Fact-checked by Grok 2 weeks ago

Cardiotoxicity

Cardiotoxicity refers to the harmful effects of toxic substances on the heart, encompassing a range of dysfunctions including arrhythmias, myocardial injury, reduced contractility, and . This condition arises primarily from exposure to pharmaceuticals, environmental toxins, or other agents that disrupt cardiac cellular processes, often manifesting as subclinical changes or severe clinical events such as or sudden cardiac death. While cardiotoxicity can occur acutely or chronically, its clinical significance lies in its potential irreversibility and impact on patient outcomes, particularly in therapeutic contexts where benefits must be weighed against cardiac risks. A primary cause of cardiotoxicity is anticancer therapy, including traditional chemotherapeutic agents like (e.g., ) and monoclonal antibodies (e.g., ), as well as emerging immunotherapies such as immune checkpoint inhibitors (e.g., ) that can induce immune-mediated . , for instance, generate (ROS) leading to and in cardiomyocytes, often resulting in type I cardiotoxicity characterized by cumulative, irreversible injury. Beyond , cardiotoxicity affects multiple drug classes, including agents like antipsychotics (e.g., , causing ) and antidepressants (e.g., , prolonging ), as well as anti-infectives (e.g., ) and anti-inflammatory drugs (e.g., ). Risk factors exacerbating these effects include patient age, preexisting , genetic predispositions, and concurrent therapies such as radiation. At the molecular level, cardiotoxicity involves multifaceted mechanisms, prominently including blockade—such as inhibition of the leading to prolongation and arrhythmias—and mitochondrial dysfunction from impaired energy production or excessive ROS. Contractility disruptions occur via altered calcium handling or pathways, while off-target effects like vascular endothelial damage contribute to or ischemia. Type II cardiotoxicity, exemplified by , is typically reversible and non-cumulative, stemming from interference with HER2 signaling rather than direct myocyte toxicity. These pathways highlight the need for vigilant monitoring, such as for left ventricular (LVEF) assessment, to detect early subclinical changes and guide interventions like dose adjustments or cardioprotective agents (e.g., ). The clinical implications of cardiotoxicity extend to and , with over 27 drugs withdrawn from markets due to cardiac risks, underscoring the importance of preclinical screening for liability and mitochondrial effects. In cancer survivors, late-onset cardiotoxicity can emerge years post-treatment, emphasizing long-term surveillance in cardio-oncology programs. Advances in precision medicine, including biomarkers like troponins and genetic profiling, aim to personalize and mitigate these toxicities, balancing therapeutic efficacy with cardiac safety.

Overview

Definition and Scope

Cardiotoxicity refers to the adverse effects on cardiac structure or function induced by exposure to toxic agents, encompassing a range of cardiac pathologies such as , arrhythmias, and . This condition arises from various xenobiotics, including chemotherapeutic drugs, environmental toxins, and other medications, and is often quantified clinically by a decline in left ventricular (LVEF), such as a ≥5% drop to below 55% with heart failure symptoms or a ≥10% drop to below 55% without symptoms. The term highlights the heart's vulnerability to insults that impair its mechanical, electrical, or structural integrity, distinguishing it from general by its specific focus on cardiovascular endpoints. The recognition of cardiotoxicity emerged prominently in the 1960s with the introduction of , such as , as breakthrough anticancer agents derived from soil bacteria, which revolutionized but revealed unexpected cardiac risks through early reports of . Initially termed in 1946 to describe cardiac effects from agents like local anesthetics and , the concept evolved in the to specifically address anthracycline-related toxicities, expanding from acute events during to , long-term manifestations observed years later. This historical shift broadened the understanding from isolated acute insults to a spectrum including delayed-onset damage, influencing modern surveillance protocols in cardio-oncology. The scope of cardiotoxicity includes both direct myocardial toxicity, which involves primary damage to cardiomyocytes—such as and from —and indirect effects, like or induced by agents such as fluoropyrimidines. Damage may be reversible, as seen in functional impairments that recover upon discontinuation of the offending agent, or irreversible, characterized by permanent cardiomyocyte loss and progressive remodeling. Fundamentally, cardiotoxicity exhibits a dose-dependent nature, where risk escalates with cumulative exposure; for instance, thresholds for significant range from 250–300 mg/m² for equivalents, though this varies by specific agent and patient-specific factors including age, genetic predispositions, and comorbidities.

Epidemiology and Risk Factors

Cardiotoxicity affects approximately 10-20% of cancer survivors receiving cardiotoxic therapies, with the incidence rising due to improved and expanded use of such treatments in an aging population. In a retrospective analysis of 96 cancer patients, the incidence was reported as 12.5%, highlighting variability across settings but underscoring the substantial burden. This contributes significantly to cardiovascular mortality, accounting for 7-27% of such events in affected individuals. Demographic factors play a key role in susceptibility, with higher risks observed in females, particularly those treated for , and in elderly patients over 65 years. Pre-existing further elevates vulnerability, as evidenced by studies from the showing synergistic effects with therapies like in older adults. For instance, in a cohort of elderly patients with , use increased congestive risk by 29%, compounded by age and prior cardiac conditions. Major risk factors include cumulative dose thresholds for , where doses exceeding 300 mg/m² are associated with substantially increased risk of . Genetic predispositions, such as the HER2 Ile655Val polymorphism, also heighten susceptibility to trastuzumab-induced cardiotoxicity in HER2-positive patients. Comorbidities like and mellitus independently amplify this risk, with showing a hazard ratio of 1.8 when combined with . Emerging trends indicate a growing incidence of cardiotoxicity beyond , driven by environmental exposures such as , which independently increases cardiovascular risk in cancer patients. Data from registries like the CARDIOTOX registry reveal a 37.5% cardiotoxicity rate during follow-up in high-risk cohorts, emphasizing the need for ongoing surveillance amid these shifts.

Pathophysiology

Cellular and Molecular Mechanisms

Cardiotoxicity arises from multiple interconnected cellular and molecular pathways that impair cardiomyocyte function and survival, primarily triggered by chemotherapeutic agents such as . These mechanisms encompass , altered calcium , mitochondrial impairment, and inflammatory signaling, each contributing to progressive cardiac damage through distinct yet overlapping processes. Oxidative stress represents a central pathway in cardiotoxicity, where toxic agents like generate (ROS) via redox cycling involving NAD(P)H oxidoreductases, leading to of cell membranes, protein oxidation, and DNA strand breaks in cardiomyocytes. In specifically, inhibition of IIβ (TOP2β) by drugs such as forms a drug-enzyme-DNA cleavage complex, resulting in persistent double-strand DNA breaks, particularly in , which amplifies ROS production and promotes cardiomyocyte death independent of canonical iron-mediated ROS pathways. Disruption of calcium handling further exacerbates cardiotoxicity by interfering with excitation-contraction coupling in cardiomyocytes. inhibit the Ca²⁺-ATPase (), reducing Ca²⁺ reuptake into the and prolonging cytosolic Ca²⁺ elevation, while also promoting leakiness to increase spontaneous Ca²⁺ release. This leads to [Ca²⁺]ᵢ dysregulation, often through direct inhibition of the Na⁺/Ca²⁺ exchanger (NCX) by or their metabolites, impairing Ca²⁺ extrusion and leading to cytosolic overload. The calcium flux imbalance can be conceptualized as: \Delta [\ce{Ca^{2+}}]_i = J_{\ce{L-type}} + J_{\ce{RyR}} - J_{\ce{[SERCA](/page/SERCA)}} - J_{\ce{NCX (forward)}} where J_{\ce{NCX (forward)}} diminishes under inhibition, thereby sustaining elevated [Ca²⁺]ᵢ and predisposing to arrhythmias. Mitochondrial dysfunction is a key downstream consequence, with ROS and direct effects inhibiting complexes (particularly complexes I and III), causing electron leakage, reduced proton gradient, and ATP depletion in cardiomyocytes. This impairment triggers the opening, facilitating release into the , which activates the intrinsic pathway via and executioner caspases, culminating in . Inflammatory cascades amplify these damages by activating the NF-κB pathway in cardiomyocytes and cardiac fibroblasts, translocating to the nucleus to upregulate proinflammatory genes, including those encoding cytokines such as TNF-α. Elevated TNF-α promotes further ROS generation, , and extracellular matrix remodeling, driving through fibroblast activation and collagen deposition. Mechanisms vary by agent, highlighting direct versus off-target effects; for instance, primarily induce ROS-dependent and TOP2β-mediated damage, whereas exerts cardiotoxicity through HER2 blockade, disrupting neuregulin-1/ signaling to impair mitochondrial and induce release without substantial ROS involvement.

