Küçükseymen S, Göktaş S and Arslan Ş: Right ventricle failure associated with trastuzumab. Ther Adv Drug Saf. 6:98–102. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Grover S, Leong DP, Chakrabarty A, Joerg L, Kotasek D, Cheong K, Joshi R, Joseph MX, DePasquale C, Koczwara B and Selvanayagam JB: Left and right ventricular effects of anthracycline and trastuzumab chemotherapy: A prospective study using novel cardiac imaging and biochemical markers. Int J Cardiol. 168:5465–5467. 2013. View Article : Google Scholar : PubMed/NCBI

53 

Zaidi A, Knight DS, Augustine DX, Harkness A, Oxborough D, Pearce K, Ring L, Robinson S, Stout M, Willis J, et al: Echocardiographic assessment of the right heart in adults: A practical guideline from the British Society of Echocardiography. Echo Res Pract. 7:G19–G41. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Keramida K and Farmakis D: Right ventricular involvement in cancer therapy-related cardiotoxicity: The emerging role of strain echocardiography. Heart Fail Rev. Mar 3–2020.(Epub ahead of print). doi: 10.1007/s10741-020-09938-8. View Article : Google Scholar : PubMed/NCBI

55 

Cardinale D, Sandri MT, Colombo A, Colombo N, Boeri M, Lamantia G, Civelli M, Peccatori F, Martinelli G, Fiorentini C and Cipolla CM: Prognostic value of troponin I in cardiac risk stratification of cancer patients undergoing high-dose chemotherapy. Circulation. 109:2749–2754. 2004. View Article : Google Scholar : PubMed/NCBI

56 

Katsurada K, Ichida M, Sakuragi M, Takehara M, Hozumi Y and Kario K: High-sensitivity troponin T as a marker to predict cardiotoxicity in breast cancer patients with adjuvant trastuzumab therapy. Springerplus. 3:6202014. View Article : Google Scholar : PubMed/NCBI

57 

Kistorp C, Raymond I, Pedersen F, Gustafsson F, Faber J and Hildebrandt P: N-terminal pro-brain natriuretic peptide, C-reactive protein, and urinary albumin levels as predictors of mortality and cardiovascular events in older adults. JAMA. 293:1609–1616. 2005. View Article : Google Scholar : PubMed/NCBI

58 

Romano S, Fratini S, Ricevuto E, Procaccini V, Stifano G, Mancini M, Di Mauro M, Ficorella C and Penco M: Serial measurements of NT-proBNP are predictive of not-high-dose anthracycline cardiotoxicity in breast cancer patients. Br J Cancer. 105:1663–1668. 2011. View Article : Google Scholar : PubMed/NCBI

59 

Ekstein S, Nir A, Rein AJ, Perles Z, Bar-Oz B, Salpeter L, Algur N and Weintraub M: N-terminal-proB-type natriuretic peptide as a marker for acute anthracycline cardiotoxicity in children. J Pediatr Hematol Oncol. 29:440–444. 2007. View Article : Google Scholar : PubMed/NCBI

60 

van Boxtel W, Bulten BF, Mavinkurve-Groothuis AM, Bellersen L, Mandigers CM, Joosten LA, Kapusta L, de Geus-Oei LF and van Laarhoven HW: New biomarkers for early detection of cardiotoxicity after treatment with docetaxel, doxorubicin and cyclophosphamide. Biomarkers. 20:143–148. 2015. View Article : Google Scholar : PubMed/NCBI

61 

Demissei BG, Hubbard RA, Zhang L, Smith AM, Sheline K, McDonald C, Narayan V, Domchek SM, DeMichele A, Shah P, et al: Changes in cardiovascular biomarkers with breast cancer therapy and associations with cardiac dysfunction. J Am Heart Assoc. 9:e0147082020. View Article : Google Scholar : PubMed/NCBI

62 

Michel L, Mincu RI, Mrotzek SM, Korste S, Neudorf U, Rassaf T and Totzeck M: Cardiac biomarkers for the detection of cardiotoxicity in childhood cancer-a meta-analysis. ESC Heart Fail. 7:423–433. 2020. View Article : Google Scholar : PubMed/NCBI

63 

Wang Y, Bao L, Chu B, Gao S, Lu M, Shi L, Fu L, Fang L and Xiang Q: Progressive elevation of NT-ProBNP during chemotherapy is related to asymptomatic cardiovascular events in patients with multiple myeloma. Clin Lymphoma Myeloma Leuk. 19:167–176.e1. 2019. View Article : Google Scholar : PubMed/NCBI

64 

Pan W, Yang DH, Yu P and Yu HZ: Comparison of predictive value of NT-proBNP, sST2 and MMPs in heart failure patients with different ejection fractions. BMC Cardiovasc Disord. 20:2082020. View Article : Google Scholar : PubMed/NCBI

65 

Aula H, Skyttä T, Tuohinen S, Luukkaala T, Hämäläinen M, Virtanen V, Raatikainen P, Moilanen E and Kellokumpu-Lehtinen PL: ST2 levels increased and were associated with changes in left ventricular systolic function during a three-year follow-up after adjuvant radiotherapy for breast cancer. Breast. 49:183–186. 2020. View Article : Google Scholar : PubMed/NCBI

66 

Huang G, Zhai J, Huang X and Zheng D: Predictive value of soluble ST-2 for changes of cardiac function and structure in breast cancer patients receiving chemotherapy. Medicine. 97:e124472018. View Article : Google Scholar : PubMed/NCBI

