Next Article in Journal
Left Atrial Mechanics and Remodeling in Paroxysmal Atrial Fibrillation: Introducing the EASE Score for Pre-Ablation Risk Prediction
Previous Article in Journal
Phenotypic and Spatial Characterization of Tumor-Associated Macrophages in Non-Metastatic Seminoma: Association with Local Tumor Progression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cardiotoxicity in Breast Cancer Therapy: Risks, Mechanisms, and Prevention Strategies

by
Annisa Eka Fitrianti
1,2,
Nadea Olyvia Wardani
1,
Astri Astuti
3,
Kusnandar Anggadiredja
4,
Lia Amalia
4,
Risani Andalasia Putri
5 and
Zulfan Zazuli
4,*
1
Master’s Program in Pharmacy, School of Pharmacy, Institut Teknologi Bandung, Bandung 40132, Indonesia
2
Department of Pharmacy, Dr. Hasan Sadikin General Hospital, Bandung 40161, Indonesia
3
Department of Cardiology, Dr. Hasan Sadikin General Hospital, Bandung 40161, Indonesia
4
Department of Pharmacology—Clinical Pharmacy, Institut Teknologi Bandung, Bandung 40132, Indonesia
5
Department of Pharmacy, Dharmais Cancer Hospital, Jakarta 11420, Indonesia
*
Author to whom correspondence should be addressed.
Med. Sci. 2025, 13(3), 130; https://doi.org/10.3390/medsci13030130
Submission received: 25 June 2025 / Revised: 4 August 2025 / Accepted: 11 August 2025 / Published: 14 August 2025

Abstract

Breast cancer is the most prevalent cancer in women. Anthracyclines are commonly used as the first line of treatment, often combined with other agents, including trastuzumab. Despite their efficacy, both drugs pose a risk of cardiotoxicity, which may impair patients’ quality of life (QoL) and hinder treatment persistence. Anthracycline-induced cardiotoxicity is dose-dependent and generally irreversible, whereas trastuzumab is associated with potentially reversible cardiac dysfunction. This review discusses the risk factors and biological mechanisms underlying chemotherapy-induced cardiotoxicity in breast cancer and explores effective strategies for prevention and treatment. It has been demonstrated that several cardioprotective strategies, such as treatments with angiotensin-converting enzyme inhibitors (ACEis), angiotensin receptor blockers (ARBs), beta-blockers, and dexrazoxane, can help lessen cardiotoxic effects. A better understanding of cardioprotective strategies may help optimize cancer treatment without compromising cardiovascular function.

1. Introduction

Breast cancer arises from the uncontrolled proliferation of epithelial cells lining the ducts or lobules of the breast, making it the most common cancer among women, accounting for approximately one-third of all cancer diagnoses in this population [1]. According to Global Burden of Cancer Study (Globocan) data for 2022, the incidence of new breast cancer cases was 66,271, constituting 16.2% of the total 408,661 new cancer cases in Indonesia, with a prevalence of 209,748 cases (20.6%) during the past five years [2].
For many years, anthracycline-based chemotherapy, including doxorubicin, has been the primary option for breast cancer treatment, frequently in conjunction with targeted therapy like trastuzumab [3,4]. However, its use is associated with toxicities ranging from mild to severe, with both short- and long-term implications [5]. Cardiotoxicity, marked by a reduction in the left ventricular ejection fraction (LVEF), is a prevalent adverse effect. According to Wang et al. patients with a cardiovascular risk may experience more severe effects from cardiotoxicity brought on by doxorubicin and trastuzumab, which may ultimately make it more difficult to continue treatment and raise the risk of death [6]. Cardiotoxicity associated with trastuzumab frequently manifests in the initial phases of treatment and typically presents without clinical symptoms [7].
This article aims to review cardiotoxicity as a significant consequence of breast cancer treatment, particularly associated with doxorubicin and trastuzumab. In conjunction with advancements in cancer treatment, the side effects manifesting as a compromised cardiac function need to be taken into consideration as they can disrupt the continuity of therapy and adversely affect the patient’s quality of life (QoL). This article is aimed at explaining the fundamental biological mechanisms and identifying risk factors associated with cardiotoxicity. This article also reviews various strategies for the prevention and therapy of cardiotoxicity to mitigate its adverse effects on cardiac function.

2. Literature Searching

This review summarizes the results of several studies related to cardiotoxicity in breast cancer therapy, particularly anthracycline-based chemotherapy and trastuzumab. The included studies were identified through the PubMed database using the keywords “cardiotoxicity,” “doxorubicin,” “trastuzumab,” “breast cancer,” and “left ventricular ejection fraction.” An initial total of 402 articles published within the past 10 years were retrieved. The search results were further refined by limiting them to studies involving human subjects, yielding 185 articles. Studies that did not specifically examine cardiotoxicity related to anthracyclines and trastuzumab were excluded. Additionally, research communications, theses, review articles, expert opinions, and irrelevant studies were also excluded. Following abstract screening based on predefined inclusion criteria, 26 studies were ultimately selected for this review. Most of these studies explored the comparative cardiotoxic effects of different treatment regimens, identified risk factors associated with cardiotoxicity, and evaluated various cardioprotective strategies. Differences in findings between studies were also discussed.

3. Breast Cancer

Breast cancer is a disease that arises from the glands, ducts, and supportive tissues within the breast. This disease is among the most prevalent forms of cancer in women, alongside cervical cancer. Cancer cells proliferate uncontrollably and may develop in the lobules (milk-producing glands), ducts (milk-transporting channels), adipose tissue, or connective tissue within the breast [8]. The initial proliferation of cancer cells often transpires within the ducts and/or lobules (in situ), which at this juncture is not life-threatening; nonetheless, cancer cells may disseminate to adjacent breast tissue (invasively), resulting in the formation of tumors that manifest as lumps or thickening. Invasive breast cancer can metastasize to adjacent lymph nodes or other organs [9].
According to the 2022 Globocan statistical data from the World Health Organization (WHO), breast cancer has the second highest incidence following lung cancer. Worldwide, there are 2,296,840 newly diagnosed cases of breast cancer, resulting in a mortality rate of 666,103, positioning it as the fourth greatest cause of cancer-related fatalities. As of the end of 2022, a total of 8,178,393 individuals were diagnosed with breast cancer within the preceding five years [2].
In Asia, 48 countries exhibit diverse trends in the breast cancer incidence throughout time. From 1990 to 2017, the incidence of breast cancer rose markedly, with the age-standardized incidence rate (ASIR) increasing by 54% in Central Asia, 89.5% in East Asia, and 76.7% in South Asia [10]. In 2018, breast cancer represented 10% of all cancer cases in Asia, totaling 845,400 cases [11]. By 2040, breast cancer diagnoses are projected to reach 1.34 million [2].
Genetic mutations mostly cause breast cancer, particularly those affecting tumor suppressor genes such as BRCA-1, BRCA-2, p53, and PTEN [1]. Mutations in the BRCA1 and BRCA2 genes, which are crucial to DNA repair and cellular growth regulation, can elevate the risk of tumorigenesis. These two genes experience heritable functional alterations that influence the expression profile of oncogenes and contribute to malignancy [12,13].
The predominant and generally accepted classification of breast cancer relies on an immunohistochemical methodology. This methodology delineates critical biomarkers, including the estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), utilized to categorize breast cancer into five subtypes: luminal A, luminal B, HER2 positive, triple-negative breast cancer (TNBC), and normal-like [14,15]. The TNBC subtype is defined by the lack of the expression of all three receptors, resulting in distinct biological features compared to other subtypes [14].
The HER2-positive subtype of breast cancer is defined by the overexpression or amplification of the HER2 protein, representing roughly 20–30% of all breast cancer [16,17]. Alongside the TNBC subtype, HER2-positive breast cancer demonstrates a more aggressive behavior and has a diminished overall prognosis relative to other types [18,19]. The National Cancer Institute study on survival rates indicates that women with the luminal A subtype have the most favorable prognosis, with a 5-year relative survival rate of 94.4%. The luminal B subtype has a survival rate of 90.7%, followed by the HER2 subtype at 84.8%, whilst the TNBC subtype exhibits the lowest survival rate at 77.1% [20].

4. Treatment of Breast Cancer

Over the past twenty years, the management of early breast cancer (stage I–IIIA) has advanced considerably, incorporating a combination of pharmacological agents, radiotherapy, and surgical interventions. Nonetheless, anthracyclines continue to be a principal element of breast cancer treatment, sometimes utilized with novel targeted therapies such as trastuzumab [4]. A meta-analysis by Bradley et al. indicates that anthracycline-based regimens, particularly when combined with taxanes, can decrease the chance of breast cancer recurrence within 10 years by as much as one-third compared to non-chemotherapy approaches. This medication also decreases the death rate from breast cancer by 20–25% [21].
Doxorubicin is a cytotoxic drug belonging to the anthracycline class, developed from Streptomyces species. The mechanism of action of anthracyclines is essentially analogous, specifically by intercalating DNA bases into the molecule, hence perturbing the DNA helical structure. This disruption impedes DNA synthesis and influences the activity of the DNA topoisomerase II enzyme. Furthermore, anthracyclines can generate iron-mediated free radicals that may target and harm rapidly proliferating cells [22,23,24]. Trastuzumab is a targeted therapeutic drug beneficial in treating HER2-positive breast cancer. Trastuzumab is a humanized recombinant monoclonal antibody specifically engineered to bind the extracellular domain of the HER2 protein, thereby inhibiting dimerization and obstructing downstream intracellular signaling pathways, which leads to cell cycle arrest and the inhibition of DNA repair following chemotherapy [25,26,27,28].
During the 2005 annual meeting of the American Society of Clinical Oncology (ASCO), the advantages of trastuzumab in conjunction with anthracycline/taxane-based adjuvant therapy for HER2-positive breast cancer were presented for the first time. The meta-analysis results indicate that one year of trastuzumab therapy in early-stage HER2-positive breast cancer markedly enhances the disease-free survival (DFS) and overall survival (OS), while diminishing the risk of recurrence and mortality [21]. Consistent with prior research findings, the amalgamation of trastuzumab with chemotherapy has demonstrated a substantial enhancement in DFS by 36–58% and OS by 24–59% [29,30,31,32].
The Neoadjuvant Herceptin (NOAH) clinical trial was performed to evaluate the efficacy of treatment in HER2-positive breast cancer patients, with 118 patients receiving chemotherapy only. In contrast, 117 more patients underwent a regimen of chemotherapy combined with trastuzumab. The trial results indicated that the 5-year event-free survival (EFS) was 58% (95% CI 48–66) for the group receiving chemotherapy with trastuzumab, in contrast to 43% (95% CI 34–52) for the group receiving chemotherapy alone [33]. A further study has shown that the use of adjuvant trastuzumab alongside paclitaxel can diminish the risk of local–regional recurrence and mitigate toxicity in HER2-positive breast cancer patients with tumors ≤ 2 cm and no lymph node involvement [34].
Other treatment types in early-stage breast cancer include the use of systemic adjuvant therapy to reduce the risk of recurrence. Treatment decisions are based on an individual’s risk of relapse and the likelihood of a response, taking into account factors such as the ER/PR and HER2 status [35]. For patients with hormone receptor-positive and HER2-negative breast cancer, endocrine therapy with tamoxifen or aromatase inhibitors remains the cornerstone of adjuvant treatment [36]. In cases of HR+/HER2− breast cancer with a high risk of recurrence, CDK4/6 inhibitors such as abemaciclib have been shown to provide significant benefits in the adjuvant setting when combined with endocrine therapy [37,38,39]. The combination of abemaciclib and endocrine therapy significantly improved the invasive disease-free survival (IDFS) compared to endocrine therapy alone (HR 0.75; 95% CI: 0.60–0.93), with a two-year IDFS rate of 92.2% in the combination group versus 88.7% in the endocrine therapy-only group [39].
In addition, for patients with germline BRCA1/2 mutations and HER2-negative status, adjuvant olaparib has been shown to significantly reduce the risk of recurrence. A study with a median follow-up of 2.5 years reported a three-year IDFS rate of 85.9% in the olaparib group, compared to 77.1% in the placebo group [40]. For patients whose breast cancer progresses after the first-line treatment with trastuzumab or pertuzumab, the standard second-line therapy is an antibody–drug conjugate such as trastuzumab emtansine (T-DM1). Treatment with trastuzumab emtansine was associated with a median progression-free survival of 9.6 months and a median overall survival of 30.9 months [41]. Furthermore, immunotherapy—particularly immune checkpoint inhibitors such as pembrolizumab and atezolizumab—has shown benefits in patients with TNBC, especially those who are PD-L1 positive. The KEYNOTE-522 study reported improvements in the pathologic complete response (pCR) and EFS in early-stage TNBC patients receiving neoadjuvant pembrolizumab [42].