Types of Cardiac Damage

Cardiotoxicity manifests in various forms of structural and functional cardiac injury, broadly classified by the affected cardiac components and the nature of the damage. These injuries range from myocardial dysfunction to rhythm disturbances and vascular complications, often detected through clinical, , and assessments. Systolic dysfunction represents a primary type of cardiotoxic injury, characterized by reduced left ventricular , typically below 50%, which can progress to overt if untreated. This form often arises from direct myocyte damage, leading to impaired contractility; early stages may be reversible with , but advanced cases result in chronic remodeling and . Subtypes include type I systolic dysfunction, which is dose-dependent and irreversible, involving histological changes such as myofibrillar loss and cytoplasmic in cardiomyocytes, and type II, which is non-dose-dependent and generally reversible upon discontinuation of the offending agent. Arrhythmias constitute another critical category of cardiac damage, encompassing both bradyarrhythmias and tachyarrhythmias that disrupt normal electrical conduction. Bradyarrhythmias, such as or , often stem from beta-adrenergic blockade or enhanced , while tachyarrhythmias include , , and accelerated idioventricular rhythms triggered by ion channel perturbations. A particularly dangerous subtype involves prolongation, which predisposes to and , reflecting abnormalities. Vascular toxicity from cardiotoxic exposures primarily involves and hemodynamic alterations, leading to conditions like , , or . Endothelial damage promotes and , which can cause acute ischemia or chronic vascular remodeling; for instance, inhibition of pathways may induce and increase thrombotic risk. , in turn, results in myocardial ischemia mimicking acute coronary syndromes. Other forms of cardiac damage include pericardial , valvular abnormalities, and , each presenting distinct histological and functional impairments. Pericardial involvement often features or due to inflammatory or hemorrhagic processes, while valvular disease manifests as thickening and regurgitation from changes. arises from interstitial , limiting diastolic filling; common histological findings across these include contraction band and myocardial lesions, indicating myocyte injury. The progression of cardiotoxic damage follows acute or timelines, with acute effects emerging within days of exposure—such as arrhythmias or transient ischemia—and manifestations developing over months to years, including progressive systolic dysfunction or . Biomarkers like elevated cardiac levels signal early myocyte injury in both phases, enabling timely detection before overt symptoms arise; for example, elevation often precedes decline in systolic damage, where contribute to initial cellular stress.

Etiology

Chemotherapeutic Agents

Chemotherapeutic agents, particularly those used in , are a leading cause of cardiotoxicity due to their direct impact on cardiac cells through mechanisms such as . These drugs, including , tyrosine kinase inhibitors, and monoclonal antibodies, can induce a spectrum of cardiac injuries ranging from left ventricular dysfunction to overt , often in a dose-dependent manner. The risk is heightened in combination regimens, necessitating careful dosing and surveillance to balance anticancer efficacy with cardiac safety. , such as , are among the most notorious for inducing cardiotoxicity, primarily through cumulative dose-related . Clinical guidelines recommend limiting the cumulative dose of to 450-550 mg/m² to minimize severe cardiac events. The risk of significantly increases above 250 mg/m², with myocardial changes evident on in over 90% of patients exceeding 240 mg/m². Tyrosine kinase inhibitors (TKIs), exemplified by , frequently cause cardiovascular adverse effects including and QT prolongation. occurs in approximately 20-30% of patients treated with , driven by receptor inhibition leading to . QT prolongation, observed in up to 4.4% of cases with receptor TKIs, arises from disruptions, increasing risk. Monoclonal antibodies like target HER2 but carry substantial cardiotoxicity risks, especially in combination with . The incidence of reaches up to 27% when is combined with such as , due to synergistic impairment of cardiac repair pathways. In the NSABP B-31 trial, cardiac events occurred in 4.0% of patients receiving after and , compared to 1.3% in the control arm without . Other chemotherapeutic classes also contribute to cardiotoxicity, though less commonly. can cause acute hemorrhagic myocarditis, particularly at high doses used in conditioning regimens, with endothelial damage leading to myocardial hemorrhage and high mortality if untreated. Taxanes, such as , are associated with bradyarrhythmias, including asymptomatic in up to 30% of patients without pre-existing cardiac risk factors, resulting from interference. Monitoring guidelines emphasize baseline and serial to detect early left ventricular declines in patients receiving these agents. For - and trastuzumab-based therapies, assessments are recommended before treatment initiation, every 3 months during therapy, and periodically post-treatment, as supported by trial data like NSABP B-31 showing sustained cardiac risks years later.

Non-Chemotherapeutic Medications

Non-chemotherapeutic medications encompass a broad range of prescription drugs used in everyday clinical practice, such as cardiovascular agents, antimicrobials, immunosuppressants, and others, which can induce cardiotoxicity through mechanisms including arrhythmias, , QT prolongation, and exacerbation of . These toxicities often arise from direct effects on cardiac channels, vascular , or secondary consequences like imbalances and fluid retention, posing risks particularly in patients with preexisting cardiovascular conditions. Cardiovascular drugs like , a used for and , carry a high risk of due to its narrow of 0.5-2 ng/mL, beyond which it can cause life-threatening arrhythmias such as or fibrillation by increasing myocardial excitability and delaying . Similarly, beta-agonists like isoproterenol, employed in acute or as a diagnostic tool, stimulate beta-1 and beta-2 adrenergic receptors, leading to and increased myocardial oxygen demand, which may precipitate ischemia or arrhythmias in susceptible individuals. Among antimicrobials, , an antifungal agent, primarily causes through renal and tubular damage, resulting in electrolyte disturbances such as that secondarily strain the heart by promoting arrhythmias or worsening fluid overload in patients with compromised cardiac function. Fluoroquinolones, such as levofloxacin used for bacterial infections, have been associated with an increased risk of or , with FDA analysis indicating approximately a twofold higher incidence compared to other antibiotics, particularly in patients with risk factors like or advanced age. Immunosuppressants like cyclosporine, commonly prescribed post-transplant to prevent rejection, induce in up to 50% or more of renal transplant patients through enhanced endothelin-1 production, which promotes and sodium retention, thereby elevating cardiac and risk of . Other classes include antipsychotics such as , which prolongs the —especially at intravenous doses exceeding 35 mg/day—potentially leading to and sudden cardiac death by blocking channels. Nonsteroidal drugs (NSAIDs), widely used for and , cause sodium and fluid retention by inhibiting prostaglandin-mediated renal , exacerbating symptoms and increasing hospitalization risk in affected patients. Risk stratification for these toxicities involves assessing drug interactions, such as those between statins and certain non-chemotherapeutic agents (e.g., antifungals or ), which can elevate levels via inhibition, heightening risk with secondary cardiac implications; FDA Adverse Event Reporting System data underscores these interactions as frequent contributors to reported cardiotoxic events. Monitoring serum levels, electrolytes, and ECGs, alongside avoiding in high-risk groups, is essential to mitigate these effects.