67 

Frères P, Bouznad N, Servais L, Josse C, Wenric S, Poncin A, Thiry J, Moonen M, Oury C, Lancellotti P, et al: Variations of circulating cardiac biomarkers during and after anthracycline-containing chemotherapy in breast cancer patients. BMC Cancer. 18:1022018. View Article : Google Scholar : PubMed/NCBI

68 

Putt M, Hahn VS, Januzzi JL, Sawaya H, Sebag IA, Plana JC, Picard MH, Carver JR, Halpern EF, Kuter I, et al: Longitudinal changes in multiple biomarkers are associated with cardiotoxicity in breast cancer patients treated with doxorubicin, taxanes, and trastuzumab. Clin Chem. 61:1164–1172. 2015. View Article : Google Scholar : PubMed/NCBI

69 

Knight D, Zumbo G, Barcella W, Steeden JA, Muthurangu V, Martinez-Naharro A, Treibel TA, Abdel-Gadir A, Bulluck H, Kotecha T, et al: Cardiac structural and functional consequences of amyloid deposition by cardiac magnetic resonance and echocardiography and their prognostic roles. JACC. Cardiovasc Imaging. 12:823–833. 2019.

70 

Tak T, Jaekel C, Gharacholou S, Dworak M and Marshall S: Measurement of ejection fraction by cardiac magnetic resonance imaging and echocardiography to monitor doxorubicin-induced cardiotoxicity. Int J Angiol. 29:45–51. 2020. View Article : Google Scholar : PubMed/NCBI

71 

Armstrong G, Plana J, Zhang N, Srivastava D, Green DM, Ness KK, Daniel Donovan F, Metzger ML, Arevalo A, Durand JB, et al: Screening adult survivors of childhood cancer for cardiomyopathy: Comparison of echocardiography and cardiac magnetic resonance imaging. J Clin Oncol. 30:2876–2884. 2012. View Article : Google Scholar : PubMed/NCBI

72 

Schneeweiss A, Chia S, Hickish T, Harvey V, Eniu A, Hegg R, Tausch C, Seo JH, Tsai YF, Ackrill A, et al: Neoadjuvant pertuzumab and trastuzumab concurrent or sequential with an anthracycline-containing or concurrent with an anthracycline-free standard regimen: A randomized phase II study (TRYPHAENA). Cancer Res. 71 (Suppl 24):S5–S6. 2011.

73 

Du XL, Xia R, Burau K and Liu CC: Cardiac risk associated with the receipt of anthracycline and trastuzumab in a large nationwide cohort of older women with breast cancer, 1998–2005. Med Oncol. 28 (Suppl 1):S80–S90. 2011. View Article : Google Scholar : PubMed/NCBI

74 

Zamorano J, Lancellotti P, Rodriguez Muñoz D, Aboyans V, Asteggiano R, Galderisi M, Habib G, Lenihan DJ, Lip GYH, Lyon AR, 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. 37:2768–2801. 2016. View Article : Google Scholar : PubMed/NCBI

75 

Armenian SH, Lacchetti C, Barac A, Carver J, Constine LS, Denduluri N, Dent S, Douglas PS, Durand JB, Ewer M, et al: Prevention and monitoring of cardiac dysfunction in survivors of adult cancers: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 35:893–911. 2017. View Article : Google Scholar : PubMed/NCBI

76 

Plana JC, Galderisi M, Barac A, Ewer MS, Ky B, Scherrer-Crosbie M, Ganame J, Sebag IA, Agler DA, Badano LP, 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. J Am Soc Echocardiogr. 27:911–939. 2014. View Article : Google Scholar : PubMed/NCBI

77 

Loap P, Kirov K and Kirova Y: Cardiotoxicity in breast cancer patients treated with radiation therapy: From evidences to controversies. Crit Rev Oncol Hematol. 156:1031212020. View Article : Google Scholar : PubMed/NCBI

78 

Common Terminology Criteria for Adverse Events (CTCAE) Version 5.0 Published, Nov 27, 2017. simplehttps://ctep.cancer.gov/protocoldevelopment/electronic_applications/docs/ctcae_v5_quick_reference_8.5×11.pdf

79 

Zipes DP, Camm AJ, Borggrefe M, Buxton AE, Chaitman B, Fromer M, Gregoratos G, Klein G, Moss AJ, Myerburg RJ, et al: ACC/AHA/ESC 2006 Guidelines for management of patients with ventricular arrhythmias and the prevention of sudden cardiac death a report of the American College of Cardiology/American Heart Association Task Force and the European Society of Cardiology Committee for practice Guidelines (Writing Committee to Develop Guidelines for management of patients with ventricular arrhythmias and the prevention of Sudden Cardiac Death): Developed in Collaboration with the European Heart Rhythm Association and the Heart Rhythm Society. Circulation. 114:e385–e484. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

February-2021
Volume 21 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guan J and Guan J: Cardiotoxicity of anthracycline‑free targeted oncological therapies in HER2‑positive breast cancer (Review). Oncol Lett 21: 100, 2021
APA
Guan, J., & Guan, J. (2021). Cardiotoxicity of anthracycline‑free targeted oncological therapies in HER2‑positive breast cancer (Review). Oncology Letters, 21, 100. https://doi.org/10.3892/ol.2020.12361
MLA
Guan, J., Zhang, M."Cardiotoxicity of anthracycline‑free targeted oncological therapies in HER2‑positive breast cancer (Review)". Oncology Letters 21.2 (2021): 100.
Chicago
Guan, J., Zhang, M."Cardiotoxicity of anthracycline‑free targeted oncological therapies in HER2‑positive breast cancer (Review)". Oncology Letters 21, no. 2 (2021): 100. https://doi.org/10.3892/ol.2020.12361