5. Cardiotoxicity

The National Cancer Institute characterizes cardiotoxicity as a toxic illness impacting the heart. This concept encompasses both the direct impact of the drug on cardiac function and the indirect effects resulting from alterations in the hemodynamic flow or thrombotic occurrences [43]. The Cardiac Review and Evaluation Committee defines cardiotoxicity as a condition encountered by patients following an anticancer treatment, characterized by one or more of the subsequent criteria:
  • Cardiomyopathy is defined by a reduction in the LVEF or a decrease in ventricular septal motion.
  • The manifestation of heart failure symptoms.
  • Tachycardia.
  • A reduction in the LVEF of ≥5% to <55% is associated with heart failure symptoms, or a decline in the LVEF of ≥10% to <55% in the absence of symptoms [44].
However, despite efforts to standardize criteria, there remains considerable heterogeneity in definitions of cardiotoxicity, especially in relation to LVEF cutoffs. Different organizations propose varying thresholds. The American Society of Echocardiography (ASE) and the European Society of Cardiovascular Imaging (EACVI) guidelines indicate that cardiotoxicity is diagnosed when there is a reduction in the ejection fraction (EF) of ≥10%, with an absolute value of <53% [45].
The European Society of Cardiology (ESC) establishes that the classification is divided into two main categories: Symptomatic Cancer Therapy-Related Cardiovascular Dysfunction (CTRCD) and Asymptomatic CTRCD. Symptomatic CTRCD applies when a patient is experiencing symptoms of heart failure. The severity is graded as follows:
  • Very severe, HF requiring inotropic support, mechanical circulatory support, or the consideration of transplantation.
  • Severe, HF hospitalization.
  • Moderate, need for the outpatient intensification of diuretic and HF therapy.
  • Mild, mild HF symptoms, no intensification of therapy required.
Cancer therapy-related cardiovascular toxicity definitions for asymptomatic CTRCD:
  • Severe, a new LVEF reduction to,40%
  • Moderate, a new LVEF reduction by ≥10 percentage points to an LVEF of 40–49% or a new LVEF reduction by 10 percentage points to an LVEF of 40–49% and either a new relative decline in GLS by >15% from baseline OR a new rise in cardiac biomarkers.
  • Mild, an LVEF ≥ 50% and a new relative decline in GLS by >15% from baseline and/or a new rise in cardiac biomarkers [46].
A reduction in the LVEF can be classified as symptomatic or asymptomatic according to its degree of reversibility, as outlined by Plana et al.:
  • If the LVEF improves by 5% points from the baseline, the condition is considered reversible.
  • If the improvement reaches ≥10% points from the lowest point but is still more than 5% below the baseline, it is classified as partially reversible.
  • If the improvement is less than 10% points from the lowest point and is still more than 5% points below the baseline, the decrease in the LVEF is considered irreversible [47].
Cardiotoxicity can develop over a long period, starting from the beginning of treatment to at least four years after chemotherapy ends [48]. This condition can appear in several forms:
  • Acute or subacute, which occurs from the start of treatment until two weeks after the therapy is complete.
  • Chronic, which develops at least one year after the end of the therapy. Chronic cardiotoxicity is further classified into two types, namely early chronic if it appears in the first year after therapy and advanced chronic if it occurs in the years following the completion of the treatment [49,50].
This lack of uniformity complicates cross-trial comparisons and clinical decision-making. The variability may also impact the patient stratification, treatment discontinuation decisions, and eligibility for cardioprotective interventions.
In addition to the LVEF, newer modalities such as global longitudinal strain (GLS) are sensitive imaging biomarkers used to detect early subclinical changes in left ventricular contractility in patients undergoing chemotherapy with a cardiotoxic potential. A relative reduction in GLS exceeding 15% from the baseline has been reported as an accurate predictor of a subsequent decrease in the LVEF [45,47,51]. This condition is considered abnormal and is an early indicator of subclinical left ventricular dysfunction. However, to date, the use of GLS and its threshold in predicting cardiotoxicity has not been standardized [52].
Troponin I have also emerged as a biomarker for early myocardial injury. Several studies have shown that troponin I measurements after the start of chemotherapy are useful in predicting the incidence and severity of cardiotoxicity, especially in patients receiving anthracyclines and those undergoing combination therapy including trastuzumab [53,54,55]. Cardiac troponin, a regulatory protein in cardiomyocytes, is released into the circulation upon a myocardial injury. Studies indicate that troponin may detect cardiotoxicity at the preclinical stage, even before reductions in the LVEF are evident, especially in patients undergoing therapy with anticancer drugs [54,55,56]. Ultimately, the diverse definitions and evolving diagnostic modalities highlight the urgent need for unified, guideline-driven criteria for diagnosing cardiotoxicity. This would enhance consistency in clinical practice, enable better risk stratification, and facilitate more accurate inter-study comparisons.

6. Treatment and Cardiotoxicity

Among antineoplastic agents, drugs that are most at risk of causing cardiotoxicity can be categorized into two types:
  • Type I works through mechanisms such as anthracyclines. Cardiotoxicity in this type is dose-dependent and causes irreversible heart damage.
  • Type II, which works through mechanisms such as trastuzumab. In this type, cardiotoxicity is reversible, so the treatment can be temporarily stopped until the patient recovers and resumed if necessary. This is possible because there is no change in the ultrastructure of the myocytes, so the heart damage is not permanent [57,58].
Anthracyclines are a group of chemotherapy agents that are best known to have the potential to cause cardiac dysfunction, with a risk five times higher than non-anthracycline agents [59]. The risk of heart failure from the use of anthracyclines increases with increasing cumulative doses. At a dose of 400 mg/m2, the risk reaches 3–5%, while at a dose of 700 mg/m2, the risk can increase to 18–48% [60]. A 2013 meta-analysis reported that after a 9-year follow-up period, clinical cardiac toxic effects occurred in 6% of patients receiving treatment with anthracyclines, while subclinical cardiac toxic effects were found in 18% of patients [61]. Although doxorubicin doses exceeding 500 mg/m2 were frequently used in the past [62], current guidelines have limited the maximum cumulative dose to 450 mg/m2 [63]. Despite the ongoing debate regarding the long-term impact of various dosing regimens—largely due to differences in follow-up durations—the recommended lifetime cumulative dose of doxorubicin is now set at less than 450 mg/m2 to minimize the risk of cardiotoxicity [64].
Trastuzumab is an effective therapy for breast cancer, but its use can cause serious side effects in the form of significant cardiotoxicity. The HER2 receptor, which is the main target of trastuzumab, is not only found in tumor cells but is also expressed in cardiomyocytes. This receptor is thought to have a cardioprotective role in responding to cellular stress. Therefore, the use of trastuzumab can trigger a significant decrease in the LVEF and potentially cause heart failure [65,66]. This risk increases when trastuzumab is combined with other chemotherapies known to be cardiotoxic, such as anthracyclines [65]. A multicenter phase III trial conducted showed that the combination of trastuzumab with anthracyclines significantly increased the risk of cardiac dysfunction and heart failure. The results of this study revealed that up to 27% of HER2-positive metastatic breast cancer patients who received the combination experienced cardiotoxicity, compared to 8% in the group that only received anthracyclines. Other studies have also revealed the same thing, that the combination of anthracyclines and trastuzumab, with a higher percentage of doxorubicin compared to other types of anthracyclines, will further decrease the LVEF value when compared to anthracyclines alone [67].
A meta-analysis involving 21 prognostic studies examined patients undergoing treatment with anthracyclines, both with and without trastuzumab. The study identified that an absolute change in GLS of 2–3% and a relative change of 10–15% can be used as a threshold for detecting subclinical cardiotoxicity [68]. Similar findings were reported in a recent meta-analysis, which revealed that patients with cardiotoxicity experienced a 14.13% greater decrease in GLS than those who did not experience the condition. This further reinforces that GLS has a high predictive value in detecting cardiotoxicity and subclinical left ventricular dysfunction [69].
Another study found that a significant increase in troponin I (>30 ng/mL) in HER2-positive breast cancer patients undergoing anthracycline therapy followed by trastuzumab may be an early indicator of cardiotoxicity [56]. The ACC/AHA guidelines provide a Class I recommendation for measuring cardiac troponin (cTn) levels in patients presenting with chest pain and advise the use of high-sensitivity cardiac troponin (hs-cTn) assays for serial measurements [70]. This recommendation is supported by a subsequent study involving 533 breast cancer patients who underwent the serial monitoring of hs-cTnI and hs-cTnT levels during trastuzumab treatment. Patients with elevated baseline troponin levels (>40 ng/L for hs-cTnI and >14 ng/L for hs-cTnT) had a fourfold higher risk of developing left ventricular dysfunction [71].
Cardiotoxicity is a serious complication of both agents because it can interfere with the QoL and overall survival of breast cancer patients. Therefore, this aspect is a major concern in patient management [72,73]. An LVEF evaluation is recommended before and after anthracycline therapy, as well as before and every three months during trastuzumab use [74]. If the LVEF decreases to below 50%, with or without symptoms, the discontinuation of trastuzumab is recommended [17]. The cardiotoxic effects of the treatment force the cessation of therapy, which ultimately risks reducing the overall patient survival [75].
Cardiotoxicity is often the reason for the discontinuation of treatment, which has an impact on the increased risk of cancer recurrence and mortality. It is reported that this risk can reach up to 40% in the five years after undergoing chemotherapy [76]. The results of a randomized trial of adjuvant trastuzumab in combination with an anthracycline–taxane-based regimen showed a 12–15% decrease in the asymptomatic LVEF. However, few patients experienced the permanent discontinuation of treatment [77,78,79,80]. Approximately 18% of all patients experience trastuzumab-induced cardiotoxicity (trastuzumab-induced cardiotoxicity/TIC), which results in a delay in therapy. In line with previous research, TIC is the main cause of the discontinuation of adjuvant trastuzumab treatments, with rates reaching 62% [81,82,83].
Other research identifies the discontinuation of trastuzumab therapy as a significant prognostic factor, with patients who discontinue therapy having up to three times worse DFS and OS than those who complete treatment for one year or experience only a delay of less than 14 days [75]. In addition, Gong et al. reported that patients who discontinue adjuvant trastuzumab have a higher risk of disease recurrence and emphasized that the early discontinuation of therapy is a stronger prognostic factor for a poorer survival than cardiotoxicity itself [84].