Environmental Toxins and Exposures

Environmental toxins encompass a range of non-pharmacological substances from industrial, occupational, and natural sources that can induce cardiotoxicity through mechanisms such as , , and direct myocardial damage. These exposures often occur involuntarily, leading to chronic or acute cardiovascular effects including , arrhythmias, and . Heavy metals like lead and mercury are prominent cardiotoxicants, particularly in occupational settings. lead exposure is linked to and , with blood lead levels exceeding 10 μg/dL associated with increased cardiovascular risk through and impaired signaling. Mercury exposure promotes arrhythmias via autonomic dysfunction and reduced , contributing to coronary heart disease and . Industrial chemicals pose significant risks through poisoning or prolonged contact. causes hypoxia-induced by binding to with high affinity, reducing oxygen delivery and directly impairing myocardial function. exposure, common in manufacturing, leads to , which secondarily affects the heart through anemia-related ischemia and potential direct cardiac abnormalities like arrhythmias. Air pollution, especially fine (PM2.5), exacerbates cardiotoxicity via and . Long-term exposure to PM2.5 in urban areas increases risk by 10-20%, as evidenced by cohort studies showing associations with and . Natural toxins from biological sources can directly damage cardiac tissue. Snake venoms, such as that of , induce myocardial through cardiotoxic components like phospholipases, leading to acute infarction-like presentations. Aflatoxins, mycotoxins contaminating in humid regions, cause cardiac and oxidative damage, contributing to in exposed populations. Occupational exposures heighten risks, particularly in where or occurs frequently. Workers in such environments face elevated cardiotoxicity from cumulative lead and mercury burdens, with long-term studies like extensions of the linking environmental pollutants to increased cardiovascular events.

Recreational and Abused Substances

Recreational and abused substances pose substantial risks to cardiovascular health, primarily through acute hemodynamic alterations and chronic structural damage. These effects are particularly pronounced in young adults engaging in , where illicit drugs like , amphetamines, and opioids contribute to a growing burden of cardiac emergencies. Cocaine, a potent , induces acute cardiotoxicity via , mediated by alpha-adrenergic stimulation and increased endothelin-1 release, which reduces myocardial oxygen supply and precipitates (ACS). This mechanism elevates the risk of up to sevenfold, even in individuals with normal , as evidenced by studies showing 6% of cases linked to recent use resulting in . Chronically, repeated exposure leads to myocardial from catecholamine toxicity and , contributing to ; among long-term users, up to 71% exhibit , including reduced and . Amphetamines and methamphetamines similarly exert cardiotoxic effects through excessive catecholamine release, causing acute and via and increased myocardial demand. These changes heighten the risk of arrhythmias, such as and QT prolongation, which account for a 27% increased incidence of sudden cardiac death among users. Chronic methamphetamine use is associated with and accelerated , with mechanisms involving and mitochondrial dysfunction; globally, amphetamine-type stimulants affect over 51 million users aged 15-64, amplifying population-level risks. Opioid abuse, particularly in overdose scenarios, results in and due to mu-receptor agonism, compounded by respiratory depression that induces and secondary cardiac stress. This can precipitate acute events like or arrest, with synthetic opioids further risking QT prolongation and . While direct myocardial toxicity is less common than with stimulants, hemodynamic instability during withdrawal or intoxication contributes to , including from injection practices. Other substances, such as anabolic-androgenic steroids (AAS), promote through activation and fluid retention, leading to systolic and diastolic dysfunction; users display elevated left ventricular mass (111±61 g/m² versus 89±18 g/m² in non-users) and increased coronary plaque volume, correlating with lifetime use duration. () induces serotonin-mediated and , potentially causing rhythm disturbances, , and , with findings revealing pathological myocardial changes in fatal cases. Epidemiological data highlight rising cardiac complications from these substances, particularly among young adults; for instance, drug-related visits involving illicit stimulants and opioids have surged, with cardiovascular manifestations like ACS and arrhythmias comprising a notable fraction—historical Drug Abuse Warning Network (DAWN) reports from 2011 indicated over 1.5 million annual drug-related visits, 22% requiring admission, and subsequent analyses showing 4-17% of cocaine-associated admissions involving ventricular arrhythmias or . Recent trends underscore increased presentations in this demographic, driven by polysubstance abuse and the .

Clinical Presentation

Symptoms and Signs

Cardiotoxicity manifests through a spectrum of acute and chronic symptoms and signs, often reflecting underlying cardiac stress from various etiologies such as chemotherapeutic agents. Acute symptoms typically include , dyspnea, and , which may arise shortly after exposure to cardiotoxic substances. Observable signs in these cases frequently involve or , signaling immediate hemodynamic instability. Chronic manifestations of cardiotoxicity often present as progressive symptoms, including fatigue, , and , which can develop months to years following initial exposure. Many cases remain , particularly in early stages, with silent myocardial ischemia occurring in approximately 15% of patients receiving 5-fluorouracil, underscoring the importance of clinical vigilance. Arrhythmia-specific presentations in cardiotoxicity may include syncope due to or other life-threatening rhythms, alongside electrocardiographic changes such as ST-segment elevation or depression. and irregular heart rhythms are common patient-reported experiences in these scenarios. The nature of symptoms can vary by subtype of cardiotoxicity; reversible forms, often linked to transient myocardial dysfunction, exhibit short-lived symptoms that resolve upon discontinuation of the offending agent, whereas irreversible damage leads to persistent or progressive decline in cardiac function. For instance, systolic dysfunction from may contribute to dyspnea in symptomatic patients. Patient functional status in cardiotoxicity-related is commonly assessed using the New York Heart Association (NYHA) classification, ranging from Class I (no limitation in physical activity) to Class IV (symptoms at rest with inability to carry out any physical activity). Symptomatic cardiotoxicity is often defined by the presence of NYHA Class III or IV symptoms alongside clinical signs of .

Diagnostic Evaluation

Diagnostic evaluation of cardiotoxicity involves a approach to detect and quantify cardiac injury, primarily through , biomarkers, , and select advanced tests, guided by established protocols for and serial . Echocardiography serves as the gold standard for assessing left ventricular (LVEF), enabling early detection of systolic dysfunction with serial measurements recommended every 3 months in high-risk patients undergoing cardiotoxic therapies. Cardiac (MRI) complements echocardiography by providing detailed evaluation of myocardial through late gadolinium enhancement, which is particularly useful for identifying subclinical structural changes not visible on standard imaging. Biomarker analysis plays a key role in risk stratification and early identification of cardiotoxicity, with elevated N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels exceeding 300 pg/mL indicating potential and warranting further investigation. Cardiac elevation signals acute myocardial injury, often preceding overt functional decline and serving as a sensitive marker for ongoing cardiotoxic effects. Electrocardiography, including Holter monitoring, is employed to detect arrhythmias such as those prompted by symptoms like , providing ambulatory rhythm assessment over 24-48 hours to capture intermittent events associated with cardiotoxic agents. Speckle-tracking enhances detection of subclinical dysfunction by measuring global longitudinal strain, which exhibits high sensitivity (up to 80%) for early cardiotoxicity changes before LVEF reduction. Advanced diagnostic tests are reserved for complex cases; endomyocardial biopsy offers definitive histological confirmation of cardiotoxicity through direct myocardial sampling, though it is rarely performed due to its invasiveness and procedural risks. Genetic testing identifies susceptibility variants, such as those in pathways, to predict individual risk of cardiotoxicity from therapies like , though it is not yet routine in clinical practice. Guidelines from the (ASCO) and (ESC) emphasize baseline cardiovascular evaluation prior to initiating potentially cardiotoxic treatments, followed by protocol-driven follow-up using and biomarkers to monitor for declines in LVEF or strain, with specificity for early detection approaching 70-90% in validated cohorts.

Management and Prevention

Acute Treatment Approaches

Acute treatment of cardiotoxicity focuses on rapid stabilization to mitigate life-threatening complications such as , arrhythmias, and hemodynamic instability. Initial interventions prioritize supportive measures to address acute cardiac , including to maintain saturation above 92% in hypoxemic patients and diuretics such as for fluid overload manifesting as . In cases of acute with reduced (HFrEF), inotropes like are administered intravenously at doses of 2-20 mcg/kg/min to enhance contractility and improve , particularly when low indicates severe systolic dysfunction. These supportive strategies align with broader acute guidelines adapted for contexts. Agent-specific antidotes are employed when applicable to counteract direct toxic effects. For anthracycline extravasation, dexrazoxane serves as a chelating agent that binds intracellular iron, thereby reducing reactive oxygen species formation and preventing severe tissue damage; it is administered intravenously at 1000 mg/m² within 6 hours of extravasation, followed by additional doses on subsequent days. In digitalis toxicity, digoxin immune Fab (digoxin-specific antibody fragments) is the first-line antidote, neutralizing free digoxin and reversing life-threatening arrhythmias or hyperkalemia; indications include serum digoxin levels ≥10 ng/mL or severe manifestations like ventricular tachycardia, with rapid administration yielding 50-90% clinical improvement within 45 minutes. Arrhythmia management in acute cardiotoxicity requires prompt intervention to prevent hemodynamic collapse. For , is a preferred antiarrhythmic, administered as a 150 mg IV bolus followed by infusion, due to its efficacy in suppressing tachyarrhythmias without exacerbating underlying . protocols follow guidelines for unstable rhythms, with immediate correction of electrolytes such as and magnesium to mitigate associated with prolongation from cardiotoxic agents. Temporary pacing may be indicated for bradycardias refractory to atropine. Discontinuation of the offending agent is a cornerstone of acute management, with immediate cessation recommended upon confirmation of moderate to severe cardiotoxicity, such as an absolute decrease in left ventricular of >10% from baseline to below 50%. Admission to the is warranted for patients exhibiting hemodynamic instability, , or <30%, where invasive monitoring and mechanical support may be required. Outcomes of acute interventions vary by toxicity type, with type II cardiotoxicity (e.g., from trastuzumab) demonstrating high reversibility; approximately 84% of cases show left ventricular ejection fraction recovery within 1.5 months following discontinuation and heart failure therapy initiation, as evidenced in clinical cohorts. Randomized trials, such as the PRADA study, support early pharmacologic intervention with ACE inhibitors and beta-blockers, achieving reversal in a majority of subclinical cases without compromising oncologic efficacy.

Long-Term Management and Monitoring

Long-term management of cardiotoxicity emphasizes preventing cardiac remodeling and optimizing heart function through targeted pharmacotherapy. Angiotensin-converting enzyme (ACE) inhibitors, such as , and beta-blockers, like , are cornerstone therapies for patients with established left ventricular systolic dysfunction (LVSD). Enalapril is typically initiated at 2.5 mg twice daily and titrated every 3–6 days to a target dose of 10 mg twice daily (20 mg total daily), provided systolic blood pressure remains above 90 mm Hg and renal function is stable (creatinine <2.5 mg/dL). Similarly, carvedilol starts at 6.25 mg twice daily, with titration every 3–6 days to a target of 25 mg twice daily (50 mg total), avoiding bradycardia below 60 beats per minute or atrioventricular block. These agents, when combined, have demonstrated efficacy in halting LV remodeling and improving ejection fraction in chemotherapy-induced cardiotoxicity, with mean achieved doses of approximately 17 mg/day for enalapril and 25 mg/day for carvedilol in clinical trials. Device therapy plays a critical role in managing high-risk complications in patients with persistent LVSD. Implantable cardioverter-defibrillators (ICDs) are recommended for individuals at elevated risk of life-threatening arrhythmias, such as sustained ventricular tachycardia, particularly when left ventricular ejection fraction (LVEF) is below 35–40% despite optimal medical therapy. Cardiac resynchronization therapy (CRT) is indicated for those with dyssynchrony, evidenced by prolonged QRS duration (>150 ms) and LVEF <35%, to improve ventricular synchrony and reduce hospitalizations. Device implantation should be considered only in patients with a life expectancy exceeding 1 year, balancing cancer prognosis with cardiac benefits. Ongoing surveillance is essential to detect subclinical progression and guide adjustments in care. Annual to assess LVEF and global longitudinal strain, combined with monitoring (e.g., high-sensitivity and B-type ), is advised for long-term survivors, with more frequent evaluations (every 3–6 months) for high-risk cases. modifications, including supervised exercise programs, are integrated to enhance cardiac ; these typically involve aerobic (e.g., 30–45 minutes, 3–5 sessions weekly) and resistance exercises, improving functional capacity and attenuating LVEF decline without increasing adverse events. A multidisciplinary approach in specialized cardio-oncology clinics facilitates coordinated care, involving oncologists, cardiologists, and specialists to monitor and modify cancer treatments as needed, such as switching to less cardiotoxic agents (e.g., from to non-anthracycline regimens). improves significantly with early intervention; studies indicate better survival among patients receiving timely and compared to untreated cases, as evidenced by follow-up data from breast cancer trials.

Prevention Strategies

Prevention of cardiotoxicity begins with comprehensive prior to initiating potentially cardiotoxic therapies. Pre-treatment cardiac evaluation typically includes a thorough history and to identify preexisting cardiovascular conditions, along with baseline assessments such as (ECG), echocardiography for left ventricular (LVEF), and biomarkers like cardiac and B-type (BNP) to establish a reference for monitoring. These evaluations help stratify patients into low-, medium-, or high-risk categories based on factors like age, cumulative dose exposure, and comorbidities, guiding personalized treatment plans. Genetic screening for polymorphisms, such as those in the 2D6 () gene, is emerging as a tool to predict susceptibility to cardiotoxicity from agents like , with poor metabolizers showing higher rates of toxicity in initial studies. Prophylactic agents play a key role in mitigating cardiotoxicity during treatment. , an iron-chelating agent, is co-administered with to reduce and has been shown to decrease the incidence of clinical by approximately 50% in high-risk patients, without compromising antitumor . Statins, such as , provide vascular protection through , anti-inflammatory, and anti-apoptotic mechanisms, with meta-analyses indicating a significant reduction in cardiotoxicity risk (relative risk 0.46) when used prophylactically in patients receiving or . Dose optimization strategies further minimize exposure to cardiotoxic peaks. Liposomal formulations, like pegylated liposomal , encapsulate the drug to reduce systemic peak concentrations and myocardial uptake, allowing cumulative doses exceeding 400 mg/m² with minimal clinically evident cardiotoxicity compared to conventional . Interval adjustments, such as biweekly dosing at reduced amounts (e.g., 40 mg/m² instead of 50 mg/m² every 4 weeks), can further lower toxicity while maintaining efficacy, particularly in relapsed settings. Lifestyle interventions complement pharmacological approaches by addressing modifiable risk factors. Smoking cessation is recommended to reduce oxidative stress and endothelial dysfunction, which exacerbate cardiotoxicity, while blood pressure control through diet and medication prevents hypertensive strain on the heart during therapy. Supervised aerobic exercise programs, tailored for at-risk patients, improve cardiovascular reserve and have been associated with lower rates of LVEF decline in those undergoing cardiotoxic treatments. Regulatory policies and clinical guidelines enforce standardized prevention practices. The U.S. (FDA) issues black-box warnings for agents like and HER2-targeted therapies, highlighting risks and mandating cardiac monitoring to limit cumulative doses. International consensus, such as the European Society for Medical Oncology (ESMO) recommendations, outlines monitoring schedules including baseline and periodic LVEF assessments, risk stratification, and multidisciplinary cardio-oncology consultation to optimize prevention across the cancer care continuum.

References

  1. [1]
    Toward a broader view of mechanisms of drug cardiotoxicity - PMC
    Cardiotoxicity generally results from the simultaneous interruption of key myocardial functions and viabilities. In this section, we focus on adverse drug ...
  2. [2]
    Chemotherapy-induced Cardiotoxicity - PMC - NIH
    Cardiotoxicity is a general term used to define "toxicity that affects the heart" (13). A growing body of researches is now studying cardiovascular events ...
  3. [3]
    Cardiotoxicity of Anticancer Drugs: Molecular Mechanisms and ...
    In this review, we summarize the knowledge regarding the cellular and molecular mechanisms contributing to the onset of cardiotoxicity.
  4. [4]
    Cardiotoxicity: precision medicine with imprecise definitions - PMC
    Jul 25, 2018 · 'Cardiotoxicity' was first coined to describe the cardiac toxicity from local anaesthetics, mercurial diuretics and digitalis in 1946. In the ...
  5. [5]
    Anthracycline Cardiotoxicity | Circulation: Heart Failure
    Mar 15, 2019 · The discovery of anthracyclines in the 1960s was a major breakthrough in oncology. And although the field of cancer therapeutics has ...
  6. [6]
    Anthracycline Cardiotoxicity in Adult Cancer Patients: JACC
    Sep 17, 2024 · Cardiotoxicity occurs at lower doses with smaller anthracycline pools or at higher doses if the patient has predisposing risk factors or ...
  7. [7]
    Current perspectives of cardio-oncology: Epidemiology, adverse ...
    Apr 27, 2023 · As the post-cancer population is growing, an increasing number of patients ... Keywords: cancer treatment; cardio-oncology; cardiotoxicity; ...
  8. [8]
    Cardiotoxicity in Cancer Patients: The Prevalence, Risk Factors, and ...
    May 3, 2024 · As a result, patients receiving high-risk cardiotoxic drugs should be monitored with caution to avoid drug-related cardiotoxicity. Furthermore, ...
  9. [9]
    Incidence of chemotherapy‐related cardiac dysfunction in cancer ...
    Apr 18, 2024 · It was found that cancer patients experience cardiotoxicity outcomes that contribute to 7%−27% of their cardiovascular mortality. , This may be ...
  10. [10]
    Cardiovascular Complications of Cancer Therapy: Incidence ... - JACC
    Jun 9, 2009 · The incidence of cardiac dysfunction ranges from 2% to 7% when trastuzumab is used as monotherapy, 2% to 13% when trastuzumab is used in ...
  11. [11]
    Doxorubicin, Cardiac Risk Factors, and Cardiac Toxicity in Elderly ...
    To our knowledge, our study is the first to show that hypertension, a known risk factor for CHF, increases the risk of doxorubicin-related cardiotoxicity. We ...
  12. [12]
    Anthracycline-Induced Clinical Heart Failure in a Cohort of 607 ...
    A cumulative dose of anthracycline higher than 300 mg/m2 was associated with an increased risk of A-CHF (relative risk, 11.8; 95% confidence interval, 1.6 to ...
  13. [13]
    Influence of the HER2 Ile655Val polymorphism on trastuzumab ...
    Our results support the role of the HER2 655 A>G polymorphism as a genetic marker of trastuzumab-induced cardiotoxicity in HER2-positive breast cancer patients.
  14. [14]
    Risk Factors for Anthracycline-Induced Cardiotoxicity - Frontiers
    Sep 28, 2021 · Hypertension, diabetes mellitus, and obesity are associated with increased risk of anthracycline-induced cardiotoxicity.
  15. [15]
    State-of-the-Art Review Air Pollution in Cardio-Oncology and ...
    Air pollution is an overlooked environmental risk factor for cardio-oncology. •. Air pollution may interact with various common risk factors for cancer and ...
  16. [16]
    the CARDIOTOX registry | European Heart Journal | Oxford Academic
    Feb 4, 2020 · Cardiotoxicity was identified in 37.5% patients during follow-up [95% confidence interval (CI) 34.22–40.8%], 31.6% with mild, 2.8% moderate, and ...Missing: Oncology | Show results with:Oncology<|control11|><|separator|>
  17. [17]
    Identification of the molecular basis of doxorubicin-induced ... - Nature
    Oct 28, 2012 · Doxorubicin is believed to cause dose-dependent cardiotoxicity through redox cycling and the generation of reactive oxygen species (ROS).
  18. [18]
    The effects of doxorubicin on cardiac calcium homeostasis and ...
    This review comprehensively summarizes the effects of DOX on cardiac Ca 2+ homeostasis and contractile function from in vitro, ex vivo, and in vivo studies.
  19. [19]
    Na/Ca exchange and contraction of the heart - PMC - NIH
    Jun 12, 2013 · In this article, we will discuss how NCX can influence contractility in the heart under physiological and pathophysiological conditions.
  20. [20]
    Drug-induced mitochondrial dysfunction and cardiotoxicity - PMC
    The free radicals produced intramitochondrially can directly inactivate the electron transport complexes by interacting with the iron-sulfur cluster, or they ...
  21. [21]
    Inhibition of ErbB2 causes mitochondrial dysfunction in ... - JACC
    The BAX oligomerization was associated with cytochrome c release and caspase activation. These alterations induced mitochondrial dysfunction, a loss of ...Alteration In Bcl-X... · Erbb-2 Inhibition Induces A... · Erbb-2 Inhibition And...
  22. [22]
    Multiple Facets of NF-κB in the Heart | Circulation Research
    Apr 29, 2011 · With chronic inhibition of NF-κB signaling, mice displayed less mortality from heart failure, reduced inflammatory cytokine secretion (IL-1β, IL ...
  23. [23]
    Tumor necrosis factor-α and its role as a mediator in myocardial ...
    Mar 22, 2015 · While NF-κB is activated by the combination of TNF-α and TNFR2, expression of inflammatory cytokines IL-6 and IL-1β is downregulated to ...
  24. [24]
    Comprehensive review of cardiovascular toxicity of drugs and ...
    The aim of this review is to summarize the current knowledge regarding xenobiotic agents and toxins that cause cardiovascular toxicity.
  25. [25]
    Cardiotoxicity of Anticancer Drugs: Molecular Mechanisms and ...
    In this review, we summarize the knowledge regarding the cellular and molecular mechanisms contributing to the onset of cardiotoxicity.Abstract · Introduction · Cardiotoxicity Mechanisms... · Strategies to Reduce...<|control11|><|separator|>
  26. [26]
    Chemotherapy-induced dysfunction - European Society of Cardiology
    Mar 17, 2017 · Although we are going to focus on myocardial damage, cardiotoxicity also produces other heart affections, including pericardial, valvular or ...<|control11|><|separator|>
  27. [27]
    Circulating Cardiovascular Biomarkers in Cancer Therapeutics ...
    Oct 27, 2023 · Trastuzumab‐induced cardiotoxicity: clinical and prognostic implications of troponin I evaluation. J Clin Oncol. 2010;28:3910–3916. Crossref.
  28. [28]
    Troponin: the biomarker of choice for the detection of cardiac injury
    Most studies found a relation between increased troponin levels and mortality.,. Chemotherapy: Cardiotoxicity is a common complication of high-dose chemotherapy ...
  29. [29]
    Anthracycline Cardiotoxicity: Prevalence, Pathogenesis and Treatment
    The family of anthracycline drugs originated in the 1950's with the identification of daunorubicin from the soil bacterium Streptomyces peucetius [1]. In the ...
  30. [30]
    Anthracycline Chemotherapy and Cardiotoxicity - PMC
    Feb 9, 2017 · Anthracycline chemotherapy maintains a prominent role in treating many forms of cancer. Cardiotoxic side effects limit their dosing and improved cancer outcomes
  31. [31]