7. Cardiotoxicity Mechanism

The cardiotoxicity mechanisms of doxorubicin and trastuzumab involve different pathways, yet both may contribute to the development of myocardial dysfunction (Figure 1). The cardiotoxic effects of doxorubicin primarily arise from three interconnected mechanisms: oxidative stress, the dysregulation of cell death pathways, and epigenetic modifications [47,85,86]. In terms of oxidative stress, doxorubicin plays a role in producing reactive oxygen species (ROS) [87,88,89], as well as increasing iron mobilization [87,90]. When ferric ions bind anthracyclines, ROS are generated, promoting the redox cycling of ferrous to ferric ions. This redox cycle induces widespread cellular damage, affecting the mitochondrial membrane, nucleus, plasma membrane, and endoplasmic reticulum. As a result, there is a decrease in intracellular calcium levels, which has an impact on the decreased heart contractility [85,91,92].
Additional studies have shown that doxorubicin induces a cardiomyocyte injury through interactions with DNA topoisomerase II beta (TOP2B) [83]. Cardiomyocyte damage is not only influenced by ROS but also by the TOP2B–ROS–DNA complex. This complex causes the suppression of transcription factors (through p53 activation), DNA degradation, and apoptosis that cannot be avoided due to mitochondrial dysfunction [89].
Doxorubicin induces cardiomyocyte death via multiple pathways, including intrinsic and extrinsic apoptosis which occurs through both intrinsic and extrinsic pathways. Recent studies have also revealed that pyroptosis and ferroptosis contribute to the loss of functioning myocytes, especially through the upregulation of non-coding RNA that plays a role in terminal differentiation. These processes result in iron dysregulation and elevated lipid peroxidation, thereby exacerbating cardiomyocyte damage [85,87,90].
Doxorubicin causes an increase in the levels of labile iron in cells, which contributes to its cytotoxic effect. In another mechanism, doxorubicin and its metabolites can disrupt the balance of iron homeostasis by interfering with the expression of genes that play a role in iron metabolism. Furthermore, doxorubicin also inhibits the glutathione peroxidase activity in the cytosol and mitochondria, which triggers lipid peroxidation and ultimately causes ferroptosis [93]. In addition, doxorubicin can damage the cardiomyocyte mitochondria, which then causes heart failure through the induction of pathological autophagy. This induction has the potential to cause cell death rather than functioning as a normal mechanism for recycling and cleaning up damaged cellular components [85,94].
Doxorubicin-associated cardiotoxicity is also mediated through epigenetic modifications. This drug is known to downregulate DNA methylation, which has an impact on changes in the mitochondrial gene expression [95]. In addition, anthracyclines also increase histone deacetylation activity, which causes the deacetylation of certain proteins, such as α-tubulin [96]. Doxorubicin also affects the regulation of various microRNAs, although the further impact of these changes is not yet fully understood [97].
The precise mechanism of trastuzumab-induced cardiotoxicity remains incompletely understood. In vivo investigations have shown that trastuzumab causes ultrastructural changes in rat heart tissue and significantly alters the expression of genes involved in the cardiac function, mitochondria, and DNA repair [98].
The inhibition of the NRG1/HER signaling pathway and its downstream pathways has long been considered the main mechanism contributing to TIC. In addition, recent research highlights the role of autophagy inhibition and changes in cellular metabolic pathways in cardiomyocytes as potential factors in the development of cardiotoxicity [99,100,101]. Mohan et al. found that treatment with trastuzumab led to the decreased expression of proteins involved in the autophagy signaling pathway, including ATG5-12, ATG7, ATG14, and Beclin. In addition, their research shows that trastuzumab-induced autophagy inhibition contributes to increased ROS production in cardiomyocytes, potentially leading to oxidative stress and cellular damage [101].
HER2 is not only expressed in tumor tissue but is also found in adult cardiomyocytes along with other family members, such as HER1, HER3, and HER4 [102], although to a lesser extent than in the embryonic heart [103]. Together with its ligand, NRG1, HER2 plays an important role in maintaining the function and survival of cardiomyocytes. When the heart experiences hemodynamic stress, the microvascular endothelial cells release NRG1 [103,104,105], which then binds to HER4 and triggers HER4/HER4 homodimerization or HER4/HER2 heterodimerization, activating downstream signaling pathways such as mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinases (PI3K)-Akt to support heart survival and function [25,106].
This study demonstrates that the HER2 deletion in the murine model causes spontaneous dilated cardiomyopathy and increases the susceptibility to cardiovascular stress. Collectively, these findings underscore the critical role of HER2 in preserving the cardiac structure and function, particularly under stress conditions, especially under conditions of physiological or pharmacological stress, which explains how the HER2 inhibition by trastuzumab can contribute to cardiotoxicity [107,108].
Akt plays an important role in cell survival by activating various proteins through phosphorylation, which ultimately prevents apoptosis and supports cell growth [109]. Research conducted by Ravingerova et al., using a mouse model with chronic cardiac ischemia, showed that the Akt activation increases the glucose and lipid metabolism in cardiomyocytes, allowing for better nutrient absorption and maintaining energy during hypoxic conditions [110]. In addition, the PI3K-Akt pathway also contributes to cell protection by stimulating the production of NO in adult ventricular myocytes, which helps fight oxidative stress. Akt also plays a role in regulating mitochondrial respiration, thus suppressing ROS production and increasing cell survival [111]. However, when HER2 is inhibited, as in the treatment with trastuzumab, the PI3K-Akt pathway cannot be activated, causing an increase in ROS accumulation in cardiomyocytes and ultimately triggering apoptosis and cardiac dysfunction [111,112].
The MAPK (MEK/ERK) pathway also plays a role in the survival of cardiomyocytes by amplifying external signals that support cell proliferation and differentiation [113,114]. The decrease in HER2 activity due to trastuzumab weakens the MEK/ERK pathway, which then triggers apoptosis [115]. The inhibition of this pathway also increases the number of mitochondrial permeability transition pores (mPTPs), causing an increased sensitivity to excess calcium (Ca2+), the excessive production of cytotoxic ROS, and a decreased gap junction permeability, which ultimately contributes to cardiomyocyte damage [116].

8. Risk Factors of Cardiotoxicity

Key risk factors for chemotherapy-induced heart failure include the cumulative drug dose, an age over 65, concurrent treatment with or prior to thoracic radiation, the co-administration of cardiotoxic agents, and pre-existing cardiovascular disease [117]. Some known risk factors associated with the development of anticancer treatment-induced cardiotoxicity are described below:
  • Age
An advancing age significantly elevates the risk of cardiotoxicity. The results showed that women aged 65 years and over who received a combination of anthracyclines and trastuzumab, as well as women aged 75 years and over who received trastuzumab alone, experienced an increased risk of heart failure [65]. In line with other studies reporting a consistently high risk in women aged 65 and over, both those receiving anthracyclines [118] and combinations of anthracyclines with trastuzumab or trastuzumab alone had higher risks compared to those receiving other types of chemotherapy [119].
2.
Hypertension
Hypertension is a prominent risk factor for doxorubicin-induced cardiotoxicity. This is most likely due to the pressure and initial damage to the heart before treatment, which then increases the risk of further damage [120]. Advances in the monoclonal antibody, tyrosine kinase, and other molecular target-based cancer therapies have significantly improved the survival of patients with advanced cancer, but these innovations also raise concerns about secondary hypertension, which can contribute to left ventricular dysfunction and be a major factor in adverse cardiotoxicity [121].
3.
Overweight and Obesity
Epidemiological studies show that being overweight or obese not only increases the risk of breast cancer but also reduces the effectiveness of the treatment, lowers the QoL after diagnosis, and increases cancer mortality [122,123]. Obesity markedly increases the risk of cardiotoxicity, especially when doxorubicin is used alone or in combination with trastuzumab. Individuals who are obese or overweight have a higher risk of cardiotoxicity after an anthracycline-based therapy [124,125].
4.
Diabetes
Diabetes is independently associated with an elevated risk of anticancer-induced heart failure. Several mechanisms that play a role in increasing this risk include lipotoxicity, hyperglycemia, and mitochondrial dysfunction, which contribute to the development of cardiotoxicity [126].
5.
Dyslipidemia
Hyperlipidemia has been investigated in the context of other cardiovascular risk factors, such as hypertension and diabetes. Research shows that patients with these conditions have a higher risk of developing heart failure due to anthracyclines than those who do not have these risk factors. Therefore, dyslipidemia has the potential to worsen anthracycline-induced cardiotoxicity [127].
The ESC recommendations support the utilization of the newly established Heart Failure Association (HFA) and International Cardio-Oncology Society (ICOS) baseline risk stratification score [128]. The HFA-ICOS score was formulated following a systematic review of the literature concerning seven categories of potentially cardiotoxic cancer treatments, including anthracycline chemotherapy and HER2-targeted therapies [46].
The likelihood of developing cardiovascular toxicity associated with cancer treatment (CRT-CVT) in patients who have had a potentially cardiotoxic therapy is directly proportional to the presence of four types of risk factor categories [128].
1.
Baseline risk evaluated using the HFA-ICOS risk assessment instruments.
This risk evaluation is predicated on the cardiovascular risk factors, prior cardiovascular disease history, cancer history, and previous cancer therapy. This evaluation entails a clinical examination, an electrocardiogram (ECG), and, in instances of a pre-existing cardiovascular disease or cancer therapies associated with a heightened risk of CTR-CVT, the measurement of the cTn, natriuretic peptides (NPs), LVEF, and GLS via echocardiography.
2.
The type and dosage of the oncological treatment
Each chemotherapy agent may present a distinct risk of CTR-CVT, ranging from a negligible to a significantly elevated risk, typically associated with the administered doses, including both individual and cumulative doses (e.g., cumulative doses of anthracyclines), as well as the concurrent or sequential administration of additional oncological therapies (e.g., anthracyclines combined with trastuzumab and radiotherapy).
3.
CRT-CVT occurring with the delivery of treatment
This encompasses nearly all types of cardiovascular injury across various cardiovascular levels, possessing diverse pathophysiological significance and therapeutic relevance.
4.
The evaluation of clinical conditions, echocardiography, and biomarker assessments
This reappraisal should occur in all clinical settings at 12 months and at 3 and 12 months for patients initially classified as high- and very-high risk. The echocardiogram must encompass a comprehensive assessment of the left and right ventricular function and, when feasible, the evaluation of the global longitudinal strain in comparison to baseline data. The clinical assessment must encompass cardiovascular risk factors in accordance with the ESC Guidelines on Prevention. This evaluation should take place at 12 months (and at 3 months for participants previously recognized as high- and extremely-high-risk during therapy) [129].

9. Cardioprotective

As a follow-up, the ESMO released updated guidelines in 2020, which suggest that in patients with a mild asymptomatic LVEF decline, trastuzumab can be continued with an additional cardioprotective therapy. This condition is defined as a decrease in the LVEF of ≥10% from baseline or a decrease to a range of 40–<50% [130]. Cardioprotective therapies, including angiotensin-converting enzyme inhibitors (ACEis), angiotensin receptor blockers (ARBs), and beta-blockers, have been shown to prevent cardiac remodeling and reduce mortality in patients with cardiac dysfunction [131]. The current evidence suggests that this therapy plays an important role in preventing the discontinuation of adjuvant trastuzumab in HER2-positive breast cancer patients [132,133]. In addition, experimental animal studies [26], observational studies [134], and small-scale randomized trials report that this cardioprotective intervention can help maintain the left ventricular function in patients undergoing cardiotoxic chemotherapy [135,136,137,138,139].
A study demonstrated that the use of lisinopril and carvedilol significantly improved the cardiotoxicity-free survival compared to the placebo. In addition, the group receiving this therapy experienced fewer treatment discontinuations. In HER2-positive breast cancer patients undergoing anthracycline and trastuzumab therapy, the use of lisinopril or carvedilol as cardioprotective agents may help reduce the risk of the trastuzumab discontinuation due to cardiotoxicity [132]. Similar findings were reported by Cardinale et al., who demonstrated that enalapril effectively prevents anthracycline-induced cardiomyopathy [140]. Kaya et al. also reported similar benefits with carvedilol [137].
Several studies have also shown that the concurrent administration of candesartan is effective in preventing a decrease in the LVEF after chemotherapy [135,136,140]. The protective effect of candesartan against a decrease in the LVEF persists for up to one year after the completion of doxorubicin chemotherapy, while the protective effect of carvedilol begins to diminish after this period. This cardioprotective effect was observed even at a minimal dose of 4 mg per day. Candesartan demonstrated a lower incidence of early subclinical cardiotoxicity and a superior preservation of the LVEF compared to carvedilol and stronger protection against the LVEF decline. Notably, all patients in the candesartan group who developed early subclinical cardiotoxicity showed a complete recovery by the end of the study, with no significant difference in the LVEF between the beginning and end of the study [140].
The results of a study conducted by Pituskin et al. found that bisoprolol (beta-blockers) was more effective in reducing the decline in the LVEF and allowed more patients to continue trastuzumab therapy without interruption compared to perindopril (ACEi) and the placebo [141]. In contrast, another study conducted by Gulati et al. reported that candesartan (ARB) has a protective effect against the decreased LVEF, while metoprolol (beta-blockers) and the placebo did not show a significant impact on patients receiving epirubicin and trastuzumab [136].
Although the specific agent selection among beta-blockers and ACEis remains debated, several studies show that carvedilol and nebivolol have the best cardioprotective effect on patients undergoing anthracycline therapy [137,142,143]. Current evidence supports the safety and efficacy of neurohormonal agents, including Renin–Angiotensin–Aldosterone System (RAAS) inhibitors and beta-blockers, in mitigating cardiotoxicity. In addition, several studies have also reported a small improvement in the left ventricular function in patients receiving cardioprotective therapy compared to the placebo group [136,144].
In addition to neurohormonal therapies, other strategies such as dexrazoxane have also been explored to prevent anthracycline-induced cardiotoxicity. One such agent, dexrazoxane, has been investigated extensively in both pediatric and adult cancer patients for its ability to reduce the cardiovascular toxicity associated with anthracyclines [145]. Dexrazoxane is believed to exert its protective effects by chelating iron and preventing the formation of the topoisomerase IIβ–anthracycline complex, a key mechanism underlying cardiac damage [146,147]. Although it received approval from the U.S. Food and Drug Administration (FDA) in 1995 and is recommended by several cardiology societies, its use in clinical practice remains limited. The FDA specifically approved dexrazoxane for patients with advanced or metastatic breast cancer who have received a cumulative anthracycline dose of at least 300 mg/m2 [47]. A meta-analysis of nine studies with 2177 patients found that dexrazoxane significantly reduced the risk of clinical heart failure (RR: 0.19) and cardiac events (RR: 0.36), without influencing the oncological outcomes, overall survival, or progression-free survival. However, given the low quality of the evidence, further randomized trials are needed before it can be widely adopted in clinical practice [145].
Complementing the use of cardioprotective agents, modified chemotherapy formulations such as liposomal doxorubicin have also been developed to address anthracycline-associated cardiotoxicity [46]. This formulation involves encapsulating doxorubicin within lipid-based vesicles (liposomes), which modifies its pharmacokinetics and reduces its accumulation in non-target organs like the heart, while enhancing the delivery to tumor sites [148]. This innovative formulation has shown a more favorable toxicity profile compared to conventional anthracyclines. A meta-analysis of 19 clinical trials across both adjuvant and metastatic settings found that the use of liposomal doxorubicin significantly reduces the incidence of cardiotoxicity [149]. Due to its enhanced cardiac safety, liposomal doxorubicin is considered a more suitable option, particularly for patients at a higher risk for cardiac side effects.

10. Discussion and Future Directions

Breast cancer therapy using anthracyclines and trastuzumab has proven to be effective; however, cardiotoxicity remains a major challenge in clinical management. Cardiotoxicity represents a serious complication, as it may disrupt the treatment continuity and significantly reduce the QoL of breast cancer patients [76]. This review highlights that anthracycline-induced cardiotoxicity is dose-dependent and generally irreversible, while trastuzumab-related cardiotoxicity tends to be reversible, yet it remains a leading cause of therapy discontinuation [45].
The cardiotoxic mechanisms of doxorubicin and trastuzumab involve different pathways, yet both contribute to myocardial dysfunction. Doxorubicin induces oxidative stress, iron dysregulation, apoptosis, ferroptosis, and epigenetic alterations; whereas, trastuzumab inhibits HER2 signaling pathways and disrupts the mitochondrial integrity [47,85,86,99,100,101]. These findings are consistent with various studies demonstrating that cardiotoxicity is multifactorial and specific to the chemotherapeutic agent used.
An advanced age, hypertension, obesity, diabetes, and dyslipidemia are key risk factors that increase the likelihood of developing cardiotoxicity [65,118,120,124,125,126,127]. However, there are inconsistencies across studies in how these risk factors are defined and assessed, indicating the need for standardized risk stratification systems such as the HFA-ICOS [46].
Current diagnostic strategies, such as the measurement of GLS, allow for the earlier detection of cardiac dysfunction, although threshold values and clinical implementation still vary [45,47]. In addition, biomarkers such as troponin and natriuretic peptides show a promising predictive value but are not yet routinely used due to the need for further validation [46]. A baseline measurement of cardiac serum biomarkers is essential if changes in these biomarkers are to be used to detect a subclinical cardiac injury during cancer treatment [46]. The study also revealed that when combined with GLS, the troponin I measurement has a negative predictive value of 91% for the possibility of developing cardiotoxicity in the future [56]. The combined use of GLS and cardiac biomarkers may enhance the early detection of cardiotoxicity in patients receiving anti-HER2 therapies, especially in high-risk populations [45,55,150].
The research pinpointed the primary ways in which CTRCD diminishes the QoL for breast cancer patients. Interviews from 12 women with a history of breast cancer and CTRCD showed that those who received chemotherapy often experienced overwhelming fatigue, the mental burden of anxiety, a lack of understanding and acceptance, a lack of knowledge and acknowledgement, and a need for personalized care. The findings indicate that patients experience a heightened disease burden from cardiovascular complications, which adversely affects their physical, social, and psychosocial well-being. Furthermore, their healthcare encounters were often compromised by an insufficient acknowledgement of their concerns and suboptimal professional communication. In response, patients articulated a strong need for more personalized follow-up care. Implementing a tailored, multidisciplinary, and holistic care model holds the potential to substantially improve both patient healthcare experiences and the long-term QoL [151].
Consistent with the prior literature, a study conducted in Romania found a correlation between a decreased LVEF and patient-reported outcomes, such as increased fatigue and reduced physical ability—effects linked to the cardiotoxicity caused by treatment. Among patients with an LVEF below 50%, the average EORTC QLQ-C30 score was 63.17, while those with normal cardiac function (LVEF between 50 and 65%) had a slightly higher average score of 64.28, indicating a comparable quality of life, albeit with more variability. The study further showed that individuals with a lower LVEF (<50%) had somewhat reduced EORTC QLQ-C30 scores compared to those with normal heart function. These findings suggest that cardiotoxicity plays a meaningful role in the decline of the quality of life, even in patients with early-stage breast cancer [152].
While treatment with heart failure medication is linked to a functional improvement, a retrospective study from Japan indicates that the LVEF recovery is often incomplete. Specifically, the mean post-recovery LVEF of 56.9% remained significantly below the baseline value of 64.3%, implying a degree of persistent, irreversible myocardial damage or remodeling induced by anthracyclines. This is further complicated by the fact that the natural course of anthracycline-induced cardiomyopathy (AIC) is not fully understood, though the spontaneous recovery of the left ventricle systolic function has been previously reported. The study aligns with this, as a LVEF recovery without heart failure medication was observed in some patients, particularly those with a less severe initial dysfunction. Given that the mechanisms underlying such a spontaneous recovery are poorly characterized, identifying clinical parameters that can predict this outcome is crucial for effective patient stratification and a timely referral for specialized heart failure management [153]. According to a key observational study, patients who develop cardiotoxicity during treatment with trastuzumab typically exhibit a functional cardiac recovery upon the discontinuation of the therapy. Ewer (2005) reported that this improvement generally manifests within a few months, with an average recovery period of 1.5 months following the drug withdrawal [153,154].
Various cardioprotective therapies have been shown to be effective in preserving the left ventricular function and preventing a chemotherapy-induced cardiac injury. Agents such as lisinopril, enalapril, carvedilol, and candesartan have demonstrated the ability to reduce the risk of treatment discontinuation due to cardiac complications, both by improving cardiac function and preventing further structural damage [131,132,133]. Beyond neurohormonal therapies, dexrazoxane has also shown efficacy in preventing anthracycline-induced cardiotoxicity. However, its clinical use remains limited due to strict regulatory guidelines and concerns about potential oncologic effects; although to date, no strong evidence has supported these concerns [46,147,155,156].
In clinical practice, several challenges remain, such as the insufficient monitoring of the cumulative anthracycline dosage—which significantly increases the risk of cardiac complications—alongside the absence of routine cardiac function assessments and limited patient awareness regarding cardiotoxicity risks. Not all healthcare facilities have standardized protocols for monitoring cardiac function before, during, and after cancer treatment, leading to variability in clinical practices across centers. Some patients may be unable or unwilling to adhere to the recommended cardiac monitoring schedules due to financial barriers, low health literacy, or logistical issues such as limited access to transportation. Furthermore, collaboration between oncologists and cardiologists remains suboptimal, often resulting in delays in the detection and management of cardiotoxicity. To address these challenges, a comprehensive and systematic approach is needed, including routine cardiac monitoring, enhanced patient education regarding cardiotoxicity risks and prevention, and stronger interdisciplinary coordination among healthcare providers.
To achieve this, further research is required, including clinical trials to evaluate the effectiveness of various cardioprotective agents, such as ACE inhibitors, ARBs, beta-blockers, and dexrazoxane, in order to determine the most effective preventive therapies. Epidemiological studies and population-based data are also essential to better understand the incidence and specific risk factors for cardiotoxicity among different patient populations. Additionally, multidisciplinary collaboration models—such as cardio-oncology clinics and telemedicine—can improve patient access to cardiac monitoring. Through these various efforts, it is hoped that strategies for the prevention and management of cardiotoxicity can be optimized, allowing patients to continue receiving the full benefit of cancer therapy without an increased risk of cardiovascular complications.

11. Conclusion

Cardiotoxicity is a major challenge in breast cancer therapy, particularly with the use of anthracyclines and trastuzumab. Although their mechanisms differ, both agents can lead to cardiac dysfunction that affects treatment continuity. The early detection through LVEF and GLS assessments—as well as cardiac biomarkers, alongside the use of cardioprotective therapies such as ACE inhibitors, ARBs, beta-blockers, and dexrazoxane—has proven to be effective in preventing cardiac damage and preserving the left ventricular function.
However, clinical implementation remains limited due to the lack of standardized protocols and multidisciplinary collaboration. Therefore, an integrated approach is needed, including individualized risk stratification, routine cardiac monitoring, and the development of cardio-oncology services, supported by further research to optimize the prevention and management of cardiotoxicity and ensure that patients receive the full benefits of cancer therapy without increasing the risk of cardiac complications.