    Incidence, Diagnosis, and Treatment of Cardiac Toxicity from ...
    Screening with multigated acquisition scan (MUGA) or echocardiogram (ECHO) is recommended to assess cardiac function prior to and during trastuzumab therapy.
  32. [32]
    Anthracycline Toxicity - StatPearls - NCBI Bookshelf - NIH
    Jan 9, 2024 · More than 90% of patients treated with an anthracycline medication manifest myocardial changes on biopsy at cumulative doses above 240 mg/m2, ...Introduction · Epidemiology · Evaluation · Toxicity and Adverse Effect...
  33. [33]
    Cardiotoxicity of Anthracyclines - PMC - NIH
    Mar 18, 2020 · The historical concept that anthracycline-induced cardiotoxicity is irreversible, with a reported mortality rate up to 60% within 2 years of ...
  34. [34]
    Cardiovascular Toxicities Associated with Tyrosine Kinase Inhibitors
    Its use has been linked to arterial ischemic events (13%), cardiac failure (2.2%), hypertension (19%), and QTc prolongation (7%) [50].
  35. [35]
    Cardiovascular toxicity of tyrosine kinase inhibitors during cancer ...
    Feb 3, 2023 · Meta-analysis of VEGFR-TKI related clinical trials demonstrated an incidence of 4.4% of all-grade QT prolongation when compared to non-TKI ...
  36. [36]
    Concurrent administration of trastuzumab and anthracyclines as ...
    However, in the neoadjuvant therapy, trastuzumab administered concurrently with anthracyclines has shown exceedingly low cardiotoxicity [8–11], and expectedly ...
  37. [37]
    The Research Progress of Trastuzumab-Induced Cardiotoxicity in ...
    Furthermore, the incidence of cardiac events reported by NSABP B-31were 1.3% in the control group and 4.0% in the trastuzumab group, with 15.5% of the ...
  38. [38]
    Cyclophosphamide-Induced Cardiomyopathy: A Case Report ... - NIH
    Higher doses of this drug may produce significant cardiac toxicity, including fatal hemorrhagic myocarditis. In this review, we present a case of ...
  39. [39]
    A reassessment of cardiac toxicity associated with Taxol - PubMed
    In patients without significant cardiac risk factors, asymptomatic sinus bradycardia is frequent (approximately 30%). Heart block and conduction abnormalities ...
  40. [40]
    Prevention and Monitoring of Cardiac Dysfunction in Survivors of ...
    Dec 5, 2016 · Echocardiography has been the most widely used method for monitoring cardiac function after chemotherapy. ... NSABP B-31. J Clin Oncol 23:7811- ...
  41. [41]
    A Review on Probable Causes of Cardiotoxicity Caused by ... - NIH
    Dec 6, 2023 · Cardiovascular toxicity is a type of medication toxicity that might show itself as symptoms like palpitations and chest blockage. The absence of ...
  42. [42]
    Amphotericin B - StatPearls - NCBI Bookshelf - NIH
    Feb 28, 2024 · Nephrotoxicity: Renal toxicity correlates with conventional amphotericin B use and can lead to renal failure and the requirement for dialysis.
  43. [43]
    Digoxin - StatPearls - NCBI Bookshelf
    Nov 25, 2024 · Digoxin toxicity is clinically significant, as it can lead to fatal cardiac arrhythmias. The estimated incidence is around 0.8% to 4% of ...
  44. [44]
    Isoproterenol - StatPearls - NCBI Bookshelf
    Isoproterenol is a beta-1 and beta-2 adrenergic receptor agonist resulting in the following: Increased heart rate. Increased heart contractility. Relaxation of ...Continuing Education Activity · Indications · Mechanism of Action · Contraindications
  45. [45]
    Atrioventricular block related to liposomal amphotericin B - PMC
    Jun 6, 2014 · The main adverse reactions associated with amphotericin are nephrotoxicity, electrolyte abnormalities (hypokalaemia, hypomagnesaemia and ...Case... · Figure 2 · Discussion<|separator|>
  46. [46]
    FDA warns about increased risk of ruptures or tears in the aorta blood
    Dec 20, 2018 · In addition, exposure to fluoroquinolones for longer than 14 days was associated with a higher risk of aortic aneurysm or dissection (for 3-14 ...
  47. [47]
    Cyclosporine and posttransplant hypertension - PubMed - NIH
    Prevalence studies suggest that hypertension is present in more than 50% of kidney transplant patients. It is more prevalent in pediatric patients, ...
  48. [48]
    Endothelin-1 plasma levels and hypertension in cyclosporine ...
    Experimental models suggest that endothelin-1 (ET-1) has a significant role in the pathogenesis of cyclosporin A (CyA)-induced hypertension.Missing: incidence | Show results with:incidence
  49. [49]
    Antipsychotics and the QTc Interval During Delirium in the Intensive ...
    Jan 22, 2024 · This prolongation is greater when haloperidol is administered at doses greater than approximately 35 mg/d. With atypical antipsychotics, the ...
  50. [50]
    Non-steroidal anti-inflammatory drugs and risk of heart failure ...
    NSAIDs are known to cause salt and fluid retention which could trigger exacerbation of heart failure (HF). In fact, this possible adverse effect has long been ...
  51. [51]
    Recommendations for Management of Clinically Significant Drug ...
    Oct 17, 2016 · Recommendations for management of clinically significant drug-drug interactions with statins and select agents used in patients with cardiovascular disease.
  52. [52]
    Important safety label changes to cholesterol-lowering statin drugs
    Feb 28, 2012 · Healthcare professionals should report adverse events involving statins to the FDA MedWatch program using the information in the "Contact FDA" ...Safety Announcement · Data Summary
  53. [53]
    Environmental Exposures and Cardiovascular Disease
    Lead, arsenic and cadmium exposure is associated with hypertension, coronary heart disease and cardiovascular mortality.<|separator|>
  54. [54]
    Heavy Metal Poisoning and Cardiovascular Disease - PMC
    Population-based studies on the cardiovascular effects of lead have focused largely on the association with hypertension, a leading risk factor for CVD ...
  55. [55]
    Mechanisms of lead-induced hypertension and cardiovascular ...
    Chronic lead exposure causes HTN and cardiovascular disease by promoting oxidative stress, limiting nitric oxide availability, impairing nitric oxide signaling.
  56. [56]
    Lead Exposure and Cardiovascular Disease—A Systematic Review
    This systematic review evaluates the evidence on the association between lead exposure and cardiovascular end points in human populations.
  57. [57]
    Mercury Exposure and Heart Diseases - PMC - PubMed Central - NIH
    Mercury toxicity is indeed strongly correlated with hypertension, coronary heart disease, myocardial infarction, cardiac arrhythmias, carotid artery obstruction ...
  58. [58]
    Role of Mercury Toxicity in Hypertension, Cardiovascular Disease ...
    The clinical consequences of mercury toxicity include hypertension, CHD, MI, cardiac arrhythmias, sudden death, reduced heart rate variability, increased ...Missing: cardiotoxicity | Show results with:cardiotoxicity
  59. [59]
    Carbon Monoxide Poisoning: Pathogenesis, Management, and ...
    Cardiovascular effects​​ Up to one-third of patients with moderate to severe CO poisoning present with myocardial injury, which may be associated with increased ...
  60. [60]
    ST Elevation Myocardial Infarction Due to Carbon Monoxide Poisoning
    Carbon monoxide is formed as a result of combustion of any carbon compound and can lead to hypoxia in many organs including the brain and the heart.
  61. [61]
    CARDIAC ABNORMALITIES INDUCED BY BENZENE EXPOSURE ...
    Benzene exposure is associated with multiple adverse health effects on the human's biological systems including its cardiac, pulmonary, respiratory, renal, ...Missing: secondary | Show results with:secondary
  62. [62]
    HEALTH EFFECTS - Toxicological Profile for Benzene - NCBI - NIH
    No studies were located regarding cardiovascular effects in humans after inhalation exposure to benzene ... Aplastic anemia is a more severe effect of benzene and ...
  63. [63]
    PM2.5 and cardiovascular diseases: State-of-the-Art review - PMC
    Oct 12, 2023 · PM2.5 is associated with many clinical risk factors for cardiovascular disease, such as hypertension. Studies have shown that both acute and ...
  64. [64]
    Particulate Matter Air Pollution: Effects on the Cardiovascular System
    The ACS study found increased risk of heart failure with PM2.5 exposure, although to a lesser degree than the association with ischemic heart disease.
  65. [65]
    Acute myocardial infarction following a Russell's viper bite
    The direct cardiotoxic effect of snake venom can result in myocarditis and extensive myocardial necrosis. It has been reported in two horses injected with ...
  66. [66]
    Acute aflatoxin B1-induced hepatic and cardiac oxidative damage in ...
    Feeding animals with AF-contaminated food led to oxidative stress, as shown by the significant lipid peroxidation increase and enzymatic antioxidant ...
  67. [67]
    Chemotherapy-related cardiotoxicity and its symptoms in patients ...
    Jun 27, 2024 · The five most common CRCT symptoms were dyspnea, chest pain, peripheral edema, fatigue, and palpitations, which were assessed by patient-reported symptom ...
  68. [68]
    Chemotherapy-Induced Cardiotoxicity: Overview of the Roles ... - NIH
    ... hypotension, tachycardia, arrhythmias, and heart failure [1]. The National Cancer Institute generally defines toxicity that affects the heart as cardiotoxicity.Missing: signs | Show results with:signs
  69. [69]
    Clinical Manifestations, Monitoring, and Prognosis: A Review of ...
    Jun 10, 2022 · Cardiotoxicity is clinically manifested by arrhythmia, decreased cardiac function, or even sudden death. The cardiotoxicity caused by ...
  70. [70]
    Myocardial Ischemia Induced by 5‐Fluorouracil: A Prospective ... - NIH
    Oct 7, 2020 · Implications for Practice. Symptomatic 5‐fluorouracil (5‐FU) cardiotoxicity occurs in 0.6%–19% of patients treated with this drug, but a small ...
  71. [71]
    Cardiotoxicity from Capecitabine Chemotherapy: Prospective Study ...
    Feb 16, 2022 · Major criteria for diagnosing cardiotoxicity at rest were: signs of cardiac ischemia (≥2 mm ST elevation or horizontal ST-segment depression on ...
  72. [72]
    Overview of Oncology: Drug-Induced Cardiac Toxicity - PMC
    The incidence of heart-related issues varies from patient to patient and depends on multiple factors, including the type of medication, dosage, duration of the ...
  73. [73]
    Echocardiographic predictors of symptomatic cardiotoxicity among ...
    Jul 15, 2022 · GLS may predict symptomatic cardiotoxicity and be used to monitor patients on chemotherapy for symptomatic cardiac dysfunction.Missing: incidence | Show results with:incidence
  74. [74]
    Anthracyclines, HER2-targeted therapies and fluoropyrimidines
    Apr 7, 2023 · ... biomarkers and cardiac imaging for monitoring cardiotoxicity in anthracycline-treated patients is suggested [4]. Figure 2 outlines the ...Anthracyclines · Her2-Targeted Therapies · Fluoropyrimidines
  75. [75]
    Practical guidance for echocardiography for cancer therapeutics ...
    The utility of point-of-care biomarkers to detect cardiotoxicity during anthracycline chemotherapy: a feasibility study. J Cardiac Fail. 2016;22:433–438 ...
  76. [76]
    Cardiac MRI in the Assessment of Cardiac Injury and Toxicity From ...
    Nov 1, 2013 · Its most promising application, however, is in the use of myocardial strain and strain rate imaging to detect early cardiac injury.
  77. [77]
    Serial Magnetic Resonance Imaging to Identify Early Stages ... - JACC
    Feb 18, 2019 · The purpose of this study was to identify early doxorubicin-induced cardiotoxicity by serial multiparametric cardiac magnetic resonance (CMR) ...
  78. [78]
    Cardiovascular Considerations Before Cancer Therapy - JACC
    Baseline NT-proBNP is recommended to detect interval change during treatment, which may indicate preclinical cardiotoxicity. •. Elevated baseline troponin is ...
  79. [79]
    [PDF] 2022 ESC Guidelines on cardio-oncology
    805–807 However, evaluating a cardiac response to treatment using a decrease in NT-proBNP levels and New York Heart Association class improvement is still.
  80. [80]
    Impact of the ESC Cardio-Oncology Guidelines Biomarker Criteria ...
    Jan 16, 2024 · This may suggest that cardiac effects of cancer and cardiotoxic cancer therapies are associated with a more pronounced cTnT than cTnI release.
  81. [81]
    P1568 Cardiac biomarkers for the prediction of cardiotoxicity in ...
    Oct 21, 2019 · Cardiac troponin qualifies as a screening test to identify patients at high risk for manifest cardiotoxicity who require referral to cardio ...<|separator|>
  82. [82]
    Value of Holter monitoring in assessing cardiac arrhythmias in ...
    However, very long periods of monitoring may be needed to make a diagnosis in those with only sporadic symptoms. MeSH terms. Adult; Aged; Arrhythmias, Cardiac / ...
  83. [83]
    Diagnostic yield of 24 to 48-hour ambulatory cardiac monitoring in ...
    Patients are instructed to wear the device for a standard of either 24 or 48 h to detect any arrythmia or record cardiac rhythm during a symptomatic event [5].
  84. [84]
    Speckle tracking echocardiography predicts early subclinical ...
    Speckle tracking echocardiography predicts early subclinical anthracycline cardiotoxicity in patients with breast cancer. J Clin Ultrasound. 2017 May;45(4): ...
  85. [85]
    Two-dimensional speckle tracking echocardiography predicts early ...
    Oct 25, 2018 · Two-dimensional speckle tracking echocardiography predicts early subclinical cardiotoxicity associated with anthracycline-trastuzumab ...
  86. [86]
    Use of Myocardial Strain Imaging by Echocardiography for the Early ...
    The reported sensitivity and specificity of GLS to predict cardiotoxicity (Table 3) is likely optimistic, given the small sample sizes and few cardiotoxicity ...State-Of-The-Art Paper · Results · Prognostic Value Of...
  87. [87]
    Endomyocardial biopsy - UpToDate
    Feb 13, 2025 · Outline · - Late AV block, arrhythmias, or refractory HF · - DCM with eosinophilia · - Anthracycline cardiotoxicity · - Restrictive cardiomyopathy ...
  88. [88]
    Current Status of Endomyocardial Biopsy - Mayo Clinic Proceedings
    Endomyocardial biopsy (EMB) is widely used for surveillance of cardiac allograft rejection and for the diagnosis of unexplained ventricular dysfunction.Analysis Of Biopsy Specimens · Indications For Emb · Anthracycline-Induced...
  89. [89]
    Genetics of Cancer Therapy-Associated Cardiotoxicity - PMC - NIH
    Recent studies demonstrate that genetics play a substantial role in susceptibility to development of cardiovascular toxicities from cancer therapies.
  90. [90]
    Genetic Testing in Evaluating Risk of Anthracycline Cardiomyopathy
    May 9, 2023 · Although there are currently no guideline-based recommendations supporting genetic testing to assess risk of cardiac dysfunction before ...Missing: susceptibility | Show results with:susceptibility
  91. [91]
    Genetic factors in the pathogenesis of cardio-oncology
    Aug 5, 2024 · Recent studies indicate that genetics play a significant role in susceptibility to the development of cardiovascular toxicity from cancer therapies.Gstm1 · Top2b-Mediated Dna Damage · Abbreviations
  92. [92]
    The 2022 European Society of Cardiology Cardio-oncology ...
    Apr 21, 2023 · The main focus of the guidelines is to ensure patients can complete their cancer treatment without significant cardiotoxicity and the correct ...Table 2: Myocarditis... · Coronary Artery Disease · Cardiac Arrhythmias
  93. [93]
    Dobutamine - StatPearls - NCBI Bookshelf - NIH
    Feb 23, 2024 · Dobutamine is approved by the US Food and Drug Administration (FDA) for short-term use in patients with decreased contractility due to heart failure or cardiac ...
  94. [94]
    Dexrazoxane - StatPearls - NCBI Bookshelf - NIH
    Jul 17, 2023 · Dexrazoxane is a medication used in the management and treatment of anthracycline-induced cardiotoxicity and extravasation injuries.Continuing Education Activity · Indications · Mechanism of Action · Adverse Effects
  95. [95]
    Digoxin Immune Fab - StatPearls - NCBI Bookshelf
    Jun 22, 2024 · Digoxin immune Fab is an antidote for digoxin toxicity, a condition typically observed in patients with atrial fibrillation and underlying heart failure.Indications · Mechanism of Action · Administration · Monitoring
  96. [96]
    Recognition, Prevention, and Management of Arrhythmias and ...
    Jun 17, 2021 · The purpose of this scientific statement is to summarize the current state of knowledge regarding arrhythmia identification and treatment in patients with ...
  97. [97]
    Severe Cardiac Toxicity Induced by Cancer Therapies Requiring ...
    Sep 3, 2021 · General criteria for ICU admission do not differ from usual indications: the development of CS, hemodynamic instability, need for invasive ...
  98. [98]
    Reversibility of Trastuzumab-Related Cardiotoxicity: New Insights ...
    Nov 1, 2005 · Our data suggest that cardiotoxicity resulting from trastuzumab is largely reversible, unlike the cardiotoxicity associated with anthracyclines, ...Trastuzumab Re-Treatment · Discussion · Authors' Disclosures Of...
  99. [99]
    The PRADA II Randomized Clinical Trial | Circulation
    Anthracycline- and trastuzumab-associated cardiotoxicity may lead to cardiac dysfunction and dose reduction or halt of potentially life-saving adjuvant ...
  100. [100]
    Enalapril and Carvedilol for Preventing Chemotherapy-Induced Left ...
    Combined treatment with enalapril and carvedilol may prevent LVSD in patients with malignant hemopathies treated with intensive chemotherapy.
  101. [101]
  102. [102]
    Management of cancer patients at high and very-high risk of ...
    In patients with HFrEF, HF therapies must be optimized before considering a cardiac implantable device such as ICD or CRT (Table 1). Table 1.
  103. [103]
    Cardio-Oncology Rehabilitation to Manage Cardiovascular ...
    Apr 8, 2019 · This document introduces the concept of cardio-oncology rehabilitation, which includes identification of patients with cancer at high risk for cardiac ...
  104. [104]
    Cardiac Toxicity of Cancer Therapies: Mechanisms, Surveillance ...
    Sep 14, 2025 · Cardiotoxicity refers to a substance's harmful effects on the heart, which can result in cardiomyopathy, heart failure (HF), or a significant ...
  105. [105]
    Monitoring for Chemotherapy-Related Cardiotoxicity in the Form of ...
    Mar 10, 2021 · Cardiotoxicity Monitoring in Patients With Cancer ... incidence of cardiotoxicity is 37.5% in patients receiving high-risk chemotherapy.Serum Biomarkers · Anthracyclines · Anti-Her2 Therapy
  106. [106]
    Strategies for risk stratification and cardiovascular toxicity prevention ...
    Feb 22, 2023 · Statins are another class of drug being explored for their cardioprotective role in cancer patients exposed to cardiotoxic agents and are ...
  107. [107]
    Baseline cardiovascular risk assessment in cancer patients ...
    The assessment occurring prior to the initiation of cancer treatment and in patients without overt CVD or previous cardiotoxicity can be considered a primary ...
  108. [108]
    Role of CYP2D6 Phenotypes in Doxorubicin-Related Cardiotoxicity
    Mar 12, 2025 · This is the first study to analyze cardiotoxicity rates based on CYP2D6 function for patients who received doxorubicin.
  109. [109]
    Dexrazoxane Significantly Reduces Anthracycline-Induced ... - NIH
    Dexrazoxane decreases the rate of cardiotoxicity at any given dose, but does not protect against accumulating late cardiotoxic effects of anthracycline ...Results · Cardiotoxicity Limited By... · Cardiotoxicity Continues To...
  110. [110]
    Statins for the Primary Prevention of Anthracycline Cardiotoxicity - NIH
    Jul 13, 2024 · Statins can prevent anthracycline-induced cardiotoxicity through mechanisms involving antioxidant, anti-inflammatory, and geno-protective ...
  111. [111]
    Statins to mitigate cardiotoxicity in cancer patients treated with ...
    This meta-analysis suggests that there is an association between statin use and decreased risk of cardiotoxicity after anthracycline and/or trastuzumab exposure ...Missing: vascular | Show results with:vascular
  112. [112]
    High Cumulative Doses of Pegylated Liposomal Doxorubicin Are ...
    Cumulative doses of PLD ≧400 mg/m 2 are not associated with clinically evident cardiac toxicity in gynecologic oncology patients.
  113. [113]
    Thirty years from FDA approval of pegylated liposomal doxorubicin ...
    Jan 9, 2025 · Pegylated liposomal doxorubicin (doxil): reduced clinical cardiotoxicity in patients reaching or exceeding cumulative doses of 500 mg/m2.Missing: optimization | Show results with:optimization
  114. [114]
    Biweekly pegylated liposomal doxorubicin in patients with relapsed ...
    There is convincing evidence that the PLD dose can be safely reduced from 50 to 40 mg/m2, but randomized trials are needed to prove that dose-splitting further ...
  115. [115]
    Optimizing Cardiovascular Health in Patients With Cancer
    Mar 26, 2020 · Optimizing the CV health of patients with cancer before starting potentially cardiotoxic therapy may help to prevent the development of cancer ...Cardiovascular Monitoring... · Cardiac Imaging Tools · Prevention Strategies
  116. [116]
    Exercise Therapy and Cardiovascular Toxicity in Cancer | Circulation
    Mar 13, 2018 · In contrast, neither cancer nor treatment with known cardiotoxic regimens is a qualifying condition for exercise rehabilitation in North America ...Current Evidence · During Therapy · Personalized...<|separator|>
  117. [117]
    Cardiovascular warnings common but slower to be issued for FDA ...
    Oct 29, 2021 · Cardiovascular disease was the most common reason for any black box warning (37.2%), with sudden death prompting 11.6% of black box warnings.
  118. [118]
    Management of Cardiac Disease | ESMO
    This ESMO consensus manuscript recommends best practices for the care of cancer patients exposed to potential cardiotoxic therapy.
  119. [119]
    Management of cardiac disease in cancer patients throughout ...
    These ESMO consensus recommendations attempt to summarise best practices for the care of cancer patients exposed to potential cardiotoxic therapy, including ...