Author Contributions

Conceptualization, A.E.F. and N.O.W.; formal analysis, A.E.F.; investigation, N.O.W.; resources, A.E.F. and N.O.W.; writing—original draft preparation, A.E.F. and N.O.W.; writing—review and editing, A.A., K.A., L.A., R.A.P., and Z.Z.; visualization, A.E.F. and N.O.W.; supervision, A.A., K.A., L.A., R.A.P., and Z.Z.; project administration, A.E.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by 2024 Research, Community Service, and Innovation (PPMI 2024) Program, School of Pharmacy, Institut Teknologi Bandung (grant no. 31/IT1.C10/SK-KU/2024).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
GlobocanGlobal Burden of Cancer Study
LVEFLeft Ventricular Ejection Fraction
QoLQuality of Life
WHOWorld Health Organization
ASIRAge-Standardized Incidence Rate
EREstrogen Receptor
PRProgesterone Receptor
HER2Human Epidermal Growth Factor Receptor 2
TNBCTriple-Negative Breast Cancer
ASCOAmerican Society of Clinical Oncology
DFSDisease-Free Survival
OSOverall Survival
NOAHNeoadjuvant Herceptin
EFSEvent-Free Survival
IDFSInvasive Disease-Free Survival
T-DM1Trastuzumab Emtansine
pCRPathologic Complete Response
ASEAmerican Society of Echocardiography
EACVIEuropean Society of Cardiovascular Imaging
ESCEuropean Society of Cardiology
CTRCDCancer Therapy-Related Cardiovascular Dysfunction
EFEjection Fraction
GLSGlobal Longitudinal Strain
cTnCardiac Troponin
hs-cTnHigh-Sensitivity Cardiac Troponin
TICTrastuzumab-Induced Cardiotoxicity
ROSReactive Oxygen Species
TOP2BTopoisomerase II Beta
dsDNADouble-Strand DNA
SQ-DOXSemiquinone Doxorubicin
O2Superoxide Anions
H2O2Hydrogen Peroxide
SODSuperoxide Dismutase
OH•Hydroxyl Radicals
TRZTrastuzumab
NRG1Neuregulin-1
NONitric Oxide
MAPKMitogen-Activated Protein Kinase
PI3KPhosphoinositide 3-Kinases
mPTPMitochondrial Permeability Transition Pores
HFAHeart Failure Association
ICOSInternational Cardio-Oncology Society
CRT-CVTCardiovascular Toxicity Associated with Cancer Treatment
ECGElectrocardiogram
NPsNatriuretic Peptides
ACEiAngiotensin-Converting Enzyme Inhibitor
ARBAngiotensin Receptor Blocker
RAASRenin–Angiotensin–Aldosterone System
FDAFood and Drug Administration
AICAnthracycline-Induced Cardiomyopathy

References

  1. Hayes, D.F.; Lippman, M.E. Breast cancer. In Harrison’s Principles of Internal Medicine, 21st ed.; Loscalzo, J., Fauci, A., Eds.; McGraw-Hill Education: New York, NY, USA, 2022; Chapter 75; pp. 1–26. [Google Scholar]
  2. Global Cancer Observatory: Cancer Today. Available online: https://gco.iarc.who.int/today (accessed on 8 January 2025).
  3. Peto, R.; Davies, C. Comparisons between different polychemotherapy regimens for early breast cancer: Meta-analyses of long-term outcome among 100,000 women in 123 randomised trials. Lancet 2012, 379, 432–444. [Google Scholar] [CrossRef]
  4. Rosa, G.M.; Gigli, L. Update on cardiotoxicity of anti-cancer treatments. Eur. J. Clin. Investig. 2016, 46, 264–284. [Google Scholar] [CrossRef]
  5. Ewer, M.S.; Lenihan, D.J. Is trastuzumab associated with adverse cardiac effects in patients with breast cancer? Nat. Clin. Pract. Oncol. 2008, 5, 192–193. [Google Scholar] [CrossRef] [PubMed]
  6. Wang, S.Y.; Long, J.B. Cardiovascular events, early discontinuation of trastuzumab, and their impact on survival. Breast Cancer Res. Treat. 2014, 146, 411–419. [Google Scholar] [CrossRef] [PubMed]
  7. Perez, E.A.; Rodeheffer, R. Clinical Cardiac Tolerability of Trastuzumab. J. Clin. Oncol. 2004, 22, 322–329. [Google Scholar] [CrossRef] [PubMed]
  8. Kementerian Kesehatan Republik Indonesia. Keputusan Menteri Kesehatan Republik Indonesia Nomor HK.01.07/MENKES/414/2018 tentang Pedoman Nasional Pelayanan Kedokteran Tata Laksana Kanker Payudara; Kementerian Kesehatan Republik Indonesia: Jakarta, Indonesia, 2018; pp. 1–11. Available online: https://kemkes.go.id/app_asset/file_content_download/170115800565659c7587dac8.09513662.pdf (accessed on 8 January 2025).
  9. World Health Organization. Breast Cancer. Available online: https://www.who.int/news-room/fact-sheets/detail/breast-cancer (accessed on 8 January 2025).
  10. Mubarik, S.; Yu, Y. Epidemiological and sociodemographic transitions of female breast cancer incidence, death, case fatality and DALYs in 21 world regions and globally, from 1990 to 2017: An Age-Period-Cohort Analysis. J. Adv. Res. 2022, 37, 185–196. [Google Scholar] [CrossRef]
  11. Bray, F.; Ferlay, J. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef]
  12. Skol, A.D.; Sasaki, M.M. The genetics of breast cancer risk in the post-genome era: Thoughts on study design to move past BRCA and towards clinical relevance. Breast Cancer Res. 2016, 18, 99. [Google Scholar] [CrossRef]
  13. Sun, Y.S.; Zhao, Z. Risk Factors and Preventions of Breast Cancer. Int. J. Biol. Sci. 2017, 13, 1387–1397. [Google Scholar] [CrossRef]
  14. Shaath, H.; Elango, R. Molecular Classification of Breast Cancer Utilizing Long Non-Coding RNA (lncRNA) Transcriptomes Identifies Novel Diagnostic lncRNA Panel for Triple-Negative Breast Cancer. Cancers 2021, 13, 5350. [Google Scholar] [CrossRef]
  15. Zubair, M.; Wang, S. Advanced Approaches to Breast Cancer Classification and Diagnosis. Front. Pharmacol. 2021, 11, 632079. [Google Scholar] [CrossRef] [PubMed]
  16. Owens, M.A.; Horten, B.C. HER2 Amplification Ratios by Fluorescence In Situ Hybridization and Correlation with Immunohistochemistry in a Cohort of 6556 Breast Cancer Tissues. Clin. Breast Cancer 2004, 5, 63–69. [Google Scholar] [CrossRef]
  17. Slamon, D.J.; Clark, G.M. Human Breast Cancer: Correlation of Relapse and Survival with Amplification of the HER-2/neu Oncogene. Science 1987, 235, 177–182. [Google Scholar] [CrossRef]
  18. Howlader, N.; Cronin, K.A. Differences in Breast Cancer Survival by Molecular Subtypes in the United States. Cancer Epidemiol. Biomark. Prev. 2018, 27, 619–626. [Google Scholar] [CrossRef]
  19. Ramamoorthy, P.; Dandawate, P. Celastrol and Triptolide Suppress Stemness in Triple Negative Breast Cancer: Notch as a Therapeutic Target for Stem Cells. Biomedicines 2021, 9, 482. [Google Scholar] [CrossRef] [PubMed]
  20. Breast Cancer Treatment (PDQ®)—Health Professional Version. Available online: https://www.cancer.gov/types/breast/hp/breast-treatment-pdq (accessed on 8 January 2025).
  21. Bradley, R.; Braybrooke, J. Trastuzumab for early-stage, HER2-positive breast cancer: A meta-analysis of 13,864 women in seven randomised trials. Lancet Oncol. 2021, 22, 1139–1150. [Google Scholar] [CrossRef]
  22. Carvalho, C.; Santos, R. Doxorubicin: The Good, the Bad and the Ugly Effect. Curr. Med. Chem. 2009, 16, 3267–3285. [Google Scholar] [CrossRef]
  23. McGowan, J.V.; Chung, R. Anthracycline Chemotherapy and Cardiotoxicity. Cardiovasc. Drugs Ther. 2017, 31, 63–75. [Google Scholar] [CrossRef]
  24. Szuławska, A.; Czyz, M. Molecular mechanisms of anthracyclines action. Postep. Hig. Med. Dosw. 2006, 60, 78–100. [Google Scholar]
  25. Hahn, V.S.; Lenihan, D.J. Cancer Therapy–Induced Cardiotoxicity: Basic Mechanisms and Potential Cardioprotective Therapies. J. Am. Heart Assoc. 2014, 3, e000665. [Google Scholar] [CrossRef] [PubMed]
  26. Iqbal, N.; Iqbal, N. Human Epidermal Growth Factor Receptor 2 (HER2) in Cancers: Overexpression and Therapeutic Implications. Mol. Biol. Int. 2014, 2014, 852748. [Google Scholar] [CrossRef]
  27. Le, X.F.; Pruefer, F. HER2-targeting Antibodies Modulate the Cyclin-dependent Kinase Inhibitor p27Kip1 via Multiple Signaling Pathways. Cell Cycle 2005, 4, 87–95. [Google Scholar] [CrossRef]
  28. Vu, T.; Claret, F.X. Trastuzumab: Updated Mechanisms of Action and Resistance in Breast Cancer. Front. Oncol. 2012, 2, 62. [Google Scholar] [CrossRef]
  29. Smith, I.; Procter, M. 2-year follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer: A randomised controlled trial. Lancet 2007, 369, 29–36. [Google Scholar] [CrossRef]
  30. Romond, E.H.; Perez, E.A. Trastuzumab plus Adjuvant Chemotherapy for Operable HER2-Positive Breast Cancer. N. Engl. J. Med. 2005, 353, 1673–1684. [Google Scholar] [CrossRef]
  31. Piccart-Gebhart, M.J.; Procter, M. Trastuzumab after Adjuvant Chemotherapy in HER2-Positive Breast Cancer. N. Engl. J. Med. 2005, 353, 1659–1672. [Google Scholar] [CrossRef] [PubMed]
  32. Jones, S.E.; Savin, M.A. Phase III Trial Comparing Doxorubicin Plus Cyclophosphamide with Docetaxel Plus Cyclophosphamide as Adjuvant Therapy for Operable Breast Cancer. J. Clin. Oncol. 2006, 24, 5381–5387. [Google Scholar] [CrossRef]
  33. Gianni, L.; Eiermann, W. Neoadjuvant chemotherapy with trastuzumab followed by adjuvant trastuzumab versus neoadjuvant chemotherapy alone, in patients with HER2-positive locally advanced breast cancer (the NOAH trial): A randomised controlled superiority trial with a parallel HER2-negative cohort. Lancet 2010, 375, 377–384. [Google Scholar] [CrossRef]
  34. Tolaney, S.M.; Barry, W.T. Seven-year (yr) follow-up of adjuvant paclitaxel (T) and trastuzumab (H) (APT trial) for node-negative, HER2-positive breast cancer (BC). J. Clin. Oncol. 2017, 35, 511. [Google Scholar] [CrossRef]
  35. Gradishar, W.J.; Moran, M.S. Breast Cancer. J. Natl. Compr. Cancer Netw. 2024, 22, 331–357. [Google Scholar] [CrossRef] [PubMed]
  36. Bradley, R.; Braybrooke, J. Aromatase inhibitors versus tamoxifen in premenopausal women with oestrogen receptor-positive early-stage breast cancer treated with ovarian suppression: A patient-level meta-analysis of 7030 women from four randomised trials. Lancet Oncol. 2022, 23, 382–392. [Google Scholar] [CrossRef] [PubMed]
  37. Sledge, G.W.; Toi, M. MONARCH 2: Abemaciclib in Combination with Fulvestrant in Women with HR+/HER2− Advanced Breast Cancer Who Had Progressed While Receiving Endocrine Therapy. J. Clin. Oncol. 2017, 35, 2875–2884. [Google Scholar] [CrossRef]
  38. Sledge, G.W.; Toi, M. The Effect of Abemaciclib Plus Fulvestrant on Overall Survival in Hormone Receptor–Positive, ERBB2-Negative Breast Cancer That Progressed on Endocrine Therapy—MONARCH 2. JAMA Oncol. 2020, 6, 116. [Google Scholar] [CrossRef]
  39. Johnston, S.R.D.; Harbeck, N. Abemaciclib Combined with Endocrine Therapy for the Adjuvant Treatment of HR+, HER2−, Node-Positive, High-Risk, Early Breast Cancer (monarchE). J. Clin. Oncol. 2020, 38, 3987–3998. [Google Scholar] [CrossRef]
  40. Tutt, A.N.J.; Garber, J.E. Adjuvant Olaparib for Patients with BRCA1—Or BRCA2—Mutated Breast Cancer. N. Engl. J. Med. 2021, 384, 2394–2405. [Google Scholar] [CrossRef]
  41. Verma, S.; Miles, D. Trastuzumab Emtansine for HER2-Positive Advanced Breast Cancer. N. Engl. J. Med. 2012, 367, 1783–1791. [Google Scholar] [CrossRef]
  42. Schmid, P.; Cortes, J. Event-free Survival with Pembrolizumab in Early Triple-Negative Breast Cancer. N. Engl. J. Med. 2022, 386, 556–567. [Google Scholar] [CrossRef]
  43. Albini, A.; Pennesi, G. Cardiotoxicity of Anticancer Drugs: The Need for Cardio-Oncology and Cardio-Oncological Prevention. JNCI J. Natl. Cancer Inst. 2010, 102, 14–25. [Google Scholar] [CrossRef]
  44. Seidman, A.; Hudis, C. Cardiac Dysfunction in the Trastuzumab Clinical Trials Experience. J. Clin. Oncol. 2002, 20, 1215–1221. [Google Scholar] [CrossRef] [PubMed]
  45. Plana, J.C.; Galderisi, M. 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. 2014, 27, 911–939. [Google Scholar] [CrossRef]
  46. Lyon, A.R.; López-Fernández, T. 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. [Google Scholar] [CrossRef]
  47. Zamorano, J.L.; Lancellotti, P. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines. Pol. Heart J. (Kardiol. Pol.) 2016, 74, 1193–1233. [Google Scholar] [CrossRef]
  48. Florescu, M.; Cinteza, M. Chemotherapy-induced Cardiotoxicity. Maedica 2013, 8, 59–67. [Google Scholar]
  49. Pai, V.B.; Nahata, M.C. Cardiotoxicity of Chemotherapeutic Agents. Drug Saf. 2000, 22, 263–302. [Google Scholar] [CrossRef] [PubMed]
  50. Shakir, D. Chemotherapy Induced Cardiomyopathy: Pathogenesis, Monitoring and Management. J. Clin. Med. Res. 2009, 11, 8–12. [Google Scholar] [CrossRef]
  51. Doherty, J.U.; Kort, S. ACC/AATS/AHA/ASE/ASNC/HRS/SCAI/SCCT/SCMR/STS 2019 Appropriate Use Criteria for Multimodality Imaging in the Assessment of Cardiac Structure and Function in Nonvalvular Heart Disease. J. Am. Coll. Cardiol. 2019, 73, 488–516. [Google Scholar] [CrossRef]
  52. Gripp, E.A.; Oliveira, G.E. Global Longitudinal Strain Accuracy for Cardiotoxicity Prediction in a Cohort of Breast Cancer Patients During Anthracycline and/or Trastuzumab Treatment. Arq. Bras. Cardiol. 2018, 110, 140–150. [Google Scholar] [CrossRef]
  53. Cardinale, D.; Colombo, A. Trastuzumab-Induced Cardiotoxicity: Clinical and Prognostic Implications of Troponin I Evaluation. J. Clin. Oncol. 2010, 28, 3910–3916. [Google Scholar] [CrossRef]
  54. Cardinale, D.; Sandri, M.T. Prognostic Value of Troponin I in Cardiac Risk Stratification of Cancer Patients Undergoing High-Dose Chemotherapy. Circulation 2004, 109, 2749–2754. [Google Scholar] [CrossRef] [PubMed]
  55. Ky, B.; Putt, M. Early Increases in Multiple Biomarkers Predict Subsequent Cardiotoxicity in Patients with Breast Cancer Treated with Doxorubicin, Taxanes, and Trastuzumab. J. Am. Coll. Cardiol. 2014, 63, 809–816. [Google Scholar] [CrossRef] [PubMed]
  56. Sawaya, H.; Sebag, I.A. Assessment of Echocardiography and Biomarkers for the Extended Prediction of Cardiotoxicity in Patients Treated with Anthracyclines, Taxanes, and Trastuzumab. Circ. Cardiovasc. Imaging 2012, 5, 596–603. [Google Scholar] [CrossRef] [PubMed]
  57. Dicato, M.A.; Van, C.E. Breast Cancer. In Side Effects of Medical Cancer Therapy Prevention and Treatment, 2nd ed.; Springer: London, UK, 2013; pp. 29–117. [Google Scholar] [CrossRef]
  58. Ewer, M.S.; Tan-Chiu, E. Reversibility of Trastuzumab Cardiotoxicity: Is the Concept Alive and Well? J. Clin. Oncol. 2007, 25, 5532–5533. [Google Scholar] [CrossRef] [PubMed]
  59. Smith, L.A.; Cornelius, V.R. Cardiotoxicity of anthracycline agents for the treatment of cancer: Systematic review and meta-analysis of randomised controlled trials. BMC Cancer 2010, 10, 337. [Google Scholar] [CrossRef]
  60. Curigliano, G.; Cardinale, D. Cardiotoxicity of anticancer treatments: Epidemiology, detection, and management. CA Cancer J. Clin. 2016, 66, 309–325. [Google Scholar] [CrossRef]
  61. Lotrionte, M.; Biondi-Zoccai, G. Review and Meta-Analysis of Incidence and Clinical Predictors of Anthracycline Cardiotoxicity. Am. J. Cardiol. 2013, 112, 1980–1984. [Google Scholar] [CrossRef]
  62. von Hoff, D.D.; Layard, M.W. Risk factors for doxorubicin-induced congestive heart failure. Ann. Intern. Med. 1979, 91, 710–717. [Google Scholar] [CrossRef]
  63. Ewer, M.S.; Ewer, S.M. Cardiotoxicity of anticancer treatments: What the cardiologist needs to know. Nat. Rev. Cardiol. 2015, 12, 547–558. [Google Scholar] [CrossRef] [PubMed]
  64. Timm, K.N.; Tyler, D.J. The Role of AMPK Activation for Cardioprotection in Doxorubicin-Induced Cardiotoxicity. Cardiovasc. Drugs Ther. 2020, 34, 255–269. [Google Scholar] [CrossRef]
  65. Bowles, E.J.A.; Wellman, R. Risk of Heart Failure in Breast Cancer Patients After Anthracycline and Trastuzumab Treatment: A Retrospective Cohort Study. JNCI J. Natl. Cancer Inst. 2012, 104, 1293–1305. [Google Scholar] [CrossRef] [PubMed]
  66. Chen, T.; Xu, T. Risk of cardiac dysfunction with trastuzumab in breast cancer patients: A meta-analysis. Cancer Treat. Rev. 2011, 37, 312–320. [Google Scholar] [CrossRef]
  67. Hamirani, Y.; Fanous, I. Anthracycline- and trastuzumab-induced cardiotoxicity: A retrospective study. Med. Oncol. 2016, 33, 82. [Google Scholar] [CrossRef]
  68. Oikonomou, E.K.; Kokkinidis, D.G. Assessment of Prognostic Value of Left Ventricular Global Longitudinal Strain for Early Prediction of Chemotherapy-Induced Cardiotoxicity. JAMA Cardiol. 2019, 4, 1007. [Google Scholar] [CrossRef]
  69. Cocco, L.D.; Chiaparini, A.F. Global Longitudinal Strain for the Early Detection of Chemotherapy-Induced Cardiotoxicity: A Systematic Review and Meta-analysis. Clin. Oncol. 2022, 34, 514–525. [Google Scholar] [CrossRef]
  70. Sandoval, Y.; Apple, F.S. High-Sensitivity Cardiac Troponin and the 2021 AHA/ACC/ASE/CHEST/SAEM/SCCT/SCMR Guidelines for the Evaluation and Diagnosis of Acute Chest Pain. Circulation 2022, 146, 569–581. [Google Scholar] [CrossRef] [PubMed]
  71. Sorodoc, V.; Sirbu, O. The Value of Troponin as a Biomarker of Chemotherapy-Induced Cardiotoxicity. Life 2022, 12, 1183. [Google Scholar] [CrossRef]
  72. Martel, S.; Maurer, C. Breast cancer treatment-induced cardiotoxicity. Expert. Opin. Drug Saf. 2017, 16, 1021–1038. [Google Scholar] [CrossRef]
  73. Zagami, P.; Trapani, D. Cardiotoxicity of Agents Used in Patients with Breast Cancer. JCO Oncol. Pr. 2024, 20, 38–46. [Google Scholar] [CrossRef] [PubMed]
  74. Curigliano, G.; Cardinale, D. Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO Clinical Practice Guidelines. Ann. Oncol. 2012, 23, 155–166. [Google Scholar] [CrossRef] [PubMed]
  75. Sardesai, S.; Sukumar, J. Clinical impact of interruption in adjuvant Trastuzumab therapy in patients with operable HER-2 positive breast cancer. Cardio-Oncology 2020, 6, 26. [Google Scholar] [CrossRef] [PubMed]
  76. Esteban-Fernández, A.; Carvajal Estupiñan, J.F.; Gavira-Gómez, J.J.; Pernas, S.; Moliner, P.; Garay, A.; Sánchez-González, Á.; Fernández-Rozas, I.; González-Costello, J. Clinical Profile and Prognosis of a Real-World Cohort of Patients with Moderate or Severe Cancer Therapy-Induced Cardiac Dysfunction. Front. Cardiovasc. Med. 2021, 8, 721080. [Google Scholar] [CrossRef] [PubMed]
  77. Russell, S.D.; Blackwell, K.L. Independent Adjudication of Symptomatic Heart Failure with the Use of Doxorubicin and Cyclophosphamide Followed by Trastuzumab Adjuvant Therapy: A Combined Review of Cardiac Data from the National Surgical Adjuvant Breast and Bowel Project B-31 and the North Central Cancer Treatment Group N9831 Clinical Trials. J. Clin. Oncol. 2010, 28, 3416–3421. [Google Scholar] [CrossRef]
  78. Goldhirsch, A.; Gelber, R.D. 2 years versus 1 year of adjuvant trastuzumab for HER2-positive breast cancer (HERA): An open-label, randomised controlled trial. Lancet 2013, 382, 1021–1028. [Google Scholar] [CrossRef]
  79. Perez, E.A.; Suman, V.J. 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. [Google Scholar] [CrossRef] [PubMed]
  80. Tan-Chiu, E.; Yothers, G. Assessment of Cardiac Dysfunction in a Randomized Trial Comparing Doxorubicin and Cyclophosphamide Followed by Paclitaxel, With or Without Trastuzumab as Adjuvant Therapy in Node-Positive, Human Epidermal Growth Factor Receptor 2–Overexpressing Breast Cancer: NSABP B-31. J. Clin. Oncol. 2005, 23, 7811–7819. [Google Scholar] [CrossRef] [PubMed]
  81. Neugut, A.I.; Hillyer, G.C. Non-initiation and early discontinuation of adjuvant trastuzumab in women with localized HER2-positive breast cancer. Breast Cancer 2014, 21, 780–785. [Google Scholar] [CrossRef]
  82. Solzbach, U.; Beuter, M. Left atrial intramural hematoma after percutaneous coronary intervention. Int. J. Cardiol. 2010, 141, e37–e38. [Google Scholar] [CrossRef]
  83. Yu, A.F.; Yadav, N.U. Trastuzumab interruption and treatment-induced cardiotoxicity in early HER2-positive breast cancer. Breast Cancer Res. Treat. 2015, 149, 489–495. [Google Scholar] [CrossRef] [PubMed]
  84. Gong, I.Y.; Verma, S. Long-term cardiovascular outcomes and overall survival of early-stage breast cancer patients with early discontinuation of trastuzumab: A population-based study. Breast Cancer Res. Treat. 2016, 157, 535–544. [Google Scholar] [CrossRef] [PubMed]
  85. Qiu, Y.; Jiang, P. Anthracycline-induced cardiotoxicity: Mechanisms, monitoring, and prevention. Front. Cardiovasc. Med. 2023, 10, 1242596. [Google Scholar] [CrossRef]
  86. Rawat, P.S.; Jaiswal, A. Doxorubicin-induced cardiotoxicity: An update on the molecular mechanism and novel therapeutic strategies for effective management. Biomed. Pharmacother. 2021, 139, 111708. [Google Scholar] [CrossRef]
  87. Nishi, M.; Wang, P. Cardiotoxicity of Cancer Treatments: Focus on Anthracycline Cardiomyopathy. Arter. Thromb. Vasc. Biol. 2021, 41, 2648–2660. [Google Scholar] [CrossRef] [PubMed]
  88. Russo, M.; Della, S.A. Metabolic Aspects of Anthracycline Cardiotoxicity. Curr. Treat. Options Oncol. 2021, 22, 18. [Google Scholar] [CrossRef]
  89. Zhang, S.; Liu, X. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat. Med. 2012, 18, 1639–1642. [Google Scholar] [CrossRef]
  90. Sawicki, K.T.; De Jesus, A. Iron Metabolism in Cardiovascular Disease: Physiology, Mechanisms, and Therapeutic Targets. Circ. Res. 2023, 132, 379–396. [Google Scholar] [CrossRef]
  91. Atallah, E.; Kantarjian, H. In reply to “Cardiotoxicity of the cancer therapeutic agent imatinib mesylate”. Nat. Med. 2007, 13, 14. [Google Scholar] [CrossRef]
  92. Kerkelä, R.; Grazette, L. Cardiotoxicity of the cancer therapeutic agent imatinib mesylate. Nat. Med. 2006, 12, 908–916. [Google Scholar] [CrossRef]
  93. Christidi, E.; Brunham, L.R. Regulated cell death pathways in doxorubicin-induced cardiotoxicity. Cell Death Dis. 2021, 12, 339. [Google Scholar] [CrossRef] [PubMed]
  94. Lu, L.; Wu, W. Adriamycin-induced autophagic cardiomyocyte death plays a pathogenic role in a rat model of heart failure. Int. J. Cardiol. 2009, 134, 82–90. [Google Scholar] [CrossRef]
  95. Ferreira, A.; Cunha-Oliveira, T. Altered mitochondrial epigenetics associated with subchronic doxorubicin cardiotoxicity. Toxicology 2017, 390, 63–73. [Google Scholar] [CrossRef] [PubMed]
  96. Song, R.; Yang, Y. HDAC6 inhibition protects cardiomyocytes against doxorubicin-induced acute damage by improving α-tubulin acetylation. J. Mol. Cell Cardiol. 2018, 124, 58–69. [Google Scholar] [CrossRef]
  97. Boen, H.M.; Cherubin, M. Circulating MicroRNA as Biomarkers of Anthracycline-Induced Cardiotoxicity. JACC CardioOncol。 2024, 6, 183–199. [Google Scholar] [CrossRef] [PubMed]
  98. ElZarrad, M.K.; Mukhopadhyay, P. Trastuzumab Alters the Expression of Genes Essential for Cardiac Function and Induces Ultrastructural Changes of Cardiomyocytes in Mice. PLoS ONE 2013, 8, e79543. [Google Scholar] [CrossRef]
  99. Hideho, U.; Oikawa, A. Neuregulin Receptor ErbB2 Localization at T-tubule in Cardiac and Skeletal Muscle. J. Histochem. Cytochem. 2005, 53, 87–91. [Google Scholar] [CrossRef] [PubMed]
  100. Lin, M.; Xiong, W. The Research Progress of Trastuzumab-Induced Cardiotoxicity in HER-2-Positive Breast Cancer Treatment. Front. Cardiovasc. Med. 2022, 8, 821663. [Google Scholar] [CrossRef]
  101. Mohan, N.; Shen, Y. Trastuzumab, but Not Pertuzumab, Dysregulates HER2 Signaling to Mediate Inhibition of Autophagy and Increase in Reactive Oxygen Species Production in Human Cardiomyocytes. Mol. Cancer Ther. 2016, 15, 1321–1331. [Google Scholar] [CrossRef] [PubMed]
  102. Vermeulen, Z.; Segers, V.F.M. ErbB2 signaling at the crossing between heart failure and cancer. Basic Res. Cardiol. 2016, 111, 60. [Google Scholar] [CrossRef]
  103. De Keulenaer, G.W.; Doggen, K. The Vulnerability of the Heart as a Pluricellular Paracrine Organ. Circ. Res. 2010, 106, 35–46. [Google Scholar] [CrossRef]
  104. Lemmens, K.; Doggen, K. Role of Neuregulin-1/ErbB Signaling in Cardiovascular Physiology and Disease. Circulation 2007, 116, 954–960. [Google Scholar] [CrossRef]
  105. Rupert, C.E.; Coulombe, K.L.K. The Roles of Neuregulin-1 in Cardiac Development, Homeostasis, and Disease. Biomark. Insights 2015, 10, 1–9. [Google Scholar] [CrossRef]
  106. Geissler, A.; Ryzhov, S. Neuregulins: Protective and reparative growth factors in multiple forms of cardiovascular disease. Clin. Sci. 2020, 134, 2623–2643. [Google Scholar] [CrossRef] [PubMed]
  107. Crone, S.A.; Zhao, Y.Y. ErbB2 is essential in the prevention of dilated cardiomyopathy. Nat. Med. 2002, 8, 459–465. [Google Scholar] [CrossRef] [PubMed]
  108. Özcelik, C.; Erdmann, B. Conditional mutation of the ErbB2 (HER2) receptor in cardiomyocytes leads to dilated cardiomyopathy. Proc. Natl. Acad. Sci. USA 2002, 99, 8880–8885. [Google Scholar] [CrossRef]
  109. Gordon, L.I.; Burke, M.A. Blockade of the erbB2 Receptor Induces Cardiomyocyte Death through Mitochondrial and Reactive Oxygen Species-dependent Pathways. J. Biol. Chem. 2009, 284, 2080–2087. [Google Scholar] [CrossRef]
  110. Ravingerová, T.; Matejíková, J. Differential role of PI3K/Akt pathway in the infarct size limitation and antiarrhythmic protection in the rat heart. Mol. Cell Biochem. 2007, 297, 111–120. [Google Scholar] [CrossRef]
  111. Rochette, L.; Guenancia, C. Anthracyclines/trastuzumab: New aspects of cardiotoxicity and molecular mechanisms. Trends Pharmacol. Sci. 2015, 36, 326–348. [Google Scholar] [CrossRef]
  112. Zeglinski, M.; Ludke, A. Trastuzumab-induced cardiac dysfunction: A “dual-hit”. Exp. Clin. Cardiol. 2011, 16, 70–74. [Google Scholar]
  113. Yang, S.; Liu, G. Targeting the Ras/Raf/MEK/ERK pathway in hepatocellular carcinoma. Oncol. Lett. 2017, 13, 1041–1047. [Google Scholar] [CrossRef]
  114. Yoon, S.; Seger, R. The extracellular signal-regulated kinase: Multiple substrates regulate diverse cellular functions. Growth Factors 2006, 24, 21–44. [Google Scholar] [CrossRef]
  115. Xia, W.; Mullin, R.J. Anti-tumor activity of GW572016: A dual tyrosine kinase inhibitor blocks EGF activation of EGFR/erbB2 and downstream Erk1/2 and AKT pathways. Oncogene 2002, 21, 6255–6263. [Google Scholar] [CrossRef]
  116. Bronte, E.; Bronte, G. Cardiotoxicity mechanisms of the combination of BRAF-inhibitors and MEK-inhibitors. Pharmacol. Ther. 2018, 192, 65–73. [Google Scholar] [CrossRef] [PubMed]
  117. Eskens, F.A.L.M. Angiogenesis inhibitors in clinical development; where are we now and where are we going? Br. J. Cancer 2004, 90, 1–7. [Google Scholar] [CrossRef] [PubMed]
  118. Pinder, M.C.; Duan, Z. Congestive Heart Failure in Older Women Treated with Adjuvant Anthracycline Chemotherapy for Breast Cancer. J. Clin. Oncol. 2007, 25, 3808–3815. [Google Scholar] [CrossRef]
  119. Thavendiranathan, P.; Abdel-Qadir, H. Breast Cancer Therapy–Related Cardiac Dysfunction in Adult Women Treated in Routine Clinical Practice: A Population-Based Cohort Study. J. Clin. Oncol. 2016, 34, 2239–2246. [Google Scholar] [CrossRef]
  120. Kim, Y.A.; Cho, H. Doxorubicin-induced heart failure in cancer patients: A cohort study based on the Korean National Health Insurance Database. Cancer Med. 2018, 7, 6084–6092. [Google Scholar] [CrossRef]
  121. Chun, W.B.; Yi, J.E. Early Cardiac Function Monitoring for Detection of Subclinical Doxorubicin Cardiotoxicity in Young Adult Patients with Breast Cancer. J. Breast Cancer 2013, 16, 178. [Google Scholar] [CrossRef] [PubMed]
  122. Calle, E.E.; Kaaks, R. Overweight, obesity and cancer: Epidemiological evidence and proposed mechanisms. Nat. Rev. Cancer 2004, 4, 579–591. [Google Scholar] [CrossRef]
  123. Renehan, A.G.; Tyson, M. Body-mass index and incidence of cancer: A systematic review and meta-analysis of prospective observational studies. Lancet 2008, 371, 569–578. [Google Scholar] [CrossRef]
  124. Guenancia, C.; Lefebvre, A. Obesity as a Risk Factor for Anthracyclines and Trastuzumab Cardiotoxicity in Breast Cancer: A Systematic Review and Meta-Analysis. J. Clin. Oncol. 2016, 34, 3157–3165. [Google Scholar] [CrossRef] [PubMed]
  125. Kaboré, E.G.; Guenancia, C. Association of body mass index and cardiotoxicity related to anthracyclines and trastuzumab in early breast cancer: French CANTO cohort study. PLoS Med. 2019, 16, e1002989. [Google Scholar] [CrossRef]
  126. Cai, Z.; Xie, Q. S100A8/A9 in Myocardial Infarction: A Promising Biomarker and Therapeutic Target. Front. Cell Dev. Biol. 2020, 8, 603902. [Google Scholar] [CrossRef]
  127. Salz, T.; Zabor, E.C. Preexisting Cardiovascular Risk and Subsequent Heart Failure Among Non-Hodgkin Lymphoma Survivors. J. Clin. Oncol. 2017, 35, 3837–3843. [Google Scholar] [CrossRef]
  128. Lyon, A.R.; Dent, S. Baseline cardiovascular risk assessment in cancer patients scheduled to receive cardiotoxic cancer therapies: A position statement and new risk assessment tools from the Cardio-Oncology Study Group of the Heart Failure Association of the European Society of Cardiology in collaboration with the International Cardio-Oncology Society. Eur. J. Heart Fail. 2020, 22, 1945–1960. [Google Scholar] [CrossRef] [PubMed]
  129. Visseren, F.L.J.; Mach, F. 2021 ESC Guidelines on cardiovascular disease prevention in clinical practice. Eur. Heart J. 2021, 42, 3227–3337. [Google Scholar] [CrossRef] [PubMed]
  130. Curigliano, G.; Lenihan, D. Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. Ann. Oncol. 2020, 31, 171–190. [Google Scholar] [CrossRef]
  131. Ponikowski, P.; Voors, A.A. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur. Heart J. 2016, 37, 2129–2200. [Google Scholar] [CrossRef]
  132. Guglin, M.; Krischer, J. Randomized Trial of Lisinopril Versus Carvedilol to Prevent Trastuzumab Cardiotoxicity in Patients with Breast Cancer. J. Am. Coll. Cardiol. 2019, 73, 2859–2868. [Google Scholar] [CrossRef]
  133. Gujral, D.M.; Lloyd, G. Effect of prophylactic betablocker or ACE inhibitor on cardiac dysfunction & heart failure during anthracycline chemotherapy ± trastuzumab. Breast 2018, 37, 64–71. [Google Scholar] [CrossRef] [PubMed]
  134. Seicean, S.; Seicean, A. Effect of Statin Therapy on the Risk for Incident Heart Failure in Patients with Breast Cancer Receiving Anthracycline Chemotherapy. J. Am. Coll. Cardiol. 2012, 60, 2384–2390. [Google Scholar] [CrossRef]
  135. Bosch, X.; Rovira, M. Enalapril and Carvedilol for Preventing Chemotherapy-Induced Left Ventricular Systolic Dysfunction in Patients With Malignant Hemopathies. J. Am. Coll. Cardiol. 2013, 61, 2355–2362. [Google Scholar] [CrossRef]
  136. Gulati, G.; Heck, S.L. Prevention of cardiac dysfunction during adjuvant breast cancer therapy (PRADA): A 2 × 2 factorial, randomized, placebo-controlled, double-blind clinical trial of candesartan and metoprolol. Eur. Heart J. 2016, 37, 1671–1680. [Google Scholar] [CrossRef]
  137. Kaya, M.G.; Ozkan, M. Protective effects of nebivolol against anthracycline-induced cardiomyopathy: A randomized control study. Int. J. Cardiol. 2013, 167, 2306–2310. [Google Scholar] [CrossRef]
  138. Yun, S.; Vincelette, N.D. Cardioprotective role of β-blockers and angiotensin antagonists in early-onset anthracyclines-induced cardiotoxicity in adult patients: A systematic review and meta-analysis. Postgrad. Med. J. 2015, 91, 627–633. [Google Scholar] [CrossRef]
  139. Cardinale, D.; Colombo, A. Prevention of High-Dose Chemotherapy–Induced Cardiotoxicity in High-Risk Patients by Angiotensin-Converting Enzyme Inhibition. Circulation 2006, 114, 2474–2481. [Google Scholar] [CrossRef]
  140. Lee, M.; Chung, W. Candesartan and carvedilol for primary prevention of subclinical cardiotoxicity in breast cancer patients without a cardiovascular risk treated with doxorubicin. Cancer Med. 2021, 10, 3964–3973. [Google Scholar] [CrossRef] [PubMed]
  141. Pituskin, E.; Mackey, J.R.; Koshman, S.; Jassal, D.; Pitz, M.; Haykowsky, M.J.; Thompson, R.; Oudit, G.; Ezekowitz, J.; Paterson, I. Abstract PD5-03: Prophylactic beta blockade preserves left ventricular ejection fraction in HER2-overexpresssing breast cancer patients receiving trastuzumab: Primary results of the MANTICORE randomized controlled trial. Cancer Res. 2021, 76, PD5-03. [Google Scholar] [CrossRef]
  142. El-Shitany, N.A.; Tolba, O.A. Protective effect of carvedilol on adriamycin-induced left ventricular dysfunction in children with acute lymphoblastic leukemia. J. Card. Fail. 2012, 18, 607–613. [Google Scholar] [CrossRef] [PubMed]
  143. Kalay, N.; Basar, E. Protective Effects of Carvedilol Against Anthracycline-Induced Cardiomyopathy. J. Am. Coll. Cardiol. 2006, 48, 2258–2262. [Google Scholar] [CrossRef]
  144. Boekhout, A.H.; Gietema, J.A. Angiotensin II–Receptor Inhibition with Candesartan to Prevent Trastuzumab-Related Cardiotoxic Effects in Patients with Early Breast Cancer. JAMA Oncol. 2016, 2, 1030. [Google Scholar] [CrossRef]
  145. Hasinoff, B.B.; Kuschak, T.I. A QSAR study comparing the cytotoxicity and DNA topoisomerase II inhibitory effects of bisdioxopiperazine analogs of ICRF-187 (dexrazoxane). Biochem. Pharmacol. 1995, 50, 953–958. [Google Scholar] [CrossRef]
  146. Lyu, Y.L.; Kerrigan, J.E. Topoisomerase IIβ–Mediated DNA Double-Strand Breaks: Implications in Doxorubicin Cardiotoxicity and Prevention by Dexrazoxane. Cancer Res. 2007, 67, 8839–8846. [Google Scholar] [CrossRef]
  147. Heidenreich, P.A.; Bozkurt, B. 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. [Google Scholar] [CrossRef] [PubMed]
  148. Batist, G.; Ramakrishnan, G. Reduced Cardiotoxicity and Preserved Antitumor Efficacy of Liposome-Encapsulated Doxorubicin and Cyclophosphamide Compared with Conventional Doxorubicin and Cyclophosphamide in a Randomized, Multicenter Trial of Metastatic Breast Cancer. J. Clin. Oncol. 2001, 19, 1444–1454. [Google Scholar] [CrossRef] [PubMed]
  149. Yamaguchi, N.; Fujii, T. Comparison of cardiac events associated with liposomal doxorubicin, epirubicin and doxorubicin in breast cancer: A Bayesian network meta-analysis. Eur. J. Cancer 2015, 51, 2314–2320. [Google Scholar] [CrossRef] [PubMed]
  150. Burrage, M.K.; Ferreira, V.M. The use of cardiovascular magnetic resonance as an early non-invasive biomarker for cardiotoxicity in cardio-oncology. Cardiovasc. Diagn. Ther. 2020, 10, 610–624. [Google Scholar] [CrossRef]
  151. Koop, Y.; van Zadelhof, N. Quality of life in breast cancer patients with cancer treatment-related cardiac dysfunction: A qualitative study. Eur. J. Cardiovasc. Nurs. 2022, 21, 235–242. [Google Scholar] [CrossRef]
  152. Murg, S.I.; Matiș, L. Cardiotoxicity in Breast Cancer: Impact of Clinical Classifications and Treatment on Heart Health. Cancers 2024, 16, 4281. [Google Scholar] [CrossRef]
  153. Ohtani, K.; Fujino, T. Recovery from left ventricular dysfunction was associated with the early introduction of heart failure medical treatment in cancer patients with anthracycline-induced cardiotoxicity. Clin. Res. Cardiol. 2019, 108, 600–611. [Google Scholar] [CrossRef]
  154. Ewer, M.S.; Vooletich, M.T. Reversibility of Trastuzumab-Related Cardiotoxicity: New Insights Based on Clinical Course and Response to Medical Treatment. J. Clin. Oncol. 2005, 23, 7820–7826. [Google Scholar] [CrossRef]
  155. Macedo, A.V.S.; Hajjar, L. Efficacy of Dexrazoxane in Preventing Anthracycline Cardiotoxicity in Breast Cancer. JACC CardioOncol 2019, 1, 68–79. [Google Scholar] [CrossRef]
  156. Armenian, S.H.; Lacchetti, C. 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. [Google Scholar] [CrossRef]
Figure 1. Doxorubicin-induced cardiotoxicity. In cardiomyocytes, DOX intercalates into DNA and inhibits topoisomerase II beta (TOP II beta), resulting in double-strand DNA (dsDNA) breaks and the activation of the tumor suppressor protein p53, which leads to apoptosis. Additionally, DOX undergoes a one-electron reduction by mitochondrial enzymes such as NADPH cytochrome P450 reductase, forming an unstable semiquinone radical. This semiquinone doxorubicin (SQ-DOX) undergoes redox cycling, generating superoxide anions (O2), which are subsequently converted into hydrogen peroxide (H2O2) either spontaneously or via superoxide dismutase (SOD). In the presence of free iron (Fe2+), H2O2 participates in Fenton reactions, producing highly reactive hydroxyl radicals (OH•). The accumulation of reactive oxygen species (ROS) causes significant oxidative damage to lipids, proteins, and DNA, particularly within the mitochondria. Furthermore, Fe2+ may form DOX–Fe complexes that amplify ROS production and promote the lipid peroxidation of mitochondrial membranes, contributing to mitochondrial dysfunction and triggering ferroptosis—a regulated, iron-dependent form of cell death. Trastuzumab-induced cardiotoxicity. Trastuzumab (TRZ), a monoclonal antibody targeting the extracellular domain of HER2, induces cardiotoxicity by disrupting the physiological HER2 signaling in cardiomyocytes. Under stress or hemodynamic stimulation, cardiac microvascular endothelial cells release neuregulin-1 (NRG1), which binds to HER4 on cardiomyocytes, facilitating HER4 homodimerization or HER4/HER2 heterodimerization. These receptor complexes activate downstream survival signaling pathways, particularly PI3K–Akt and MAPK, which play crucial roles in cell proliferation, mitochondrial integrity, and the protection against oxidative stress. Trastuzumab blocks HER2, thereby inhibiting HER4/HER2 heterodimerization and attenuating NRG1-mediated signaling. Consequently, the PI3K–Akt pathway is suppressed, leading to decreased nitric oxide (NO) production, impaired mitochondrial function, increased ROS accumulation, and ultimately cardiomyocyte apoptosis. The MAPK pathway, which also contributes to mitochondrial stability, is similarly disrupted.
Figure 1. Doxorubicin-induced cardiotoxicity. In cardiomyocytes, DOX intercalates into DNA and inhibits topoisomerase II beta (TOP II beta), resulting in double-strand DNA (dsDNA) breaks and the activation of the tumor suppressor protein p53, which leads to apoptosis. Additionally, DOX undergoes a one-electron reduction by mitochondrial enzymes such as NADPH cytochrome P450 reductase, forming an unstable semiquinone radical. This semiquinone doxorubicin (SQ-DOX) undergoes redox cycling, generating superoxide anions (O2), which are subsequently converted into hydrogen peroxide (H2O2) either spontaneously or via superoxide dismutase (SOD). In the presence of free iron (Fe2+), H2O2 participates in Fenton reactions, producing highly reactive hydroxyl radicals (OH•). The accumulation of reactive oxygen species (ROS) causes significant oxidative damage to lipids, proteins, and DNA, particularly within the mitochondria. Furthermore, Fe2+ may form DOX–Fe complexes that amplify ROS production and promote the lipid peroxidation of mitochondrial membranes, contributing to mitochondrial dysfunction and triggering ferroptosis—a regulated, iron-dependent form of cell death. Trastuzumab-induced cardiotoxicity. Trastuzumab (TRZ), a monoclonal antibody targeting the extracellular domain of HER2, induces cardiotoxicity by disrupting the physiological HER2 signaling in cardiomyocytes. Under stress or hemodynamic stimulation, cardiac microvascular endothelial cells release neuregulin-1 (NRG1), which binds to HER4 on cardiomyocytes, facilitating HER4 homodimerization or HER4/HER2 heterodimerization. These receptor complexes activate downstream survival signaling pathways, particularly PI3K–Akt and MAPK, which play crucial roles in cell proliferation, mitochondrial integrity, and the protection against oxidative stress. Trastuzumab blocks HER2, thereby inhibiting HER4/HER2 heterodimerization and attenuating NRG1-mediated signaling. Consequently, the PI3K–Akt pathway is suppressed, leading to decreased nitric oxide (NO) production, impaired mitochondrial function, increased ROS accumulation, and ultimately cardiomyocyte apoptosis. The MAPK pathway, which also contributes to mitochondrial stability, is similarly disrupted.
Medsci 13 00130 g001
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fitrianti, A.E.; Wardani, N.O.; Astuti, A.; Anggadiredja, K.; Amalia, L.; Putri, R.A.; Zazuli, Z. Cardiotoxicity in Breast Cancer Therapy: Risks, Mechanisms, and Prevention Strategies. Med. Sci. 2025, 13, 130. https://doi.org/10.3390/medsci13030130

AMA Style

Fitrianti AE, Wardani NO, Astuti A, Anggadiredja K, Amalia L, Putri RA, Zazuli Z. Cardiotoxicity in Breast Cancer Therapy: Risks, Mechanisms, and Prevention Strategies. Medical Sciences. 2025; 13(3):130. https://doi.org/10.3390/medsci13030130

Chicago/Turabian Style

Fitrianti, Annisa Eka, Nadea Olyvia Wardani, Astri Astuti, Kusnandar Anggadiredja, Lia Amalia, Risani Andalasia Putri, and Zulfan Zazuli. 2025. "Cardiotoxicity in Breast Cancer Therapy: Risks, Mechanisms, and Prevention Strategies" Medical Sciences 13, no. 3: 130. https://doi.org/10.3390/medsci13030130

APA Style

Fitrianti, A. E., Wardani, N. O., Astuti, A., Anggadiredja, K., Amalia, L., Putri, R. A., & Zazuli, Z. (2025). Cardiotoxicity in Breast Cancer Therapy: Risks, Mechanisms, and Prevention Strategies. Medical Sciences, 13(3), 130. https://doi.org/10.3390/medsci13030130

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop