Journal List > J Cardiovasc Ultrasound > v.26(1) > 1018512

Kim, Chung, Cho, Kim, Seo, Park, Kim, Lee, Kim, and Youn: Diagnosis, Treatment, and Prevention of Cardiovascular Toxicity Related to Anti-Cancer Treatment in Clinical Practice: An Opinion Paper from the Working Group on Cardio-Oncology of the Korean Society of Echocardiography

Abstract

Cardiovascular (CV) toxicity associated with anti-cancer treatment is commonly encountered and raises critical problems that often result in serious morbidity or mortality. Most cardiac toxicities are related to the cumulative dose of chemotherapy; however, the type of chemotherapy, concomitant agents, and/or conventional CV risk factors have been frequently implicated in CV toxicity. Approximately half of the patients exhibiting CV toxicity receive an anthracycline-based regimen. Therefore, serologic biomarkers or cardiac imagings are important during anti-cancer treatment for early detection and the decision of appropriate management of cardiotoxicity. However, given the difficulty in determining a causal relationship, a multidisciplinary collaborative approach between cardiologists and oncologists is required. In this review, we summarize the CV toxicity and focus on the role of cardiac imaging in management strategies for cardiotoxicity associated with anti-cancer treatment.

Introduction

Overview of cardiovascular toxicity

Cardiotoxicity associated with chemotherapy frequently raises an important issue in cancer treatment, because it can influence the mortality and morbidity of patients with cancer by causing a delay or discontinuation of chemotherapy.1)2) Generally, some requisites are needed to define cardiotoxicity caused by chemotherapy: 1) a cause-and-effect relationship between cardiotoxicity and chemotherapy, 2) a clear mechanism for chemotherapy-induced cardiotoxicity, and 3) available indicators or biomarkers for the early detection or evaluation of cardiotoxicity. Considering these findings, physicians should decide whether to continue, discontinue, or delay chemotherapy or whether to reduce the dose of chemotherapeutic agents in case of cardiotoxicity development.3)4)5)6)
For anthracyclines, the mechanism of cardiotoxicity, including left ventricular (LV) dysfunction, has been previously evaluated, and monitoring or detection of cardiotoxicity can be conducted using biomarkers, including troponin-I (Tn-I) and N-terminal pro-B-type natriuretic peptide (NT-proBNP).7)8)9) Moreover, to some extent, cardiotoxicity can be prevented with the use of a protectant (e.g., dexrazoxane).10)11) In contrast, 5-fluorouracil (FU) has an unclear mechanism of cardiovascular (CV) toxicity; cardiotoxicity related to this agent is difficult to detect or monitor and occurs at a variable incidence of 1% to 8%.12)13) These uncertainties make it difficult to establish consensus on handling cardiotoxicity. Therefore, greater extension of the cardio-oncologic field in cancer treatment is warranted.

Crucial cardiovascular toxicities associated with anti-cancer treatment

A wide spectrum of CV toxicity associated with anti-cancer treatment has been described, and nearly all chemotherapeutic agents can elicit CV toxicity. The prototype of cardiotoxicity is cardiomyopathy or LV dysfunction caused by anthracyclines. Common CV toxicities (Appendix 1 for breast cancer) include 1) cardiomyopathy or heart failure (HF) due to myocardial injury, 2) ischemic heart disease or coronary artery disease, 3) QT prolongation or cardiac arrhythmias, 4) hypertension, 5) thromboembolism, 6) pulmonary artery disease, and 7) pericardial disease.

Cardiomyopathy

Classically, cardiotoxicity associated with anti-cancer treatment has been usually based on the cardiomyopathy with a decreased LV ejection fraction (EF) and HF symptoms (or signs). Cardiotoxicity seems to be limited to a structural disorder, revealed by decreased LVEF, in relation with systolic dysfunction. Therefore, cardiac dysfunction has been defined as a decrease of 10% point from baseline LVEF or an absolute value of LVEF < 53%.3)4)14)15) However, from a cardiologic perspective, the term “cardiomyopathy” can encompass the preserved LVEF or diastolic dysfunction, when LV strain, rather than LVEF is used to assess cardiomyopathy.3)15)
Among cardiac toxicities, cardiomyopathy is frequently encountered in the cardio-oncologic field (Table 1). Cardiomyopathy caused by anti-cancer treatment can be classified into type I and type II toxicities.3)4)15)16)17) Type I toxicity is characterized by irreversible myocardial damage and is frequently associated with anthracycline use. Type II toxicity is associated with targeted therapy such as trastuzumab, which can cause reversible cardiac dysfunction regardless of chemotherapeutic dose.

Anthracyclines

The suggested mechanism of anthracycline-induced cardiotoxicity is oxidative stress with the reactivation of oxygen free radicals or superoxide by iron-anthracycline complex within the mitochondria.6)8)17) The interaction between anthracycline and topoisomerase II beta results in changes in the transcriptome, mitochondrial dysfunction, and the production of reactive oxygen species.7) These consequences disrupt the DNA double strand and damage the myocardium. The injured myocardium experiences programmed apoptosis and cell necrosis accelerated by reactive oxygen species.8)18)19) Other studies have suggested that the anthracycline-mediated mechanism involves the inhibition of adenosine triphosphate in the myocardium and homeostatic changes in calcium metabolism associated with inhibited messenger RNA transcription of Ca-ATPase in the sarcoplasmic reticulum.2)6) Recently, anthracycline-associated cardiomyopathy has been considered to occur more frequently under conditions of myocardial fibrosis or pressure overload such as hypertension.19)

Cyclophosphamide/ifosfamide

The suggested CV toxic mechanism of cyclophosphamide, a drug used in lymphoma and breast cancer treatment, has been the extravasation of chemotherapeutic agents with endothelial damage, leading to interstitial edema, hemorrhagic perimyocarditis, and myocardial necrosis with the production of fibrin microthrombi.2)20)21)22) Likewise, pericardial effusion can be frequently caused by cyclophosphamide.
Ifosfamide, used for germ-cell testicular cancer treatment, seems to have a similar cardiotoxic pathway, but less frequently causes hemorrhagic myocarditis compared to cyclophosphamide. Other explanations of LV dysfunction can include an indirect pathway, such as an electrolyte imbalance and volume changes aggravated by direct nephrotoxicity.23)24)

Trastuzumab

Trastuzumab, a monoclonal antibody against HER2-ErbB2, has been commonly used in HER2 (+) breast cancer.25)26) The cardiotoxic mechanism of this agent is mediated by inhibition of the ErbB2 receptor, which seems to play a major role in the cardiomyocyte proliferation and cardiac development required for cardiac contractility. Cardiotoxicity occurs more frequently when this agent is administered concurrently with anthracyclines.25)26)

Bevacizumab

Bevacizumab, a vascular endothelial growth factor (VEGF) inhibitor used for metastatic breast cancer or colorectal cancer, can worsen underlying HF, hypertension, and arterial thromboembolism.27) The mechanism of CV toxicity of this agent seems to be related to the inhibition of VEGF signaling on vascular smooth muscle cells and the endothelium: CV complications are primarily related with the inhibition of nitric oxide (NO) pathway, activation of endothelin system and vasomotor tone, and promoting changes in the microvasculature.28)29)30) Compared to that observed with trastuzumab, cardiac dysfunction associated with bevacizumab is not common. The risk factors for cardiomyopathy include pre-existing CV disease, concomitant anthracyclines treatment, and old age.31)

Ischemic heart disease

Chest pain can be related to ischemic coronary artery disease or non-ischemic disease. Ischemic heart disease is frequently associated with vasoconstriction or vasomotor reactions to chemotherapeutic agents such as 5-FU (Table 2). Vascular toxicities can be classified into type I (irreversible and processed) and type II (reversible and transient) responses. Type I responses are frequently associated with toxic effects on endothelial cells caused by cisplatin, bleomycin, or vinca alkaloids. Type II responses are associated with vasoconstriction or vasomotion induced by 5-FU, capecitabine, paclitaxel, gemcitabine, rituximab, and sorafenib.32)33)34)

Fluorouracil/capecitabine

Up to 20% of cases of vascular toxicity are estimated to be caused by 5-FU.12)13)35) The action of this agent is relatively acute or associated with early-onset symptoms, including vasoconstriction. The suggested mechanism involves activation of endothelin-1 from smooth muscle cell hyper-reactivity due to postreceptor alterations of protein kinase-C.36)

Paclitaxel

The vascular toxicity of taxanes is mediated by vasospasm which can be induced by overactivated histamine release.37) It can cause myocardial ischemia (up to 4%), particularly in patients with underlying coronary artery disease.

Bevacizumab

Bevacizumab, an inhibitor of VEGF signaling, impairs endothelial NO synthase activity, resulting in aggravated or worsening hypertension.27) It can induce Takotsubo cardiomyopathy with reduced coronary blood flow.38) Moreover, it can prompt platelet aggregation, thrombus formation, and increased blood viscosity.

Arrhythmias

Patients with cancer can experience various arrhythmias, including conduction disorders, tachycardia, or bradycardia.2)39) Briefly, cancer enhances the arrhythmogenic milieu; however, arrhythmias may accompany LV systolic dysfunction, ischemia, or hypertension rather than being directly related to chemotherapy-induced cardiotoxicity. Some major chemotherapeutic agents have been related with specific arrhythmias (Table 3). Therefore, decisions regarding anti-arrhythmic drugs or devices usually require a risk-and-benefit evaluation in patients with cancer.40)

QT prolongation

QT prolongation is one of the important causes of torsade de pointes, which is associated with sudden cardiac death. The duration of the QT interval is influenced by many risk factors, including female sex, old age (> 65 years), LV hypertrophy, congenital long QT syndrome, hormonal disturbances, electrolyte imbalances, and concomitant drugs use.2)41) Several classes of chemotherapeutic agents related with ischemic Several classes of chemotherapeutic agents should be used with caution in patients (Table 4); particularly, arsenic trioxide is considered a common drug (with a prevalence of up to 90%).42) Therefore, to prevent fatal cardiac arrhythmic events, frequent electrocardiographic (ECG) monitoring is recommended and abnormal triggering conditions must be corrected in patients with risk factors.

Supraventricular arrhythmia

Regarding supraventricular arrhythmia, premature atrial contraction and atrial fibrillation are common, but their clinical significance is not clear in patients without symptoms. Atrial fibrillation is frequently observed during treatment with alkylating agents (up to 30%), anthracyclines, and antimetabolites.43) The suggested mechanism of arrhythmia induced by alkylating agents is thought to involve direct irritation of the myocardium.44)

Ventricular arrhythmias

LV dysfunction, ischemia, or QT prolongation are associated with ventricular arrhythmias such as premature ventricular contraction, ventricular tachycardia (VT), or ventricular fibrillation (VF) (torsade de pointes). Despite the lack of arrhythmic data, nonsustained VT can occur during treatment with doxorubicin (5–10%) and taxanes (< 1%).3) Tyrosine kinase inhibitors and alkylating agents can rarely induce VT.

Sinus node dysfunction and conduction disorders

Together with thalidomide, taxanes (paclitaxel) and anti-microtubule agents are usually associated with bradycardia or conduction disorders, such as first degree atrioventricular (AV) block or rarely, asystole.39)43) The incidence of taxanes-associated bradycardia or AV block has been estimated to be approximately 20%.45)46) The mechanism seems to involve histamine receptor stimulation and ischemia with reduced coronary perfusion.47)

Hypertension

Hypertension is frequently encountered in patients with cancer (particularly in renal cancer); apart from old age, it is the most common comorbid condition.48)49) Anti-cancer treatment-associated hypertension was first described with the anti-angiogenic agent (sunitinib). The VEGF signaling pathway is a critical mediator of tumor angiogenesis. VEGF inhibition contributes to endothelial dysfunction such as inhibition of NO production and enhancing of endothelin system, causing vasomotion and microvascular permeability.29)30)50) Furthermore, VEGF inhibitor promotes activation of renin-angiotensin-system with renal function deteriorated.29)30) VEGF inhibitor (bevacizumab, sorafenib, pazopanib, and sunitinib)-associated hypertension is a common side-effect in more than 50% of patients.51)52) Frequently, sunitinib can increase blood pressure (> 150/100 mm Hg) within the first 4 weeks of therapy.53)
While anti-angiogenic therapy-associated hypertension was not fully elucidated, diverse underlying mechanisms of this entity have been proposed, including impaired endothelial function secondary to decreased NO availability, increased vascular tone, and decreased microvascular density (rarefaction).54) In addition to these suggestions, renal dysfunction secondary to thrombotic microangiopathy is considered a specific contributing factor of bevacizumab toxicity.55) VEGF inhibitor-associated hypertension can occur within a few hours to several months after chemotherapy treatment. Therefore, after the initiation of VEGF inhibitors, frequent blood pressure monitoring is necessary for early detection of hypertension.56)

Thromboembolic disease

Vascular toxicity

Among vascular toxicities associated with chemotherapy, two components should be addressed. One form of vascular toxicity is acute vasospasm.57) 5-FU is the most common agent associated with vascular toxicity. Its mechanism is linked with vasoconstriction. Therefore, in patients with underlying atherosclerosis, 5-FU can worsen or aggravate vascular spasm, resulting in acute coronary syndrome, peripheral arterial disease, cerebrovascular disease, or stress-induced cardiomyopathy. The second form of vascular toxicities is arterial thrombosis, which is primarily elicited by reduced NO/prostacyclin signaling and platelet activation. VEGF inhibitors cause endothelial dysfunction and platelet function inhibition, resulting in arterial thrombosis in approximately 4% of patients in addition to venous thrombosis.58)59) The common drugs associated with arterial thrombosis include tyrosine kinase inhibitors (ponatinib, nilotinib, and dasatinib) and cisplatin.

Venous thrombosis and thromboembolism

Malignancy itself can contribute to hypercoagulability. The incidence of venous thromboembolism is estimated to be approximately 7% in patients with malignancies.60) Generally, thrombogenic procoagulants production is related to cancer-associated factors and patient-associated factors such as immobility. As a treatment-related factor, vascular toxicity caused by chemotherapy can aggravate hypercoagulability, which may result in activation of the coagulation pathway, induction of the inflammation response, and inhibition of fibrinolytic activity.
Mucin-producing malignancies, such as pancreatic and gastric cancers, metastatic cancers, and other high-risk malignancies (gastrointestinal, bladder, brain, renal, testicular, lung, and lymphoma) are commonly associated with venous thromboembolism.60)61) A high pre-chemotherapy white-blood cell count (> 11000/L) or platelet count (> 350000/L), anemia (hemoglobin < 11 g/dL), red-blood cell growth factors use, and a high body mass index (> 35 kg/m2) are considered risk factors for venous thromboembolism.62) With respect to chemotherapeutic agents, cisplatin, VEGF inhibitors (e.g., bevacizumab), and radiotherapy are associated with venous thromboembolism.62)63)

Pulmonary hypertension

The relationship between pulmonary hypertension and chemotherapy remains poorly understood and the definite cause-and-effect relationship is unclear. Dasatinib, a tyrosine kinase inhibitor of Bcr-Abl used for leukemia, can cause precapillary pulmonary hypertension. The incidence of dasatinib induced pulmonary hypertension is up to 12%.64)65) It seems to be associated with VEGF receptor-2 dysfunction or inhibition.64)66)

Pericardial disease

Pericardial diseases, such as pericardial effusion or acute pericarditis can develop during or after anti-cancer treatment. In cases of pericardial effusion, the cardiac involvement of the underlying malignancy should be considered. Malignancies associated with cardiac involvement are melanoma, leukemia, lymphoma, and lung, breast, and gastrointestinal tumors. However, radiotherapy-induced pericardial effusion is also common during cancer treatment or after two months of treatment or up to 10 years after radiotherapy.67) Several important chemotherapeutic agents can cause pericardial effusion (Table 5).

Highlights

  • Anthracyclines are common drugs that cause myocardial injury and fibrosis which are mediated by the production of reactive oxygen species and the formation of iron-anthracycline complexes.

  • Cyclophosphamide is associated with the extravasation of chemotherapeutic agent and endothelial damage, causing hemorrhagic perimyocarditis, myocardial necrosis, and pericardial effusion.

  • Trastuzumab, an endothelial growth factor receptor targeting agent, inhibits the ErbB2 receptor and prevents the cardiomyocyte proliferation and cardiac development required for cardiac contractility.

  • Bevacizumab, a VEGF inhibitor, can aggravate underlying HF, hypertension, and arterial thromboembolism by inhibiting VEGF signaling on vascular smooth muscle cells and the endothelium.

  • Type I (irreversible and processed) vascular toxicity is frequently associated with toxic effects on endothelial cells caused by cisplatin, while type II (reversible and transient) toxicity is associated with vasoconstriction, primarily induced by 5-FU.

  • Regular and frequent monitoring is required to early detect arrhythmic events or pericardial disease, particularly for anthracycline treatment.

Chemotherapy Associated with Cardiovascular Toxicity

Anthracyclines

General consideration

Anthracyclines are well-known antitumor agents that are used to treat many different cancers, including lymphoma, leukemia, breast cancer, sarcoma, and ovarian cancer. Anthracyclines include doxorubicin, liposomal doxorubicin, epirubicin, daunorubicin, idarubicin, and mitoxantrone. Common side-effects of anthracyclines include nausea, vomiting, alopecia, mucositis, bone marrow suppression, and cardiotoxicity.

Clinical considerations in anthracycline-induced cardiotoxicity

Anthracycline-induced cardiotoxicity is traditionally classified as acute or late onset type. Acute immediate cardiotoxicity after the injection of anthracycline has been reported in < 1% of patients and is characterized by supraventricular arrhythmia, ECG changes, and transient LV dysfunction.68) Early-onset cardiotoxicity (up to one year after treatment) can occur in 1.6–2.1% of patients, while late-onset cardiotoxicity can occur in 1.6–5% of patients at least one year after treatment.69)70)71) Chronic toxicity may occur up to 20 years after the first dose of anti-cancer treatment.2) Both early- and late-onset chronic cardiotoxicities present as progressive, dilated cardiomyopathy, or HF.4) A recent continuum paradigm of anthracycline-induced cardiotoxicity has emerged, reflecting the development of subclinical myocardial cell injury, early asymptomatic LVEF reduction, and symptomatic HF if left untreated.7)
Anthracycline-induced cardiotoxicity is the prototype for type I chemotherapy-induced cardiotoxicity, which is characterized by dose-dependent, irreversible myocardial cell death. The risk for cardiotoxicity increases with cumulative dose of anthracycline (Table 6). A cumulative doxorubicin dose of 400 mg/m2 determines a 3–5% risk of doxorubicin-induced HF, which increases to 7–26% at 550 mg/m2 and to 18–48% at 700 mg/m2.72) The maximal standard cumulative dose for doxorubicin is 400 to 450 mg/m2, which is considered to achieve the best anticancer effect at a HF risk of 5%.9) The relative cardiotoxicities of anthracyclines and infusion schedules are noted in Table 6. The cardiotoxicity index is derived by dividing 400 mg/m2 as the recommended maximum dose of rapid doxorubicin infusion.

Alkylating agents

Cisplatin

Cisplatin is used to treat osteosarcoma and ovarian, head and neck, esophageal, bladder, and lung cancers.73) Cisplatin has been reported to cause atrial fibrillation, supraventricular tachycardia, left bundle branch block, myocardial ischemia, and myocardial infarction.74) It is frequently related to vascular toxicities, hypertension, and cerebral ischemia. Cisplatin is also associated with venous thromboembolism, with an incidence of up to 8% in a retrospective cohort study.2)75)

Cyclophosphamide

Cyclophosphamide is used to treat lymphoma, leukemia, multiple myeloma, lung cancer, and breast cancer. At low doses, cyclophosphamide is rarely associated with cardiotoxicity. The risk of cardiotoxicity is associated with a high dose of cyclophosphamide (> 150 mg/kg and 1.5 g/m2/day) and the incidence of HF is 7–28%.2) The common findings are tachyarrhythmias, low QRS complex voltage, non-specific T or ST segment abnormalities, and AV conduction disturbances.20)21) In breast cancer, cyclophosphamide is commonly used concurrently and sequentially with anthracyclines, or can be substituted after anthracycline failure.

Ifosfamide

Ifosfamide is a cyclophosphamide analogue that is used to treat soft tissue sarcoma and non-small cell lung cancer. Ifosfamide is associated with arrhythmias, ST segment changes, and HF.24)74) The incidence of HF is 17% and ifosfamide-induced HF occurs dose-dependently within 6–23 days after the initial dose (more than 12.5 g/m2).2)

Antimetabolites

5-fluorouracil

The synthetic pyrimidine metabolite 5-FU is used to treat breast, gastrointestinal, head and neck, and ovarian cancers. Angina-like chest pain is common during 5-FU treatment, particularly with the use of continuous infusion. This type of chest pain is difficult to discriminate from cardiac ischemia or infarction, irrespective of ECG changes. Myocardial ischemia, HF, arrhythmias (including atrial fibrillation, VT, and VF), and cardiogenic shock have rarely been reported.74) The incidence of myocardial ischemia associated with 5-FU ranged from 1–68%.2)75) Usually, either chest pain or arrhythmia shows good response to 5-FU discontinuation or calcium channel blockers (CCBs) administration.75) Although the mechanisms of cardiotoxicity associated with 5-FU are not fully understood, small coronary artery thrombosis, arteritis of small-sized vessels, and vasospasm have been suggested as possible mechanisms.76)

Capecitabine

Capecitabine is an oral prodrug of 5-FU used to treat metastatic breast and colorectal cancers. The incidence and risk factors of capecitabine-associated cardiotoxicity are not as well defined. ECG changes mimicking ST-segment elevation and normal cardiac markers have been noted in many cases.2)

HER2-targeted therapy

Trastuzumab, a humanized monoclonal antibody to the extracellular domain of the HER2 receptor, has been widely used as a combination treatment option with paclitaxel for metastatic breast cancer since it was approved in 1988. It has been available as a first-line therapy for HER2 (+) breast cancer since 2005. The estimated incidence of trastuzumab-associated cardiotoxicity is 2–28%: 2–7% with single trastuzumab treatment, 2–13% in combination with paclitaxel, and up to 27% with concomitant treatment with anthracycline/cyclophosphamide.77)78)79)
Unlike anthracycline-induced type I toxicity, the cardiac toxicity caused by trastuzumab is considered type II toxicity, which can resolve almost completely if the drug is discontinued. Despite the limited data for Asian patients, the risk factors for cardiac toxicity include old age (> 50 years), a mildly decreased LVEF, underlying CV diseases, and a previous history of accumulated doses of doxorubicin (> 300 mg/m2).80)81)82)83) Recently, regular evaluation of cardiac function every three months has been recommended in trastuzumab treatment.3) Furthermore, cardiac Tn-I level and two-dimensional (2D) strain on echocardiography are useful tools for the early detection of LV dysfunction or toxicity and are recommended in every cycle of trastuzumab treatment in high-risk patients.84)85)86)

VEGF inhibitor

VEGF, a primary regulator of angiogenesis, is activated through VEGF receptor signaling. It seems likely that VEGF receptor inhibition can prevent tumors with overexpressed VEGF receptors from progression and metastasis. VEGF inhibitors are currently used in combination with many other chemotherapeutic agents for solid tumors, such as metastatic colon and renal cell malignancies. VEGF inhibitors prevent vascular endothelial cells from proliferation, induce their apoptosis, and impair the production of endothelial NO. Therefore, hypertension can occur commonly, and its incidence may be up to 50%. Blood pressure control is frequently poor, particularly in patients with underlying hypertension.53)87) Additionally, VEGF inhibitors have been associated with thromboembolism because of disturbances in endothelial function.88) According to these vascular toxicity mechanisms, VEGF inhibitors (sunitinib, sorafenib, and pazopanib) can cause myocardial dysfunction within the early treatment period or several months after treatment, although the incidence is not as common as that with anthracyclines or tyrosine kinase inhibitors.89)90) In high-risk patients with a previous history of cardiac toxicity, monitoring of cardiac Tn-I or NT-proBNP levels and echocardiography every 2–3 months would be helpful to detect LV dysfunction.

Highlights

  • Anthracycline-induced cardiotoxicity can occur during the early or late phase of anti-cancer treatment in proportion to the cumulative dose (> 400 mg/m2 of doxorubicin).

  • Alkylating agents, such as cyclophosphamide and cisplatin can cause vascular toxicity, arrhythmic events, and cardiomyopathy.

  • Antimetabolites (5-FU)-associated cardiotoxicity commonly manifests as angina-like chest pain with or without myocardial ischemia.

  • Trastuzumab, an HER2-targeted agent, can cause reversible and transient cardiomyopathy, particularly when used concomitantly with anthracycline/cyclophosphamide.

  • VEGF inhibitors are frequently associated with vascular toxicities, including hypertension and thromboembolism, through endothelial dysfunction.

Prevention of Cardiovascular Toxicity

Preventive treatment for cardiovascular toxicity

Limited data and studies are available regarding primary prophylactic strategies for chemotherapy. The main goal is to prevent cardiac dysfunction, hemodynamic compromise, and vascular damage, regardless of early- or late-phase treatment. Common cardiac side-effects of anti-cancer treatments include cardiomyopathy caused by type I or II cardiotoxicity, ischemic heart disease, and arrhythmias. Endothelial dysfunction/damage or accelerating atherosclerosis also aggravates vascular toxicity. To prevent these adverse CV events, surveillance-based strategies or serial monitoring can be cost-effective and helpful. In anti-cancer treatments, primary prevention or early detection of LV dysfunction has been a primary focus in breast cancer (Appendix 2).

Beta-blockers

Beta-blockers (BB), important agents in HF treatment, exhibit an anti-oxidant mechanism in preventing cardiotoxicity. A meta-analysis revealed their efficacy in reducing cardiac toxicity by 70% in most anthracycline-based breast cancer studies.11) Carvedilol has been shown to prevent doxorubicin or anthracycline cardiotoxicity.91) In the PRevention of cArdiac Dysfunction during Adjuvant breast cancer therapy (PRADA) study, however, metoprolol had no effect on LVEF maintenance.92) Recently, nebivolol was reported to prevent LV systolic dysfunction in a small study.93)

Angiotensin converting enzyme inhibitors or angiotensin receptor blockers

Enalapril, as a prototype, has been evaluated for the prevention of cardiotoxicity and its early administration was shown to be effective in LVEF recovery.94) As combination therapy, enalapril and carvedilol exhibited protective effects against decreased LVEF in a small study.95) The relative risk reduction in angiotensin converting enzyme inhibitors /angiotensin receptor blockers (ACEIs/ARBs) was 70–90%.96) According to the PRADA study, candesartan showed protective effects in patients with early breast cancer.92)

Dexrazoxane

Dexrazoxane prevents anthracycline toxicity by reducing toxic radical species and inhibiting anthracycline-iron binding and subsequent reactive oxygen species due to iron chelation. Furthermore, dexrazoxane can induce the depletion of topoisomerase II beta which can cause to damage DNA through interaction with anthracycline.10) According to the results of a meta-analysis, dexrazoxane can prevent HF (risk ratio 0.29) without a difference in response rate or survival in patients with advanced breast cancer.97) However, because of concerns regarding its interference with anti-tumor effects, dexrazoxane use is suggested by the American Society of Clinical Oncology only for metastatic breast cancer treated with a cumulative doxorubicin dose of more than 300 mg/m2.98)

Statins

The pleiotropic mechanism of statins action was found related to their prophylactic effects, such as reducing oxidative stress, cellular inflammation, and cytokine release. To date, however, few consistent data on statin usefulness for prevention or treatment of cardiotoxicity are available. Based on limited studies, statins may be possibly involved LVEF maintenance or reduction in HF risk, including in patients with breast cancer.99)100) Their relative risk reduction ranged widely from 20–80% compared to placebo.96) Overall, although clear consensus about the preventive role of statins is currently lacking, they may be emerging promising agents for CV toxicity associated with anti-cancer treatment.

Early detection of cardiac dysfunction

Malignancy is occurring at an increasing rate in elderly patients who frequently have many comorbidities, such as hypertension, diabetes, and stroke. Therefore, screening and early identification of high-risk patients is complex and difficult. It is important to consider risk-stratification for cardiac and vascular complications. The risk of CV complications should be defined and evaluated to optimize anti-cancer treatment without detrimental CV side-effects. Therefore, the goal of cardiooncologic pretreatment is the comprehensive risk-stratification and prevention of detrimental CV side-effects in proceeding with anti-cancer treatment.

Cardio-oncologic consideration for chemotherapy

In type I cardiotoxicity, the most important factor for preventing cardiac dysfunction is reducing the cumulative dose of anthracyclines. According to the current guidelines, the general risk factors for cardiomyopathy or HF include exposure to multiple cardiotoxic agents, cumulative doses (> 360 mg/m2 for doxorubicin, > 800 mg/m2 for daunorubicin, > 120 mg/m2 for mitoxantrone, > 90 mg/m2 for idarubicin, and > 720 mg/m2 for epirubicin), the form of administration (continuous infusion over 6 hours vs. bolus administration), combined treatment modality (radiotherapy or chemotherapy), age (< 15 years or > 65 years), and female sex.4)40)101)102)103) A liposome-encapsulated formulation rather than conventional anthracyclines can serve as a strategy to prevent cardiac dysfunction because it could not cross the gap junctions of the cardiac endothelium.104)105) Regarding the cardiac safety about the combination of trastuzumab with doxorubicin/cyclophophamide/paclitaxel, the potential CV toxicity should be carefully considered in case of adjuvant therapy.83)
Other conventional risk factors (Table 7) include hypertension, diabetes, dyslipidemia, myocardial ischemia, arterial thrombosis, and arrhythmias. During or after chemotherapy, uncontrolled hypertension is commonly encountered.
Regarding arrhythmias, anthracycline (doxorubicin), arsenic trioxide, tyrosine kinase inhibitors (sunitinib and vandetanib), and histone deacetylase inhibitors were found associated with QT prolongation, and paclitaxel and thalidomide were found associated with bradycardia.5) In cases of corrected QT (QTc) prolongation > 450 ms in men (> 460 ms in women), arrhythmic events should be considered. According to National Cancer Institute cancer therapy evaluation program, frequent ECG monitoring is required in cases of grade III or IV QTc prolongation (Grade III; QTc ≥ 501 ms without signs and symptoms, Grade IV; QTc ≥ 501 ms with clinical signs or symptoms).106) Avoiding drugs that prolong QTc interval and correction of the QTc interval should be considered, or discontinuation of the scheduled chemotherapy regimen may be considered if necessary.5)40) Therefore, ECG monitoring helps detecting early cardiotoxicity at baseline, at 2–4 weeks, monthly during the first three months, every three months, and then periodically during treatment, depending on the chemotherapy regimen and the patient's status. Patients experiencing diarrhea can be monitored more frequently because of possible electrolyte imbalances, and those receiving arsenic trioxide should be also considered for ECG monitoring.
Overall, the decision to initiate anti-cancer treatment in riskstratified patients should be made through a multi-team approach. During risk-stratification for breast cancer treatment, patients with high-risk symptoms of HF or asymptomatic LV dysfunction can benefit from a cardio-oncologic consultation.107)

Imaging for cardiac dysfunction

Whereas chemotherapy frequently causes cardiac toxicity or LV dysfunction, the appropriate time to initiate HF treatment for recovery of LVEF is thought to be within 6 months from chemotherapy to start of HF therapy.94) To prevent irreversible dysfunction of myocardium, imaging modalities for early detection of LV dysfunction would be required. Regarding imaging studies, echocardiography can be easily used to evaluate LV systolic function, especially using LVEF or strain (Fig. 1). However, to detect early LV dysfunction in an asymptomatic patient, LV strain may be preferred to LVEF. With doxorubicin-based treatment, echocardiography is recommended at baseline (before chemotherapy), every six months within five years after chemotherapy, and when an additional doxorubicin dose of 50 mg/m2 is administered after a cumulative dose of 240 mg/m2.15)108)
Before trastuzumab treatment, a careful evaluation of biomarkers, conventional risk factors, and echocardiography is necessary. If patients show LV dysfunction before or during chemotherapy, low-dose ACEIs or ARBs, BB, and statins may be useful.109) Echocardiographic follow-up is recommended at baseline and every three months during treatment with trastuzumab for LV dysfunction surveillance.4)15)110) During chemotherapy, if LVEF declines to 40% (or ≥ 10% point decrease from baseline), chemotherapeutic agents can be reduced or stopped for at least four weeks (or up to eight weeks).4)111)112) If LVEF recovers to normal range, or reaches at least 45% with a decrease < 10% point from baseline within four weeks, agents can be resumed.4)15)110)113)
Other imaging techniques, including 2D-global longitudinal strain (GLS), three-dimensional (3D)-echocardiography-based LVEF, cardiac magnetic resonance (CMR), or radionuclide planar multi-gated angiography (MUGA), can be used to monitor LV systolic function. However, MUGA has problems in its radiation exposure and cannot evaluate cardiac structure, whereas CMR is not readily accessible. In case of GLS, breast cancer studies, including those on trastuzumab, showed that a relative reduction of 2–15% from baseline GLS was associated with increased rate of cardiotoxicity, and accordingly, reductions > 15% from baseline in GLS have been suggested for detection of LV dysfunction by most experts' consensus statements.15)86)114)115)116)117)

Biological markers for cardiac dysfunction

As a complement to imaging modalities, biomarkers such as Tn-I may be helpful to detect myocardial damage. Cardiac Tn-I level has an excellent negative predictive value (approximately 99%) for cardiotoxicity when measured 1–3 days after each dose and one month after high-dose chemotherapy, and it may be useful even in low-risk patients.108) Particularly, elevated Tn-I level during or within three days of chemotherapy has been associated with the occurrence of LV dysfunction.84)118) Based on these data, Tn-I level measurement at baseline and periodically, or at each cycle of anti-cancer treatment, seems to be a strong tool for surveillance of myocardial injury, and could be a useful biomarker to screen for subsequent CV events.4)114)118)
Contrary to the Tn-I level, the role of the NT-proBNP level in cancer treatment has not been widely evaluated for early detection of LV dysfunction. Generally, the NT-proBNP level is useful for initial risk-stratification and to predict the prognosis of LV dysfunction; persistent elevation of NT-proBNP during a 3-day period of chemotherapy corresponds to a progressive decline in LVEF.119)

Highlights

  • A baseline CV risk assessment including conventional factors should be performed before anti-cancer treatment.

  • For anthracycline-based chemotherapy, serial echocardiography should be performed at baseline, every six months within five years after chemotherapy, and when an additional dose of 50 mg/m2 is administered (if the cumulative dose > 240 mg/m2).

  • For early detection of trastuzumab-related LV dysfunction, imaging modalities such as echocardiography are recommended at baseline and every three months during treatment.

  • As a biomarker, the cardiac Tn-I level is useful for monitoring cardiac tissue injury and can be measured at baseline, within three days, and at each cycle of chemotherapy.

  • In cases of LV dysfunction or elevated Tn-I level, BB, ACEIs/ARBs, or statin can be useful to prevent worsening of cardiac function.

Diagnosis of Cardiovascular Toxicity

Cardiac biomarkers

Cardiac biomarkers have been extensively used to detect cardiotoxicity. Current guidelines suggest Tn-I and NT-proBNP (or BNP) as cardiac biomarkers to detect cardiotoxicity.3) Although several studies are ongoing, limited data are available regarding optimal assessment times or frequencies.

Troponin I

Out of patients receiving high-dose combination chemotherapy, newly elevated cardiac Tn-I levels can identify those who may have a poor prognosis, when an elevated Tn-I level persists.120) Particularly, patients with persistence of Tn-I increase one month after anti-cancer treatment would likely show a great risk of future cardiac event.118) Furthermore, in patients treated with trastuzumab with previous exposure to anthracyclines, Tn-I elevation can predict the development of cardiac dysfunction that may not recover despite treatment for HF.84) Sawaya et al.115) demonstrated that both elevated high-sensitivity (hs) Tn-I levels at three months and a decrease in GLS of echocardiography between baseline and three months were independent predictors of later cardiotoxicity in patients with breast cancer who received anthracyclines and trastuzumab. Ky et al.85) also demonstrated that new elevation of serum hsTn-I in patients receiving anthracyclines and/or trastuzumab predicted subsequent LV dysfunction.

N-terminal pro-B-type natriuretic peptide

The use of NT-proBNP levels for HF has been widely evaluated and it can be useful to detect high-risk patients, as was reflected in the guideline recommendations.121) Recently, a meta-analysis showed that the NT-proBNP level is associated with anthracycline-induced cardiotoxicity and may serve as a useful index for monitoring anthracycline-induced cardiotoxicity at an early stage.122)

Imaging tests for cardiovascular toxicity

Anti-cancer treatment is necessary to control a patient's quality of life and to prolong his or her life, but it is also accompanied by increased morbidity and mortality due to CV toxicity.123) Cardiac function should be evaluated before, during, and after anti-cancer treatment. Particularly, when the baseline cardiac dysfunction is encountered, the etiology or treatment of LV dysfunction might affect the schedule or protocol of anti-cancer treatment, and thus, the cardio-oncologic evaluation with multi-modalities is frequently required (Fig. 2). Several diagnostic modalities are available to assess LV dysfunction, including ECG, echocardiography, nuclear imaging, CMR, and biomarkers.124)

Echocardiography in cardio-oncology

2-dimensional echocardiography

LVEF is most commonly used to evaluate LV function. Current guidelines recommend the measurement of LV function at baseline followed by periodic assessments of LV function to detect cardiotoxicity using LVEF calculated by the modified biplane Simpson's method.15) Echocardiography is the useful tool for assessing myocardial dysfunction before, during, and after anti-cancer treatment.15)125) The currently available echocardiographic criteria for cardiotoxicity are a decrease in LVEF by 10% point or a reduced LVEF < 53%.3)15)

Tissue Doppler and speckle tracking echocardiography

Tissue Doppler imaging and myocardial deformation imaging [strain or strain rate (SR)] might reveal signs of early cardiac dysfunction. Strain, a dimensionless index, reflects the deformation of the ventricular myocardium during the cardiac cycle as a percentage of its initial length, and SR is the rate of deformation or stretch.86) The three components of strain and SR are represented in the longitudinal, radial, and circumferential directions.126)
The deformation imaging can be provided to evaluate subclinical LV dysfunction caused by anti-cancer treatment.15)127)128)129) Strain and SR have allowed myocardial deformation analysis, and peak systolic SR would be a useful parameter for discrimination of intrinsic contractility.130)
Recently, GLS has been reported to accurately predict subtle or subclinical LV dysfunction before a decrease in LVEF is evident.15)130)131) While the definite, absolute value of GLS based on 2D strain for detecting cardiotoxicity has not been yet clearly elucidated, an optimal cut-off value of 17.5–19% of GLS (Vendor, GE Medical systems) would be useful to detect early cardiotoxicity that needs to be further evaluated.86)128) In addition, a reduction in GLS by 15% from baseline is suggested as an indicator of early LV dysfunction, and seems to be more reliable than the absolute GLS value (Fig. 3).86)127) However, the same vendor for GLS follow-ups might be necessary with expertise at laboratories whose results show good accuracy and reproducibility.131) Even in patients with breast cancer showing normal LVEF, standard anti-cancer treatment could reduce GLS values in addition to other conventional echocardiographic parameters.129)

Limitations of echocardiography

Echocardiography is the standard method for cardiac evaluation of patients with cancer, because it is noninvasive, cost-effective, and widely available. Echocardiography can be performed to detect many complications of cancer therapy, including pulmonary hypertension.132)133) It is possible to detect diastolic dysfunction with normal LV systolic function by echocardiography.
Despite the advantages of echocardiography, some limitations should be considered. First, the most important limitation is that echocardiography is operator-dependent; therefore, its reproducibility could be lower compared to that of other imaging modalities. Second, image quality can be poor in some patients. Third, for GLS measurements, there are inter-vendor variabilities and some technical requirements are necessary. To improve reproducibility, the use of 3D echocardiography may be helpful; this technique is associated with the best test-retest variability.134)

Cardiac magnetic resonance in cardio-oncology

CMR is a useful imaging methodology because of excellent spatial resolution and the lack of exposure to ionizing radiation. The most attractive advantage is its ability to detect early or subclinical CV injury before overt manifestations of clinical side-effects of chemotherapeutic agents emerge.135) Despite the increasing application of CMR in cardio-oncology, the risk of side-effects such as nephrogenic systemic fibrosis in patients with renal dysfunction (e.g., estimated glomerular filtration rate < 30–40 mL/min/1.73 m2) or other contraindications to CMR should be always considered.136)

Cardiac tissue characterization

Tissue injury caused by chemotherapy leads to early (acute or subacute) toxicity, including inflammation and edema, and late (chronic) toxicity, including fibrosis or scar formation. These cellular features can be identified by CMR with T2-weighted images, late gadolinium enhancement (LGE) images, or mapping techniques.137)138)139) T2-weighted imaging or T2-short tau inversion recovery (STIR) can reveal inflammation and cellular edema among early histopathological changes. In early hyperemia caused by chemotherapy, increased blood volume or capillary leakage can lead to an increased uptake of gadolinium which shortens T1 time in the phase of myocardial early enhancement.137) In T2-weighted imaging or T2-STIR, water content causes high signal intensity in injured myocardium.140) Moreover, within three months after chemotherapy, contrast-enhanced T1-weighted imaging can reveal early signs of subclinical injury or reduced LVEF.140)141)
After chronic exposure to chemotherapy, chronic, irreversible pathologic changes such as interstitial fibrosis or cellular death appear bright with high signal intensity on LGE images. With recently developed mapping techniques using T2 or T1-LGE, T2-mapping can measure the water content in edematous regions. For extracellular volume (ECV) mapping, T1 mapping before and after enhancement (shortened T1 value) can be used to quantify ECV parameters, which show good correlations with the severity of diffuse fibrosis.142)143) However, limited evidence that these early myocardial changes contribute to late sequelae or the prognosis of CV events is currently available.

Cardiac function

With respect to LV dysfunction, LVEF is the most important parameter for the diagnosis and evaluation of HF. CMR using a cine steady-state free precession sequence has been regarded as the gold standard for evaluating LVEF, including LV volume, right ventricular volume, and LV mass.135) With early subtle changes beginning just after the initiation of chemotherapy, LV mass could increase within several days of treatment due to myocardial edema.144)
During early cardiac toxicity, it is uncertain whether the changes in LV strain can be observed. However, after 1–3 months of chemotherapy, a significant decrease in LVEF and an increase in the LV end-systolic dimension may reflect early cardiac injury. With these findings, subtle diffuse LV fibrosis can be detected via LGE-T1 signal intensity without focal regional enhancement or myocardial edema.139)140)141)
Relative to late cardiac toxicity, approximately a quarter of patients treated with chemotherapy show a decrease in LVEF to less than 50–55%, which is usually accompanied by increased LV end-systolic dimensions. Decreased LV mass is also a crucial marker of late toxicity and seems to be an independent predictor of adverse CV outcomes.144) The features of fibrosis indicated by LGE T1 imaging are frequently associated with irreversible LV dysfunction and poor prognosis.135)139)142)

Vascular injury

In addition to cardiac toxicity, chemotherapy can accelerate or contribute to the development of vascular events, such as hypertension, peripheral arterial disease, or cerebrovascular disease. Unlike atherosclerotic changes, arterial stiffening can be observed via 4D-flow pulse-wave velocity early after the administration of chemotherapy using the phase contrast CMR technique.135)145)146) Vascular injuries are associated with endothelial or vasa-vasorum dysfunction according to pulse-wave velocity or flow-mediated dilation.147) Vascular wall thickness and plaque composition can be also assessed by CMR.

Radionuclide imaging in cardio-oncology

Although radionuclide imaging has been used to evaluate cardiac function before and after anti-cancer treatment, its use in clinical practice is decreasing due to limited availability, low spatial resolution, and radiation exposure.148) Furthermore, it cannot provide information about cardiac structure or LV diastolic function. 2D-echocardiography has been the most widely used modality to assess cardiac function because of its feasibility and accessibility. 2D or 3D echocardiography-based LV volume or LVEF could be an excellent replacement of planar MUGA. Despite the disadvantages of radionuclide imaging, myocardial biological studies on the reversibility or early detection of cellular injury may provide a promising modality to evaluate cardiac toxicity in anti-cancer treatment.149) Therefore, novel nuclear imaging techniques are currently under investigation for the detection of early LV dysfunction.

Cardiac function

Multi-gated radioactivity counts over several cardiac cycles are performed to assess LVEF using 99mTc-erythrocytes from the cardiac blood pool. The change in radioactivity between diastole and systole reflects the LV volume change, although soft tissue attenuation is a troublesome limitation.148) Because of the advantages of accuracy and the lack of an association with geometrical assumptions of LV, MUGA has been frequently used for serial measurements of LVEF.150) In doxorubicin chemotherapy, a decline in LVEF > 15–20% point from baseline or an LVEF < 30% indicates cardiac toxicity, warranting frequent monitoring and possible treatment-discontinuation.151)
Furthermore, 99mTc-gated blood pool single photon emission computed tomography (SPECT) can be used to evaluate LV wall motion and LV volume changes. Recently, 3D-MUGA was proposed as a suitable method for assessing LV volume or dys-synchrony.152)153)

Cardiac cellular injury

Nuclear imaging has been developed for functional cellular study. Among the techniques using radiotracers, iodine-123 metaiodobenzylguanidine (123I-MIBG) and 111In-antimyosin are frequently used in clinical practice.154)155)156)157) In cardiac sympathetic imaging, 123I-MIBG uptake is related to the efferent sympathetic nervous innervation around the heart. Therefore, using planar scintigraphy, both early and late ratios of heart to mediastinal (H/M) uptake can be calculated.155)156) 123I-MIBG shows dose-dependent decreased uptake after chemotherapy, reflecting LV dysfunction in late cardiac toxicity. Patients with an H/M ratio < 1.6 showed adverse cardiac events.158) Therefore, 123I-MIBG can serve as a monitoring tool for the risk of developing cardiomyopathy due to cardiac sympathetic damage.
111In-antimyosin can bind to intracellular myosin which is released from disrupted cardiac sarcolemma, when myocardial necrosis or irreversible cellular damage occurs in myocarditis or myocardial infarction. Regarding myocardial cell injury, 111In-antimyosin Inantimyosin uptake during or after anti-cancer treatment could be helpful in risk-stratification for cardiac prognosis or in identifying subclinical LV systolic dysfunction.156)157) 123I-methylpentadecanoic acid, a radiotracer of free fatty acids, can be used for the early detection of myocardial dysfunction due to decreased mitochondrial uptake of free fatty acids in taxanes-based chemotherapy.159)160) In particular, 111In-trastuzumab SPECT has been under investigation to determine the direct effect of trastuzumab on the HER2 receptor.161)162)163)

Positron emission tomography

Cardiac positron emission tomography has been used to detect highly metabolic tissues, such as malignancy or inflammation, using two radionuclides relative to cardiac metabolism and perfusion. However, its role in detecting early LV dysfunction is still limited due to its high cost and low availability.

Highlights

  • To early detect LV dysfunction, echocardiography and cardiac biomarkers are preferentially recommended.

  • In cases of suboptimal or imprecise findings on echocardiography, CMR can be used.

  • CMR can provide an excellent imaging modality to evaluate myocardial tissue injury, such as inflammation or edema.

  • Regarding LV dysfunction, LGE by CMR can provide prognostic information for adverse CV outcomes.

  • The measurement of LVEF can be assessed by MUGA when 2D/3D echocardiography or CMR is unavailable.

  • Subclinical biological evaluations of cardiomyocytes using SPECT or radionuclide imaging with 111In-antimyosin, 123I-MIBG, or 111In-trastuzumab may serve as a promising strategy to detect cardiac toxicity.

Management of Cardiovascular Toxicity

Heart failure

Although patients receiving potentially toxic chemotherapy were excluded from large randomized HF trials, the management of chemotherapy-related LV dysfunction is considered the same as that for any form of LV dysfunction.164)165) In the American College of Cardiology Foundation/the American Heart Association guidelines, patients with cancer treated with potentially cardiotoxic agents are classified as patients at a high risk for HF development (stage A).164) In patients in stage A, the risk factors for coronary artery disease should be controlled, and ACEIs or ARBs should be considered for the treatment of hypertension.164) For patients with asymptomatic LV dysfunction (stage B), limited data showed some benefits of slowing or reversing LV dilation; despite the lack of evidence, treatment with ACEIs (or ARBs) and a BB should be considered for patients with asymptomatic LV dysfunction or a decline in LVEF < 10% point of the lower normal limit.3) For patients with symptomatic LV dysfunction (stage C and D), ACEIs (or ARBs if there is a contraindication or intolerance) and BB are recommended, and mineralocorticoid receptor antagonists can be given if symptoms persist (Table 8). Recent trials and updated guidelines recommend an angiotensin receptor-neprilysin inhibitor (ARNI) (sacubitril/valsartan) rather than ACEIs (or ARBs) if symptoms persist, and ivabradine may be recommended in symptomatic patients with sinus rhythm and a heart rate above 70 bpm.166) However, the data on ARNI and ivabradine are currently limited.
Another important point about the recovery of cardiac dysfunction is the timing of treatment initiation with ACEIs and BB. In patients with type I cardiotoxicity, early treatment with ACEIs (enalapril 2.5–20 mg/day) and/or BB (carvedilol 12.5–50 mg/day) immediately after cardiac dysfunction is detected may result in better cardiac outcomes.94)167) However, the role of HF medications in the management of type II cardiotoxicity with trastuzumab remains uncertain. Initial studies on trastuzumab-induced cardiomyopathy revealed that cardiac function could be restored by withholding trastuzumab (for a mean of 1.5 months).168) However, trastuzumab is a crucial treatment for breast cancer and a “drug holiday (cessation)” may be detdetrimental.169)170)171) Whether continuation of trastuzumab with cardioactive protective medication improves outcomes also remains uncertain.
Another issue is the lack of rationale for the management of patients with recovered LV function. It seems reasonable to maintain treatment during periods in which a patient needs more chemotherapy. After cure is achieved, some experts recommend maintaining treatment for at least one year, although others suggest that treatment should be extended in the absence of contraindications.172)

Myocardial ischemia and coronary artery disease

The administration of cytotoxic agents in anti-cancer treatment increases the risk of myocardial ischemia through various mechanisms (coronary vasospasm, thrombosis, and vascular dysfunction). In anti-cancer agent-induced coronary spasm, it is important to identify the temporal relationship between drug administration and the occurrence of chest pain. If symptoms occur during drug administration, the drug should be stopped and then symptoms can be treated with sublingual nitroglycerin and opioids.112) Although prophylactic therapy with nitrates and CCBs does not appear to be universally effective, these are the only available options.173)
In radiation-associated ischemia, patients with stable angina can be managed in the same manner as patients with stable angina due to atherosclerotic coronary artery disease and those with unstable symptoms can be managed according to the existing acute coronary syndrome guidelines.174)175)
Invasive treatment can be considered based on the severity of coronary artery disease, the stage of malignancy, the condition of patient, thrombocytopenia, and the need for future cancer surgery. For revascularization of coronary stenosis, cancer surgery planned in the near future and occurrence of severe thrombocytopenia should be addressed in case of percutaneous coronary intervention (PCI) using dual antiplatelet therapy (DAPT). In other cases, coronary artery bypass graft (CABG) surgery could be an alternative. However, the disadvantage of CABG surgery, including an impaired wound healing, post-operative infection, or prolonged recovery time should be also considered. Therefore, therapeutic options may be individualized according to the risk of the revascularization methods, the urgency of coronary artery disease, the risk of the surgery, and the likelihood of severe coronary artery disease.176)177) According to the Society for Cardiovascular Angiography and Interventions Consensus, when selecting a stent, bare-metal stents or new-generation drug-eluting stents are recommended because of their low risk of thrombosis.178) To reduce the need for DAPT, drug-eluting balloons, or bio-absorbable polymers/scaffolds may be used. In all patients receiving PCI, an activated clotting time > 250 seconds is necessary during the procedure. For patients with cancer and severe thrombocytopenia (< 50000/mL), lower doses of unfractionated (30–50 U/kg) heparin can achieve a therapeutic activated clotting time.178) Antiplatelet treatment should be individualized. The platelet count > 10000/mL usually allows treatment of aspirin, and DAPT may require a platelet count > 30000–50000/mL.178)

Arrhythmias

QT prolongation

Although anti-cancer drug-induced QT prolongation may lead to substantial risks of life-threatening cardiac arrhythmias such as torsade de pointes, the potential benefit of chemotherapy over the risk of QTc prolongation may be considered in patients with cancer.179)180)181) Discontinuation of anti-cancer drugs is required in case of significant QTc prolongation, serious arrhythmias (torsade de pointes), symptomatic HF, shock, or syncope. Above all things, however, it is important that arrhythmic triggering conditions are corrected.

Atrial fibrillation

It may be reasonable to consider recovery and maintenance of sinus rhythm using cardioversion and/or antiarrhythmic therapy if symptoms persist despite appropriate heart rate control with BB or non-dihydropyridine (DHP) CCBs.182) The clinical situation dictates the decision to continue the use of the presumed offending anticancer agent. However, the presence of atrial fibrillation alone could not provide a sufficient reason for the cessation of anti-cancer treatment.
The use of warfarin or novel oral anticoagulants (NOAC) in the setting of chemotherapy is another issue. Generally, the risk of thromboembolism and bleeding in patients with atrial fibrillation can be generally assessed by CHA2DS2-VASc and HAS-BLED scores despite the lack of validation in patients with cancer.183) In patients with CHA2DS2-VASc scores ≥ 2, anticoagulation can be considered if the platelet count is > 50000/mL.184) Even in low-risk patients, given the risk of venous thromboembolism, prophylaxis may be considered.
Anticoagulation options include low-molecular weight heparin, vitamin K antagonist, or NOAC. Warfarin is often avoided in patients with metastatic cancer or a high bleeding risk. In patients requiring multiple procedures, patients with a high potential for drug-drug interactions, or patients with a high risk for bleeding, alternative drugs such as low-molecular weight heparin may be suitable for anticoagulation.185) Although there is a lack of consensus, some clinical studies for active cancer patients with atrial fibrillation showed the comparable safety and efficacy of NOAC for stroke prevention.186)187)

Hypertension

Treatment for anti-cancer drug-induced hypertension can be initiated with diuretics, BB, ACEIs, ARBs, or CCBs. The choice of anti-hypertensive drug should be tailored to the clinical situation, including consideration of possible drug-drug interactions, volume status, and renal function.188) ACEIs or ARBs are usually recommended for patients with metabolic syndrome, proteinuria, or a high risk for chronic kidney disease. While DHP CCBs are useful for old age, non-DHP CCBs are not recommended for patients receiving cytochrome-P450 inhibitors.98)189) Weekly blood pressure monitoring is recommended during the first cycle of therapy and then every 2–3 weeks during anticancer therapy.112)

Thromboembolism

Malignancy is frequently associated with a prothrombotic or accelerated coagulation cascade. A patient's life expectancy and bleeding risk should be always assessed when deciding whether to administer anticoagulants. Thromboembolic events can be aggravated by several drugs, such as cisplatin, thalidomide, lenalidomide, or vorinostat. Therefore, thromboprophylaxis with low-molecular weight heparin or low-dose aspirin is recommended in acute hospitalized patients with active malignancy, patients undergoing major cancer surgery, and patients with ambulatory multiple myeloma receiving chemotherapy.190) In ambulatory patients receiving chemotherapy, the role of prophylactic antithrombotic agents remains controversial and the bleeding tendency with anticoagulation drugs should be considered in cases of primary prevention even though venous thromboembolism is common.191)
In acute venous thromboembolism, a 3 to 6-month regimen of low-molecular weight heparin is recommended in hemodynamically stable patients.3) Low-molecular weight heparin would be superior to warfarin in reducing recurrent thromboembolic events with comparable mortality and bleeding risk in patients with cancer.192) The clinical evidence of NOAC efficacy in patients with cancer is limited. In clinical trials, NOAC and warfarin showed no difference in recurrent thromboembolism and bleeding events.193)194) However, few direct comparative studies between NOAC and low-molecular weight heparin are available.
In order to decide whether anticoagulation should be continued, careful discussion with a detailed overview of cancer therapy, thromboembolic risk, bleeding risk, and patient preference is necessary.3) In cases of recurrent venous thromboembolism despite anticoagulation treatment, switching from warfarin to low-molecular weight heparin or increasing the dose of low-molecular weight heparin may be considered.195) Long-term anticoagulation may be required to prevent recurrent thromboembolic events after acute-phase treatment until a cure is achieved because patients with cancer have a high risk of recurrent thromboembolic events. An inferior vena cava filter may be implanted if anticoagulation therapy is contraindicated or fails.195) However, the risk of filter-related complications, such as filter thrombosis and occlusion should also be considered before implantation.
Because of the increased risk of bleeding, the benefits of thrombolytic therapy in patients with hemodynamically unstable pulmonary thromboembolism remain controversial.3) Thrombolytic therapy is absolutely contraindicated for malignant brain tumors.196) Surgical thrombectomy may be considered, but surgical treatment may carry a high bleeding risk and significant morbidity.194) Venous thromboembolism may be detected incidentally during imaging for cancer. Since recurrent thromboembolic events and mortality are increased in these patients, incidentally detected asymptomatic venous thromboembolic disease can be treated as a symptomatic event.197)

Pulmonary hypertension

Baseline assessment, including CV risk factors, functional capacity (e.g., NYHA/WHO functional class and 6-minute walking distance), cardiac biomarkers (NT-proBNP), and echocardiography is recommended in all patients receiving chemotherapeutic agents known to cause pulmonary hypertension.3) In patients complaining of dyspnea or exercise limitations during cancer therapy, an echocardiographic examination may be considered.3) Further cardiologic examinations such as right heart catheterization may be needed in patients with echocardiographic evidence of pulmonary hypertension and right ventricular overload to confirm pulmonary hypertension and to determine the etiology.198) Assessment of functional capacity and echocardiographic surveillance may be considered every 3–6 months in the setting of treatment with drugs associated with pulmonary hypertension, even in asymptomatic patients.
If drug-induced pulmonary arterial hypertension is detected, referral to a specialized pulmonary hypertension center and a multidisciplinary team approach are recommended to determine whether or not to continue anti-cancer treatment even using pulmonary arterial hypertension-specific drugs.198) Particularly, dasatinib, a tyrosine kinase inhibitor, frequently induces severe precapillary pulmonary hypertension, but this is often reversible after drug discontinuation.65) In cases of pulmonary arterial hypertension associated with drugs and toxins (WHO group I), ambrisentan or iloprost inhaler can be used as target therapy. Cyclophosphamide and other alkylating agents may cause pulmonary veno-occlusive disease which has no effective pharmacologic treatment.199)

Pericardial disease

Several chemotherapeutic agents and chest radiotherapy can cause various pericardial diseases, including acute pericarditis, pericardial effusion, and constrictive pericarditis. Each specific management strategy follows general treatment principles.200) Aspirin or non-steroidal anti-inflammatory drugs are the mainstays of therapy for acute pericarditis and colchicine is recommended to reduce inflammation and to prevent recurrence.201) Pericardiocentesis should be considered in patients with hemodynamically unstable pericardial effusion, and it may be considered to relieve symptoms and establish a diagnosis in patients with extensive pericardial effusion.202) Pericardiotomy can be considered if pericardiocentesis cannot be performed.203) Surgical pericardiectomy is rarely indicated for constrictive pericarditis or for the management of procedure-related complications.

Mechanical circulatory support in patients with cancer

Quality-adjusted life expectancy and underlying cancer status are always considered before the implantation of mechanical circulatory devices. Mechanical circulatory support as a bridge to transplantation or destination therapy is not recommended in patients with an active hematologic or solid organ malignancy and a life expectancy < 2-year. However, patients who have been treated for cancer and have maintained disease-free status may be candidates for mechanical circulatory support as a bridge to transplantation. Mechanical circulatory support as destination therapy may also be considered in patients with a life expectancy > 2-year.204)

Highlights

  • The conventional treatment of HF requires the use of BB, ACEIs (or ARBs), or aldosterone blockers according to guidelines, but the data are limited.

  • In cases of symptomatic ischemic disease, anti-cancer treatment should be stopped, and evaluation with treatment is recommended.

  • Significant symptomatic QT prolongation should be addressed through the correction of triggering factors; discontinuation of anti-cancer treatment may be considered.

  • Prophylactic anticoagulation for venous thromboembolism is recommended after cancer surgery for a minimum four weeks. Long-term anticoagulation may be considered to prevent recurrent thromboembolic events after the acute phase of treatment until the cancer is cured.

Future Directions

The CV side-effects or toxicities caused by anti-cancer treatment have a critical impact on the evaluation and future treatment of malignancies. Unfortunately, few studies regarding the cause-and-effect relationship between cardiotoxicity, treatment, monitoring, and biomarkers in the prevention of LV dysfunction are currently available. To increase treatment effectiveness for cardiotoxicity, primary prevention using precise, easy, and simple methods to detect side-effects is necessary. Advances in imaging modalities and the availability of cardiac biomarkers can contribute to early diagnosis, leading to better quality of life in patients with cancer. Together with these developments, a prediction model for CV toxicity is needed for risk-stratification relative to CV prognosis. Future studies in the cardio-oncologic field should gather data regarding the mechanism and prevention of the cardiotoxicity associated with anti-cancer treatment. Therefore, close collaboration between cardiology and oncology teams is necessary.

Reviewers

This paper was reviewed by the members of the Cardio-oncology of the Korean Society of Echocardiography. Reviewers included Sung Hoon Sim, MD, Jeong-Yoon Song, MD, Sang Jae Rhee, MD, Sung-Eun Lee, MD, Jieun Lee, MD, Hyewon Lee, MD, Joo Han Lim, MD, Jung Hyun Choi, MD, and Byung Joo Chae, MD.

The Cardio-oncology of the Korean Society of Echocardiography Committee

Ho-Joong Youn (Chairperson), Cardiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul; Woo-Baek Chung (General Secretary), Cardiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul; Hyungseop Kim (Guideline Enactment Committee), Cardiology, Keimyung University Dongsan Medical Center, Daegu; Il Suk Sohn, (Scientific Committee), Cardiology, Kyung Hee University Hospital at Gangdong, Seoul; Seongwoo Han (Registration Committee), Cardiology, Dongtan Sacred Heart Hospital, Hallym University College of Medicine, Hwaseong, Korea.
Hak Jin Kim/Mi Hyang Kwak/Eun Jeong Cho (Center for Clinical Specialty), Hyeon-Seok Eom/Hyewon Lee (Center for Hematologic Malignancy), Sung Hoon Sim (Center for Breast Cancer), National Cancer Center, Goyang; Chi Young Shim (Cardiology), Gun Min Kim (Medical Oncology), Yonsei University College of Medicine, Seoul; Kee Sik Kim (Cardiology), Daegu Catholic University Medical Center, Daegu; Wan-Joo Shim/Seong-Mi Park (Cardiology), Korea University Anam Hospital, Korea University College of Medicine, Seoul; Seong Hwan Kim (Cardiology), Korea University Ansan Hospital, Korea University College of Medicine, Ansan; Eun Kyoung Kim (Cardiology), Ji-Yeon Kim (Hematology-Oncology), Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul; Yun-Gyoo Lee (Oncology), Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul; Chung Il Noh (Pediatrics), Seoul National University, Seoul; Chang-Ki Min/Sung-Eun Lee (Hematology), Jieun Lee (Oncology), Woo-Chan Park/Byung Joo Chae (Breast Surgery), Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul; Byung Joo Song (Breast Surgery), Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Bucheon; Hui-Kyung Jeon (Cardiology), Uijeongbu St. Mary's Hospital, The Catholic University of Korea, Uijeongbu; Jung Sun Cho (Cardiology), Daejeon St. Mary's Hospital, The Catholic University of Korea, Daejeon; Jin-Sun Park (Cardiology), Ajou University School of Medicine, Suwon; Jeong-Sook Seo (Cardiology), Busan Paik Hospital, Inje University, Busan; Jeong-Yoon Song (General Surgery), Wan Kyu Eo (Hemato-Oncology), Kyung Hee University Hospital at Gangdong, Seoul; Gil Ja Shin/Jeong Eun Yi (Cardiology), Ewha Womans University School of Medicine, Seoul; Hyun Ju Yoon (Cardiology), Chonnam National University Hospital, Gwangju; Jin-Won Jeong/Sang Jae Rhee (Cardiology), Wonkwang University Hospital, Iksan; Kyoung Im Cho/Bong-Joon Kim (Cardiology), Eunmi Lee (Hemato-Oncology), Kosin University College of Medicine, Busan; Joo Han Lim (Hemato-Oncology), Inha University Hospital, Incheon; Ju-Hee Lee (Cardiology), Chungbuk National University Hospital, Cheongju; Jung Hyun Choi (Cardiology), Pusan National University School of Medicine, Busan, Korea.

Acknowledgements

This position paper is financially supported by the Korean Society of Echocardiography.

Appendices

Appendix 1

Adverse cardiovascular side-effects

1. Cardiomyopathy
1) The most common cause of drugs
  • (1) Anthracycline: Doxorubicin (a cumulative dose of > 400 mg/m2)

    Epirubicin (a cumulative dose of > 900 mg/m2)
  • (2) Trastuzumab

  • (3) Small molecule tyrosine kinase inhibitors: sorafenib, sunitinib, pazopanib

  • (4) 5-fluorouracil

  • (5) Cyclophosphamide (in combination with anthracycline)

  • (6) Docetaxel

2) Time onset: few months to years after treatment
2. Ischemic heart disease
5-fluorouracil, capecitabine (Xeloda), taxanes, bevacizumab (Avastin), cisplatin
3. Venous thromboembolism/stroke
Cisplatin, selective estrogen receptor modulator (SERM): tamoxifen
4. Hypertension
1) Small molecule tyrosine kinase inhibitors: sorafenib, sunitinib, pazopanib
2) Vascular endothelial growth factor inhibitors: bevacizumab
5. QT prolongation
Doxorubicin, SERM (tamoxifen)

Appendix 2

Prevention and treatment of cardiovascular side-effects

1. Beta-blocker, angiotensin-converting enzyme inhibitor/-receptor blocker, statin
1) For asymptomatic or symptomatic structural heart disease
2) For elevated cardiac Tn-I or NT-proBNP
2. Dexrazoxane
For metastatic breast cancer (> 300 mg/m2 of doxorubicin)
3. Monitoring of cardiac function and cardiac biomarkers
1) Echocardiography, cardiac MR
2) Tn-I, NT-proBNP

References

1. Meinardi MT, Gietema JA, van Veldhuisen DJ, van der Graaf WT, de Vries EG, Sleijfer DT. Long-term chemotherapy-related cardiovascular morbidity. Cancer Treat Rev. 2000; 26:429–447. PMID: 11139373.
2. Yeh ET, Bickford CL. Cardiovascular complications of cancer therapy: incidence, pathogenesis, diagnosis, and management. J Am Coll Cardiol. 2009; 53:2231–2247. PMID: 19520246.
3. Zamorano JL, Lancellotti P, Rodriguez Muñoz D, Aboyans V, Asteggiano R, Galderisi M, Habib G, Lenihan DJ, Lip GY, Lyon AR, Lopez Fernandez T, Mohty D, Piepoli MF, Tamargo J, Torbicki A, Suter TM, Zamorano JL, Aboyans V, Achenbach S, Agewall S, Badimon L, Barón-Esquivias G, Baumgartner H, Bax JJ, Bueno H, Carerj S, Dean V, Erol Ç, Fitzsimons D, Gaemperli O, Kirchhof P, Kolh P, Lancellotti P, Lip GY, Nihoyannopoulos P, Piepoli MF, Ponikowski P, Roffi M, Torbicki A, Vaz Carneiro A, Windecker S. Authors/Task Force Members. ESC Committee for Practice Guidelines (CPG). Document Reviewers. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines: the Task Force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC). Eur J Heart Fail. 2017; 19:9–42. PMID: 27565769.
4. Curigliano G, Cardinale D, Suter T, Plataniotis G, de Azambuja E, Sandri MT, Criscitiello C, Goldhirsch A, Cipolla C, Roila F. ESMO Guidelines Working Group. Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO clinical practice guidelines. Ann Oncol. 2012; 23(Suppl 7):vii155–vii166. PMID: 22997448.
5. Maurea N, Spallarossa P, Cadeddu C, Madonna R, Mele D, Monte I, Novo G, Pagliaro P, Pepe A, Tocchetti CG, Zito C, Mercuro G. A recommended practical approach to the management of target therapy and angiogenesis inhibitors cardiotoxicity: an opinion paper of the working group on drug cardiotoxicity and cardioprotection, Italian Society of Cardiology. J Cardiovasc Med (Hagerstown). 2016; 17(Suppl 1):S93–S104. PMID: 27183530.
6. Albini A, Pennesi G, Donatelli F, Cammarota R, De Flora S, Noonan DM. Cardiotoxicity of anticancer drugs: the need for cardio-oncology and cardio-oncological prevention. J Natl Cancer Inst. 2010; 102:14–25. PMID: 20007921.
7. Groarke JD, Nohria A. Anthracycline cardiotoxicity: a new paradigm for an old classic. Circulation. 2015; 131:1946–1949. PMID: 25948539.
8. Distefano G. Molecular pathogenetic mechanisms and new therapeutic perspectives in anthracycline-induced cardiomyopathy. Ital J Pediatr. 2009; 35:37. PMID: 19930562.
9. Ewer MS. Anthracycline cardiotoxicity: clinical aspects, recognition, monitoring, treatment, and prevention. In : Ewer MS, Yeh ET, editors. Cancer and the heart. 2nd ed. Shelton (CT): People's Medical Publishing House - USA, Ltd;2013. p. 11–41.
10. Lyu YL, Kerrigan JE, Lin CP, Azarova AM, Tsai YC, Ban Y, Liu LF. Topoisomerase IIbeta mediated DNA double-strand breaks: implications in doxorubicin cardiotoxicity and prevention by dexrazoxane. Cancer Res. 2007; 67:8839–8846. PMID: 17875725.
11. Schimmel KJ, Richel DJ, van den Brink RB, Guchelaar HJ. Cardiotoxicity of cytotoxic drugs. Cancer Treat Rev. 2004; 30:181–191. PMID: 15023436.
12. Kosmas C, Kallistratos MS, Kopterides P, Syrios J, Skopelitis H, Mylonakis N, Karabelis A, Tsavaris N. Cardiotoxicity of fluoropyrimidines in different schedules of administration: a prospective study. J Cancer Res Clin Oncol. 2008; 134:75–82. PMID: 17636329.
13. Meyer CC, Calis KA, Burke LB, Walawander CA, Grasela TH. Symptomatic cardiotoxicity associated with 5-fluorouracil. Pharmacotherapy. 1997; 17:729–736. PMID: 9250550.
14. Khouri MG, Douglas PS, Mackey JR, Martin M, Scott JM, Scherrer-Crosbie M, Jones LW. Cancer therapy-induced cardiac toxicity in early breast cancer: addressing the unresolved issues. Circulation. 2012; 126:2749–2763. PMID: 23212997.
15. Plana JC, Galderisi M, Barac A, Ewer MS, Ky B, Scherrer-Crosbie M, Ganame J, Sebag IA, Agler DA, Badano LP, Banchs J, Cardinale D, Carver J, Cerqueira M, DeCara JM, Edvardsen T, Flamm SD, Force T, Griffin BP, Jerusalem G, Liu JE, Magalhães A, Marwick T, Sanchez LY, Sicari R, Villarraga HR, Lancellotti P. 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. PMID: 25172399.
16. Ewer MS, Lippman SM. Type II chemotherapy-related cardiac dysfunction: time to recognize a new entity. J Clin Oncol. 2005; 23:2900–2902. PMID: 15860848.
17. Chung WB, Youn HJ. Pathophysiology and preventive strategies of anthracycline-induced cardiotoxicity. Korean J Intern Med. 2016; 31:625–633. PMID: 27378126.
18. Scherrer-Crosbie M. Cardiotoxicity of oncologic treatments. Is there a uniform definition?. In : Lancellotti P, Zamorano J, Galderisi M, editors. Anticancer treatments and cardiotoxicity mechanisms, diagnostic, and therapeutic interventions. 1st ed. London: Elsvier;2016. p. 35–38.
19. Youn HJ, Kim HS, Jeon MH, Lee JH, Seo YJ, Lee YJ, Lee JH. Induction of caspase-independent apoptosis in H9c2 cardiomyocytes by adriamycin treatment. Mol Cell Biochem. 2005; 270:13–19. PMID: 15792349.
20. Taniguchi I. Clinical significance of cyclophosphamide-induced cardiotoxicity. Intern Med. 2005; 44:89–90. PMID: 15750266.
21. O'Connell TX, Berenbaum MC. Cardiac and pulmonary effects of high doses of cyclophosphamide and isophosphamide. Cancer Res. 1974; 34:1586–1591. PMID: 4833912.
22. Braverman AC, Antin JH, Plappert MT, Cook EF, Lee RT. Cyclophosphamide cardiotoxicity in bone marrow transplantation: a prospective evaluation of new dosing regimens. J Clin Oncol. 1991; 9:1215–1223. PMID: 2045862.
23. Kandylis K, Vassilomanolakis M, Tsoussis S, Efremidis AP. Ifosfamide cardiotoxicity in humans. Cancer Chemother Pharmacol. 1989; 24:395–396. PMID: 2791194.
24. Madeddu C, Deidda M, Piras A, Cadeddu C, Demurtas L, Puzzoni M, Piscopo G, Scartozzi M, Mercuro G. Pathophysiology of cardiotoxicity induced by nonanthracycline chemotherapy. J Cardiovasc Med (Hagerstown). 2016; 17(Suppl 1):e12–e18. PMID: 27755238.
25. Ewer SM, Ewer MS. Cardiotoxicity profile of trastuzumab. Drug Saf. 2008; 31:459–467. PMID: 18484781.
26. Nowsheen S, Viscuse PV, O'Sullivan CC, Sandhu NP, Haddad TC, Blaes A, Klemp J, Nhola L, Herrmann J, Ruddy KJ. Incidence, diagnosis, and treatment of cardiac toxicity from trastuzumab in patients with breast cancer. Curr Breast Cancer Rep. 2017; 9:173–182. PMID: 29225726.
27. Force T, Krause DS, Van Etten RA. Molecular mechanisms of cardiotoxicity of tyrosine kinase inhibition. Nat Rev Cancer. 2007; 7:332–344. PMID: 17457301.
28. Chen ZI, Ai DI. Cardiotoxicity associated with targeted cancer therapies. Mol Clin Oncol. 2016; 4:675–681. PMID: 27123262.
29. Lankhorst S, Kappers MH, van Esch JH, Danser AH, van den Meiracker AH. Hypertension during vascular endothelial growth factor inhibition: focus on nitric oxide, endothelin-1, and oxidative stress. Antioxid Redox Signal. 2014; 20:135–145. PMID: 23458507.
30. Pandey AK, Singhi EK, Arroyo JP, Ikizler TA, Gould ER, Brown J, Beckman JA, Harrison DG, Moslehi J. Mechanisms of VEGF (vascular endothelial growth factor) inhibitor-associated hypertension and vascular disease. Hypertension. 2018; 71:e1–e8. PMID: 29279311.
31. Scappaticci FA, Skillings JR, Holden SN, Gerber HP, Miller K, Kabbinavar F, Bergsland E, Ngai J, Holmgren E, Wang J, Hurwitz H. Arterial thromboembolic events in patients with metastatic carcinoma treated with chemotherapy and bevacizumab. J Natl Cancer Inst. 2007; 99:1232–1239. PMID: 17686822.
32. Samuels BL, Vogelzang NJ, Kennedy BJ. Severe vascular toxicity associated with vinblastine, bleomycin, and cisplatin chemotherapy. Cancer Chemother Pharmacol. 1987; 19:253–256. PMID: 2438065.
33. Lu JI, Carhart RL, Graziano SL, Gajra A. Acute coronary syndrome secondary to fluorouracil infusion. J Clin Oncol. 2006; 24:2959–2960. PMID: 16782933.
34. Frickhofen N, Beck FJ, Jung B, Fuhr HG, Andrasch H, Sigmund M. Capecitabine can induce acute coronary syndrome similar to 5-fluorouracil. Ann Oncol. 2002; 13:797–801. PMID: 12075751.
35. Polk A, Vistisen K, Vaage-Nilsen M, Nielsen DL. A systematic review of the pathophysiology of 5-fluorouracil-induced cardiotoxicity. BMC Pharmacol Toxicol. 2014; 15:47. PMID: 25186061.
36. Schöber C, Papageorgiou E, Harstrick A, Bokemeyer C, Mügge A, Stahl M, Wilke H, Poliwoda H, Hiddemann W, Köhne-Wömpner CH, Weiss J, Preiss J, Schmoll HJ. Cardiotoxicity of 5-fluorouracil in combination with folinic acid in patients with gastrointestinal cancer. Cancer. 1993; 72:2242–2247. PMID: 8374883.
37. Ward S, Simpson E, Davis S, Hind D, Rees A, Wilkinson A. Taxanes for the adjuvant treatment of early breast cancer: systematic review and economic evaluation. Health Technol Assess. 2007; 11:1–144.
38. Franco TH, Khan A, Joshi V, Thomas B. Takotsubo cardiomyopathy in two men receiving bevacizumab for metastatic cancer. Ther Clin Risk Manag. 2008; 4:1367–1370. PMID: 19337443.
39. Tamargo J, Caballero R, Delpón E. Cancer chemotherapy and cardiac arrhythmias: a review. Drug Saf. 2015; 38:129–152. PMID: 25577497.
40. Priori SG, Blomström-Lundqvist C, Mazzanti A, Blom N, Borggrefe M, Camm J, Elliott PM, Fitzsimons D, Hatala R, Hindricks G, Kirchhof P, Kjeldsen K, Kuck KH, Hernandez-Madrid A, Nikolaou N, Norekvål TM, Spaulding C, Van Veldhuisen DJ. 2015 ESC guidelines for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death: The Task Force for the management of patients with ventricular arrhythmias and the prevention of sudden cardiac death of the European Society of Cardiology (ESC). Endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC). Eur Heart J. 2015; 36:2793–2867. PMID: 26320108.
41. Vorchheimer DA. What is QT interval prolongation. J Fam Pract. 2005; (Suppl):S4–S7. PMID: 15938992.
42. Roboz GJ, Ritchie EK, Carlin RF, Samuel M, Gale L, Provenzano-Gober JL, Curcio TJ, Feldman EJ, Kligfield PD. Prevalence, management, and clinical consequences of QT interval prolongation during treatment with arsenic trioxide. J Clin Oncol. 2014; 32:3723–3728. PMID: 25245447.
43. Markman TM, Nazarian S. Arrhythmia and electrophysiological effects of chemotherapy: a review. Oncology. 2016; 91:61–68. PMID: 27256307.
44. Ifran A, Kaptan K, Beyan C. High-dose cyclophosphamide and MESNA infusion can cause acute atrial fibrillation. Am J Hematol. 2005; 80:247. PMID: 16247745.
45. Rowinsky EK, McGuire WP, Guarnieri T, Fisherman JS, Christian MC, Donehower RC. Cardiac disturbances during the administration of taxol. J Clin Oncol. 1991; 9:1704–1712. PMID: 1678781.
46. Kamineni P, Prakasa K, Hasan SP, Ravi A, Dawkins F. Cardiotoxicities of paclitaxel in African Americans. J Natl Med Assoc. 2004; 96:995. PMID: 15253334.
47. McGuire WP, Rowinsky EK, Rosenshein NB, Grumbine FC, Ettinger DS, Armstrong DK, Donehower RC. Taxol: a unique antineoplastic agent with significant activity in advanced ovarian epithelial neoplasms. Ann Intern Med. 1989; 111:273–279. PMID: 2569287.
48. Jain M, Townsend RR. Chemotherapy agents and hypertension: a focus on angiogenesis blockade. Curr Hypertens Rep. 2007; 9:320–328. PMID: 17686384.
49. Colt JS, Schwartz K, Graubard BI, Davis F, Ruterbusch J, DiGaetano R, Purdue M, Rothman N, Wacholder S, Chow WH. Hypertension and risk of renal cell carcinoma among white and black Americans. Epidemiology. 2011; 22:797–804. PMID: 21881515.
50. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003; 9:669–676. PMID: 12778165.
51. Milan A, Puglisi E, Ferrari L, Bruno G, Losano I, Veglio F. Arterial hypertension and cancer. Int J Cancer. 2014; 134:2269–2277. PMID: 23784914.
52. Izzedine H, Ederhy S, Goldwasser F, Soria JC, Milano G, Cohen A, Khayat D, Spano JP. Management of hypertension in angiogenesis inhibitor-treated patients. Ann Oncol. 2009; 20:807–815. PMID: 19150949.
53. Chu TF, Rupnick MA, Kerkela R, Dallabrida SM, Zurakowski D, Nguyen L, Woulfe K, Pravda E, Cassiola F, Desai J, George S, Morgan JA, Harris DM, Ismail NS, Chen JH, Schoen FJ, Van den Abbeele AD, Demetri GD, Force T, Chen MH. Cardiotoxicity associated with tyrosine kinase inhibitor sunitinib. Lancet. 2007; 370:2011–2019. PMID: 18083403.
54. Aparicio-Gallego G, Afonso-Afonso FJ, León-Mateos L, Fírvida-Pérez JL, Vázquez-Estévez S, Lázaro-Quintela M, Ramos-Vázquez M, Fernández-Calvo O, Campos-Balea B, Antón-Aparicio LM. Molecular basis of hypertension side effects induced by sunitinib. Anticancer Drugs. 2011; 22:1–8. PMID: 20938340.
55. Eremina V, Jefferson JA, Kowalewska J, Hochster H, Haas M, Weisstuch J, Richardson C, Kopp JB, Kabir MG, Backx PH, Gerber HP, Ferrara N, Barisoni L, Alpers CE, Quaggin SE. VEGF inhibition and renal thrombotic microangiopathy. N Engl J Med. 2008; 358:1129–1136. PMID: 18337603.
56. Qi WX, He AN, Shen Z, Yao Y. Incidence and risk of hypertension with a novel multi-targeted kinase inhibitor axitinib in cancer patients: a systematic review and meta-analysis. Br J Clin Pharmacol. 2013; 76:348–357. PMID: 23617405.
57. Di Nisio M, Ferrante N, Feragalli B, De Tursi M, Iacobelli S, Cuccurullo F, Porreca E. Arterial thrombosis in ambulatory cancer patients treated with chemotherapy. Thromb Res. 2011; 127:382–383. PMID: 21056904.
58. Choueiri TK, Schutz FA, Je Y, Rosenberg JE, Bellmunt J. Risk of arterial thromboembolic events with sunitinib and sorafenib: a systematic review and meta-analysis of clinical trials. J Clin Oncol. 2010; 28:2280–2285. PMID: 20351323.
59. Schutz FA, Je Y, Azzi GR, Nguyen PL, Choueiri TK. Bevacizumab increases the risk of arterial ischemia: a large study in cancer patients with a focus on different subgroup outcomes. Ann Oncol. 2011; 22:1404–1412. PMID: 21115602.
60. Riedl J, Kaider A, Reitter EM, Marosi C, Jäger U, Schwarzinger I, Zielinski C, Pabinger I, Ay C. Association of mean platelet volume with risk of venous thromboembolism and mortality in patients with cancer. Results from the Vienna Cancer and Thrombosis Study (CATS). Thromb Haemost. 2014; 111:670–678. PMID: 24306221.
61. Ay C, Pabinger I, Cohen AT. Cancer-associated venous thromboembolism: burden, mechanisms, and management. Thromb Haemost. 2017; 117:219–230. PMID: 27882374.
62. Khorana AA, Kuderer NM, Culakova E, Lyman GH, Francis CW. Development and validation of a predictive model for chemotherapy-associated thrombosis. Blood. 2008; 111:4902–4907. PMID: 18216292.
63. Lee YG, Lee E, Kim I, Lee KW, Kim TM, Lee SH, Kim DW, Heo DS. Cisplatin-based chemotherapy is a strong risk factor for thromboembolic events in small-cell lung cancer. Cancer Res Treat. 2015; 47:670–675. PMID: 25672586.
64. Guignabert C, Phan C, Seferian A, Huertas A, Tu L, Thuillet R, Sattler C, Le Hiress M, Tamura Y, Jutant EM, Chaumais MC, Bouchet S, Manéglier B, Molimard M, Rousselot P, Sitbon O, Simonneau G, Montani D, Humbert M. Dasatinib induces lung vascular toxicity and predisposes to pulmonary hypertension. J Clin Invest. 2016; 126:3207–3218. PMID: 27482885.
65. Montani D, Bergot E, Günther S, Savale L, Bergeron A, Bourdin A, Bouvaist H, Canuet M, Pison C, Macro M, Poubeau P, Girerd B, Natali D, Guignabert C, Perros F, O'Callaghan DS, Jaïs X, Tubert-Bitter P, Zalcman G, Sitbon O, Simonneau G, Humbert M. Pulmonary arterial hypertension in patients treated by dasatinib. Circulation. 2012; 125:2128–2137. PMID: 22451584.
66. Alias S, Redwan B, Panzenboeck A, Winter MP, Schubert U, Voswinckel R, Frey MK, Jakowitsch J, Alimohammadi A, Hobohm L, Mangold A, Bergmeister H, Sibilia M, Wagner EF, Mayer E, Klepetko W, Hoelzenbein TJ, Preissner KT, Lang IM. Defective angiogenesis delays thrombus resolution: a potential pathogenetic mechanism underlying chronic thromboembolic pulmonary hypertension. Arterioscler Thromb Vasc Biol. 2014; 34:810–819. PMID: 24526692.
67. Pohjola-Sintonen S, Tötterman KJ, Salmo M, Siltanen P. Late cardiac effects of mediastinal radiotherapy in patients with Hodgkin's disease. Cancer. 1987; 60:31–37. PMID: 3581032.
68. Hayek ER, Speakman E, Rehmus E. Acute doxorubicin cardiotoxicity. N Engl J Med. 2005; 352:2456–2457. PMID: 15944435.
69. Bristow MR, Thompson PD, Martin RP, Mason JW, Billingham ME, Harrison DC. Early anthracycline cardiotoxicity. Am J Med. 1978; 65:823–832. PMID: 707541.
70. Chung WB, Yi JE, Jin JY, Choi YS, Park CS, Park WC, Song BJ, Youn HJ. Early cardiac function monitoring for detection of subclinical doxorubicin cardiotoxicity in young adult patients with breast cancer. J Breast Cancer. 2013; 16:178–183. PMID: 23843850.
71. Lotrionte M, Biondi-Zoccai G, Abbate A, Lanzetta G, D'Ascenzo F, Malavasi V, Peruzzi M, Frati G, Palazzoni G. Review and meta-analysis of incidence and clinical predictors of anthracycline cardiotoxicity. Am J Cardiol. 2013; 112:1980–1984. PMID: 24075281.
72. Suter TM, Ewer MS. Cancer drugs and the heart: importance and management. Eur Heart J. 2013; 34:1102–1111. PMID: 22789916.
73. Bickford CL, Yeh ET. Cardiotoxicity of other anti-cancer treatment. In : Ewer MS, Yeh ET, editors. Cancer and the heart. 2nd ed. Shelton (CT): People's Medical Publishing House - USA, Ltd;2013. p. 69–82.
74. Floyd JD, Nguyen DT, Lobins RL, Bashir Q, Doll DC, Perry MC. Cardiotoxicity of cancer therapy. J Clin Oncol. 2005; 23:7685–7696. PMID: 16234530.
75. Czaykowski PM, Moore MJ, Tannock IF. High risk of vascular events in patients with urothelial transitional cell carcinoma treated with cisplatin based chemotherapy. J Urol. 1998; 160(6 Pt 1):2021–2024. PMID: 9817314.
76. Kim SM, Kwak CH, Lee B, Kim SB, Sir JJ, Cho WH, Choi SK. A case of severe coronary spasm associated with 5-fluorouracil chemotherapy. Korean J Intern Med. 2012; 27:342–345. PMID: 23019400.
77. Seidman A, Hudis C, Pierri MK, Shak S, Paton V, Ashby M, Murphy M, Stewart SJ, Keefe D. Cardiac dysfunction in the trastuzumab clinical trials experience. J Clin Oncol. 2002; 20:1215–1221. PMID: 11870163.
78. Romond EH, Perez EA, Bryant J, Suman VJ, Geyer CE Jr, Davidson NE, Tan-Chiu E, Martino S, Paik S, Kaufman PA, Swain SM, Pisansky TM, Fehrenbacher L, Kutteh LA, Vogel VG, Visscher DW, Yothers G, Jenkins RB, Brown AM, Dakhil SR, Mamounas EP, Lingle WL, Klein PM, Ingle JN, Wolmark N. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med. 2005; 353:1673–1684. PMID: 16236738.
79. Perez EA, Suman VJ, Davidson NE, Sledge GW, Kaufman PA, Hudis CA, Martino S, Gralow JR, Dakhil SR, Ingle JN, Winer EP, Gelmon KA, Gersh BJ, Jaffe AS, Rodeheffer RJ. 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. PMID: 18250349.
80. Guarneri V, Lenihan DJ, Valero V, Durand JB, Broglio K, Hess KR, Michaud LB, Gonzalez-Angulo AM, Hortobagyi GN, Esteva FJ. Long-term cardiac tolerability of trastuzumab in metastatic breast cancer. J Clin Oncol. 2006; 24:4107–4115. PMID: 16908934.
81. Gianni L, Salvatorelli E, Minotti G. Anthracycline cardiotoxicity in breast cancer patients: synergism with trastuzumab and taxanes. Cardiovasc Toxicol. 2007; 7:67–71. PMID: 17652806.
82. Guglin M, Cutro R, Mishkin JD. Trastuzumab-induced cardiomyopathy. J Card Fail. 2008; 14:437–444. PMID: 18514938.
83. Tan-Chiu E, Yothers G, Romond E, Geyer CE Jr, Ewer M, Keefe D, Shannon RP, Swain SM, Brown A, Fehrenbacher L, Vogel VG, Seay TE, Rastogi P, Mamounas EP, Wolmark N, Bryant J. 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. PMID: 16258083.
84. Cardinale D, Colombo A, Torrisi R, Sandri MT, Civelli M, Salvatici M, Lamantia G, Colombo N, Cortinovis S, Dessanai MA, Nolè F, Veglia F, Cipolla CM. Trastuzumab-induced cardiotoxicity: clinical and prognostic implications of troponin I evaluation. J Clin Oncol. 2010; 28:3910–3916. PMID: 20679614.
85. Ky B, Putt M, Sawaya H, French B, Januzzi JL Jr, Sebag IA, Plana JC, Cohen V, Banchs J, Carver JR, Wiegers SE, Martin RP, Picard MH, Gerszten RE, Halpern EF, Passeri J, Kuter I, Scherrer-Crosbie 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. PMID: 24291281.
86. Thavendiranathan P, Poulin F, Lim KD, Plana JC, Woo A, Marwick TH. Use of myocardial strain imaging by echocardiography for the early detection of cardiotoxicity in patients during and after cancer chemotherapy: a systematic review. J Am Coll Cardiol. 2014; 63(25 Pt A):2751–2768. PMID: 24703918.
87. Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, Negrier S, Chevreau C, Solska E, Desai AA, Rolland F, Demkow T, Hutson TE, Gore M, Freeman S, Schwartz B, Shan M, Simantov R, Bukowski RM. TARGET Study Group. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007; 356:125–134. PMID: 17215530.
88. Verheul HM, Pinedo HM. Possible molecular mechanisms involved in the toxicity of angiogenesis inhibition. Nat Rev Cancer. 2007; 7:475–485. PMID: 17522716.
89. Miller K, Wang M, Gralow J, Dickler M, Cobleigh M, Perez EA, Shenkier T, Cella D, Davidson NE. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med. 2007; 357:2666–2676. PMID: 18160686.
90. Miller KD, Chap LI, Holmes FA, Cobleigh MA, Marcom PK, Fehrenbacher L, Dickler M, Overmoyer BA, Reimann JD, Sing AP, Langmuir V, Rugo HS. Randomized phase III trial of capecitabine compared with bevacizumab plus capecitabine in patients with previously treated metastatic breast cancer. J Clin Oncol. 2005; 23:792–799. PMID: 15681523.
91. Elitok A, Oz F, Cizgici AY, Kilic L, Ciftci R, Sen F, Bugra Z, Mercanoglu F, Oncul A, Oflaz H. Effect of carvedilol on silent anthracycline-induced cardiotoxicity assessed by strain imaging: a prospective randomized controlled study with six-month follow-up. Cardiol J. 2014; 21:509–515. PMID: 24142687.
92. Gulati G, Heck SL, Ree AH, Hoffmann P, Schulz-Menger J, Fagerland MW, Gravdehaug B, von Knobelsdorff-Brenkenhoff F, Bratland Å, Storås TH, Hagve TA, Røsjø H, Steine K, Geisler J, Omland T. 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. PMID: 26903532.
93. Kaya MG, Ozkan M, Gunebakmaz O, Akkaya H, Kaya EG, Akpek M, Kalay N, Dikilitas M, Yarlioglues M, Karaca H, Berk V, Ardic I, Ergin A, Lam YY. Protective effects of nebivolol against anthracycline-induced cardiomyopathy: a randomized control study. Int J Cardiol. 2013; 167:2306–2310. PMID: 22727976.
94. Cardinale D, Colombo A, Lamantia G, Colombo N, Civelli M, De Giacomi G, Rubino M, Veglia F, Fiorentini C, Cipolla CM. Anthracycline-induced cardiomyopathy: clinical relevance and response to pharmacologic therapy. J Am Coll Cardiol. 2010; 55:213–220. PMID: 20117401.
95. Bosch X, Rovira M, Sitges M, Domènech A, Ortiz-Pérez JT, de Caralt TM, Morales-Ruiz M, Perea RJ, Monzó M, Esteve J. Enalapril and carvedilol for preventing chemotherapy-induced left ventricular systolic dysfunction in patients with malignant hemopathies: the OVERCOME trial (preventiOn of left Ventricular dysfunction with Enalapril and caRvedilol in patients submitted to intensive ChemOtherapy for the treatment of Malignant hEmopathies). J Am Coll Cardiol. 2013; 61:2355–2362. PMID: 23583763.
96. Kalam K, Marwick TH. Role of cardioprotective therapy for prevention of cardiotoxicity with chemotherapy: a systematic review and meta-analysis. Eur J Cancer. 2013; 49:2900–2909. PMID: 23706982.
97. van Dalen EC, Caron HN, Dickinson HO, Kremer LC. Cardioprotective interventions for cancer patients receiving anthracyclines. Cochrane Database Syst Rev. 2011; 6. 15. [Epub]. DOI: 10.1002/14651858.CD003917.pub4.
98. Hensley ML, Hagerty KL, Kewalramani T, Green DM, Meropol NJ, Wasserman TH, Cohen GI, Emami B, Gradishar WJ, Mitchell RB, Thigpen JT, Trotti A 3rd, von Hoff D, Schuchter LM. American Society of Clinical Oncology 2008 clinical practice guideline update: use of chemotherapy and radiation therapy protectants. J Clin Oncol. 2009; 27:127–145. PMID: 19018081.
99. Acar Z, Kale A, Turgut M, Demircan S, Durna K, Demir S, Meriç M, Ağaç MT. Efficiency of atorvastatin in the protection of anthracycline-induced cardiomyopathy. J Am Coll Cardiol. 2011; 58:988–989. PMID: 21851890.
100. Seicean S, Seicean A, Plana JC, Budd GT, Marwick TH. Effect of statin therapy on the risk for incident heart failure in patients with breast cancer receiving anthracycline chemotherapy: an observational clinical cohort study. J Am Coll Cardiol. 2012; 60:2384–2390. PMID: 23141499.
101. Chow EJ, Chen Y, Kremer LC, Breslow NE, Hudson MM, Armstrong GT, Border WL, Feijen EA, Green DM, Meacham LR, Meeske KA, Mulrooney DA, Ness KK, Oeffinger KC, Sklar CA, Stovall M, van der Pal HJ, Weathers RE, Robison LL, Yasui Y. Individual prediction of heart failure among childhood cancer survivors. J Clin Oncol. 2015; 33:394–402. PMID: 25287823.
102. Mendelsohn ME, Karas RH. Molecular and cellular basis of cardiovascular gender differences. Science. 2005; 308:1583–1587. PMID: 15947175.
103. El-Kareh AW, Secomb TW. A mathematical model for comparison of bolus injection, continuous infusion, and liposomal delivery of doxorubicin to tumor cells. Neoplasia. 2000; 2:325–338. PMID: 11005567.
104. van Dalen EC, Michiels EM, Caron HN, Kremer LC. Different anthracycline derivates for reducing cardiotoxicity in cancer patients. Cochrane Database Syst Rev. 2010; 05–12. [Epub]. DOI: 10.1002/14651858.CD005006.pub4.
105. Seymour LW. Passive tumor targeting of soluble macromolecules and drug conjugates. Crit Rev Ther Drug Carrier Syst. 1992; 9:135–187. PMID: 1386002.
106. Ctep.cancer.gov [Internet] National Cancer Institute. Cancer Therapy Evaluation Program, Common Terminology Criteria for Adverse Events (CT-CAE), ver 5.0. updated 2017 Nov 27. cited Feb 26, 2018. Available from: https://ctep.cancer.gov/protocolDevelopment/electronic_applications/ctc.htm#ctc_50.
107. Ezaz G, Long JB, Gross CP, Chen J. Risk prediction model for heart failure and cardiomyopathy after adjuvant trastuzumab therapy for breast cancer. J Am Heart Assoc. 2014; 3:e000472. PMID: 24584736.
108. Witteles RM, Fowler MB, Telli ML. Chemotherapy-associated cardiotoxicity: how often does it really occur and how can it be prevented? Heart Fail Clin. 2011; 7:333–344. PMID: 21749885.
109. Carver JR, Shapiro CL, Ng A, Jacobs L, Schwartz C, Virgo KS, Hagerty KL, Somerfield MR, Vaughn DJ. ASCO Cancer Survivorship Expert Panel. American Society of Clinical Oncology clinical evidence review on the ongoing care of adult cancer survivors: cardiac and pulmonary late effects. J Clin Oncol. 2007; 25:3991–4008. PMID: 17577017.
110. Suter TM, Procter M, van Veldhuisen DJ, Muscholl M, Bergh J, Carlomagno C, Perren T, Passalacqua R, Bighin C, Klijn JG, Ageev FT, Hitre E, Groetz J, Iwata H, Knap M, Gnant M, Muehlbauer S, Spence A, Gelber RD, Piccart-Gebhart MJ. Trastuzumab-associated cardiac adverse effects in the herceptin adjuvant trial. J Clin Oncol. 2007; 25:3859–3865. PMID: 17646669.
111. Jones AL, Barlow M, Barrett-Lee PJ, Canney PA, Gilmour IM, Robb SD, Plummer CJ, Wardley AM, Verrill MW. Management of cardiac health in trastuzumab-treated patients with breast cancer: updated United Kingdom National Cancer Research Institute recommendations for monitoring. Br J Cancer. 2009; 100:684–692. PMID: 19259090.
112. Virani SA, Dent S, Brezden-Masley C, Clarke B, Davis MK, Jassal DS, Johnson C, Lemieux J, Paterson I, Sebag IA, Simmons C, Sulpher J, Thain K, Thavendiranathan P, Wentzell JR, Wurtele N, Côté MA, Fine NM, Haddad H, Hayley BD, Hopkins S, Joy AA, Rayson D, Stadnick E, Straatman L. Canadian Cardiovascular society guidelines for evaluation and management of cardiovascular complications of cancer therapy. Can J Cardiol. 2016; 32:831–841. PMID: 27343741.
113. Hamo CE, Bloom MW, Cardinale D, Ky B, Nohria A, Baer L, Skopicki H, Lenihan DJ, Gheorghiade M, Lyon AR, Butler J. Cancer therapy-related cardiac dysfunction and heart failure: part 2: prevention, treatment, guidelines, and future directions. Circ Heart Fail. 2016; 9:e002843. PMID: 26839395.
114. Sawaya H, Sebag IA, Plana JC, Januzzi JL, Ky B, Tan TC, Cohen V, Banchs J, Carver JR, Wiegers SE, Martin RP, Picard MH, Gerszten RE, Halpern EF, Passeri J, Kuter I, Scherrer-Crosbie M. 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. PMID: 22744937.
115. Sawaya H, Sebag IA, Plana JC, Januzzi JL, Ky B, Cohen V, Gosavi S, Carver JR, Wiegers SE, Martin RP, Picard MH, Gerszten RE, Halpern EF, Passeri J, Kuter I, Scherrer-Crosbie M. Early detection and prediction of cardiotoxicity in chemotherapy-treated patients. Am J Cardiol. 2011; 107:1375–1380. PMID: 21371685.
116. Gulati G, Zhang KW, Scherrer-Crosbie M, Ky B. Cancer and cardiovascular disease: the use of novel echocardiography measures to predict subsequent cardiotoxicity in breast cancer treated with anthracyclines and trastuzumab. Curr Heart Fail Rep. 2014; 11:366–373. PMID: 25079445.
117. King A, Thambyrajah J, Leng E, Stewart MJ. Global longitudinal strain: a useful everyday measurement. Echo Res Pract. 2016; 3:85–93. PMID: 27628098.
118. Cardinale D, Sandri MT, Colombo A, Colombo N, Boeri M, Lamantia G, Civelli M, Peccatori F, Martinelli G, Fiorentini C, Cipolla CM. Prognostic value of troponin I in cardiac risk stratification of cancer patients undergoing high-dose chemotherapy. Circulation. 2004; 109:2749–2754. PMID: 15148277.
119. Sandri MT, Salvatici M, Cardinale D, Zorzino L, Passerini R, Lentati P, Leon M, Civelli M, Martinelli G, Cipolla CM. N-terminal pro-B-type natriuretic peptide after high-dose chemotherapy: a marker predictive of cardiac dysfunction. Clin Chem. 2005; 51:1405–1410. PMID: 15932966.
120. Cardinale D, Sandri MT, Martinoni A, Tricca A, Civelli M, Lamantia G, Cinieri S, Martinelli G, Cipolla CM, Fiorentini C. Left ventricular dysfunction predicted by early troponin I release after high-dose chemotherapy. J Am Coll Cardiol. 2000; 36:517–522. PMID: 10933366.
121. Ledwidge M, Gallagher J, Conlon C, Tallon E, O'Connell E, Dawkins I, Watson C, O'Hanlon R, Bermingham M, Patle A, Badabhagni MR, Murtagh G, Voon V, Tilson L, Barry M, McDonald L, Maurer B, McDonald K. Natriuretic peptide-based screening and collaborative care for heart failure: the STOP-HF randomized trial. JAMA. 2013; 310:66–74. PMID: 23821090.
122. Wang YD, Chen SX, Ren LQ. Serum B-type natriuretic peptide levels as a marker for anthracycline-induced cardiotoxicity. Oncol Lett. 2016; 11:3483–3492. PMID: 27123140.
123. Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JW, Comber H, Forman D, Bray F. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur J Cancer. 2013; 49:1374–1403. PMID: 23485231.
124. Todaro MC, Oreto L, Qamar R, Paterick TE, Carerj S, Khandheria BK. Cardioncology: state of the heart. Int J Cardiol. 2013; 168:680–687. PMID: 23639459.
125. Lancellotti P, Nkomo VT, Badano LP, Bergler-Klein J, Bogaert J, Davin L, Cosyns B, Coucke P, Dulgheru R, Edvardsen T, Gaemperli O, Galderisi M, Griffin B, Heidenreich PA, Nieman K, Plana JC, Port SC, Scherrer-Crosbie M, Schwartz RG, Sebag IA, Voigt JU, Wann S, Yang PC. European Society of Cardiology Working Groups on Nuclear Cardiology and Cardiac Computed Tomography and Cardiovascular Magnetic Resonance. American Society of Nuclear Cardiology; Society for Cardiovascular Magnetic Resonance. Society of Cardiovascular Computed Tomography. Expert consensus for multi-modality imaging evaluation of cardiovascular complications of radiotherapy in adults: a report from the European Association of Cardiovascular Imaging and the American Society of Echocardiography. Eur Heart J Cardiovasc Imaging. 2013; 14:721–740. PMID: 23847385.
126. Mor-Avi V, Lang RM, Badano LP, Belohlavek M, Cardim NM, Derumeaux G, Galderisi M, Marwick T, Nagueh SF, Sengupta PP, Sicari R, Smiseth OA, Smulevitz B, Takeuchi M, Thomas JD, Vannan M, Voigt JU, Zamorano JL. Current and evolving echocardiographic techniques for the quantitative evaluation of cardiac mechanics: ASE/EAE consensus statement on methodology and indications endorsed by the Japanese Society of Echocardiography. Eur J Echocardiogr. 2011; 12:167–205. PMID: 21385887.
127. Negishi K, Negishi T, Hare JL, Haluska BA, Plana JC, Marwick TH. Independent and incremental value of deformation indices for prediction of trastuzumab-induced cardiotoxicity. J Am Soc Echocardiogr. 2013; 26:493–498. PMID: 23562088.
128. Charbonnel C, Convers-Domart R, Rigaudeau S, Taksin AL, Baron N, Lambert J, Ghez S, Georges JL, Farhat H, Lambert J, Rousselot P, Livarek B. Assessment of global longitudinal strain at low-dose anthracycline-based chemotherapy, for the prediction of subsequent cardiotoxicity. Eur Heart J Cardiovasc Imaging. 2017; 18:392–401. PMID: 28064155.
129. Ho E, Brown A, Barrett P, Morgan RB, King G, Kennedy MJ, Murphy RT. Subclinical anthracycline- and trastuzumab-induced cardiotoxicity in the long-term follow-up of asymptomatic breast cancer survivors: a speckle tracking echocardiographic study. Heart. 2010; 96:701–707. PMID: 20424152.
130. Greenberg NL, Firstenberg MS, Castro PL, Main M, Travaglini A, Odabashian JA, Drinko JK, Rodriguez LL, Thomas JD, Garcia MJ. Doppler-derived myocardial systolic strain rate is a strong index of left ventricular contractility. Circulation. 2002; 105:99–105. PMID: 11772883.
131. Voigt JU, Pedrizzetti G, Lysyansky P, Marwick TH, Houle H, Baumann R, Pedri S, Ito Y, Abe Y, Metz S, Song JH, Hamilton J, Sengupta PP, Kolias TJ, d'Hooge J, Aurigemma GP, Thomas JD, Badano LP. Definitions for a common standard for 2D speckle tracking echocardiography: consensus document of the EACVI/ASE/Industry Task Force to standardize deformation imaging. Eur Heart J Cardiovasc Imaging. 2015; 16:1–11. PMID: 25525063.
132. Hering D, Faber L, Horstkotte D. Echocardiographic features of radiation-associated valvular disease. Am J Cardiol. 2003; 92:226–230. PMID: 12860232.
133. Hull MC, Morris CG, Pepine CJ, Mendenhall NP. Valvular dysfunction and carotid, subclavian, and coronary artery disease in survivors of hodgkin lymphoma treated with radiation therapy. JAMA. 2003; 290:2831–2837. PMID: 14657067.
134. Thavendiranathan P, Grant AD, Negishi T, Plana JC, Popović ZB, Marwick TH. Reproducibility of echocardiographic techniques for sequential assessment of left ventricular ejection fraction and volumes: application to patients undergoing cancer chemotherapy. J Am Coll Cardiol. 2013; 61:77–84. PMID: 23199515.
135. Thavendiranathan P, Wintersperger BJ, Flamm SD, Marwick TH. Cardiac MRI in the assessment of cardiac injury and toxicity from cancer chemotherapy: a systematic review. Circ Cardiovasc Imaging. 2013; 6:1080–1091. PMID: 24254478.
136. Cheong BY, Muthupillai R. Nephrogenic systemic fibrosis: a concise review for cardiologists. Tex Heart Inst J. 2010; 37:508–515. PMID: 20978560.
137. Friedrich MG, Sechtem U, Schulz-Menger J, Holmvang G, Alakija P, Cooper LT, White JA, Abdel-Aty H, Gutberlet M, Prasad S, Aletras A, Laissy JP, Paterson I, Filipchuk NG, Kumar A, Pauschinger M, Liu P. International Consensus Group on Cardiovascular Magnetic Resonance in Myocarditis. Cardiovascular magnetic resonance in myocarditis: A JACC White Paper. J Am Coll Cardiol. 2009; 53:1475–1487. PMID: 19389557.
138. Fallah-Rad N, Lytwyn M, Fang T, Kirkpatrick I, Jassal DS. Delayed contrast enhancement cardiac magnetic resonance imaging in trastuzumab induced cardiomyopathy. J Cardiovasc Magn Reson. 2008; 10:5. PMID: 18272009.
139. Tham EB, Haykowsky MJ, Chow K, Spavor M, Kaneko S, Khoo NS, Pagano JJ, Mackie AS, Thompson RB. Diffuse myocardial fibrosis by T1-mapping in children with subclinical anthracycline cardiotoxicity: relationship to exercise capacity, cumulative dose and remodeling. J Cardiovasc Magn Reson. 2013; 15:48. PMID: 23758789.
140. Jordan JH, D'Agostino RB Jr, Hamilton CA, Vasu S, Hall ME, Kitzman DW, Thohan V, Lawrence JA, Ellis LR, Lash TL, Hundley WG. Longitudinal assessment of concurrent changes in left ventricular ejection fraction and left ventricular myocardial tissue characteristics after administration of cardiotoxic chemotherapies using T1-weighted and T2-weighted cardiovascular magnetic resonance. Circ Cardiovasc Imaging. 2014; 7:872–879. PMID: 25273568.
141. Wassmuth R, Lentzsch S, Erdbruegger U, Schulz-Menger J, Doerken B, Dietz R, Friedrich MG. Subclinical cardiotoxic effects of anthracyclines as assessed by magnetic resonance imaging-a pilot study. Am Heart J. 2001; 141:1007–1013. PMID: 11376317.
142. Iles L, Pfluger H, Phrommintikul A, Cherayath J, Aksit P, Gupta SN, Kaye DM, Taylor AJ. Evaluation of diffuse myocardial fibrosis in heart failure with cardiac magnetic resonance contrast-enhanced T1 mapping. J Am Coll Cardiol. 2008; 52:1574–1580. PMID: 19007595.
143. Ugander M, Oki AJ, Hsu LY, Kellman P, Greiser A, Aletras AH, Sibley CT, Chen MY, Bandettini WP, Arai AE. Extracellular volume imaging by magnetic resonance imaging provides insights into overt and sub-clinical myocardial pathology. Eur Heart J. 2012; 33:1268–1278. PMID: 22279111.
144. Neilan TG, Coelho-Filho OR, Pena-Herrera D, Shah RV, Jerosch-Herold M, Francis SA, Moslehi J, Kwong RY. Left ventricular mass in patients with a cardiomyopathy after treatment with anthracyclines. Am J Cardiol. 2012; 110:1679–1686. PMID: 22917553.
145. Grover S, Lou PW, Bradbrook C, Cheong K, Kotasek D, Leong DP, Koczwara B, Selvanayagam JB. Early and late changes in markers of aortic stiffness with breast cancer therapy. Intern Med J. 2015; 45:140–147. PMID: 25404097.
146. Chaosuwannakit N, D'Agostino R Jr, Hamilton CA, Lane KS, Ntim WO, Lawrence J, Melin SA, Ellis LR, Torti FM, Little WC, Hundley WG. Aortic stiffness increases upon receipt of anthracycline chemotherapy. J Clin Oncol. 2010; 28:166–172. PMID: 19901105.
147. Vasu S, Hundley WG. Understanding cardiovascular injury after treatment for cancer: an overview of current uses and future directions of cardiovascular magnetic resonance. J Cardiovasc Magn Reson. 2013; 15:66. PMID: 23902649.
148. Schwartz RG, Jain D, Storozynsky E. Traditional and novel methods to assess and prevent chemotherapy-related cardiac dysfunction noninvasively. J Nucl Cardiol. 2013; 20:443–464. PMID: 23572315.
149. de Geus-Oei LF, Mavinkurve-Groothuis AM, Bellersen L, Gotthardt M, Oyen WJ, Kapusta L, van Laarhoven HW. Scintigraphic techniques for early detection of cancer treatment-induced cardiotoxicity. J Nucl Med Technol. 2013; 41:170–181. PMID: 23929800.
150. Altena R, Perik PJ, van Veldhuisen DJ, de Vries EG, Gietema JA. Cardiovascular toxicity caused by cancer treatment: strategies for early detection. Lancet Oncol. 2009; 10:391–399. PMID: 19341970.
151. Schwartz RG, McKenzie WB, Alexander J, Sager P, D'Souza A, Manatunga A, Schwartz PE, Berger HJ, Setaro J, Surkin L, Wackers FJ, Zaret BL. Congestive heart failure and left ventricular dysfunction complicating doxorubicin therapy. Seven-year experience using serial radionuclide angiocardiography. Am J Med. 1987; 82:1109–1118. PMID: 3605130.
152. Groch MW, DePuey EG, Belzberg AC, Erwin WD, Kamran M, Barnett CA, Hendel RC, Spies SM, Ali A, Marshall RC. Planar imaging versus gated blood-pool SPECT for the assessment of ventricular performance: a multicenter study. J Nucl Med. 2001; 42:1773–1779. PMID: 11752072.
153. Castro P, Winter JL, Verdejo H, Orellana P, Quintana JC, Greig D, Enríquez A, Sepúlveda L, Concepción R, Sepúlveda P, Rossel V, Chiong M, García L, Lavandero S. Relationship between mechanical and metabolic dyssynchrony with left bundle branch block: evaluation by 18-fluorodeoxyglucose positron emission tomography in patients with non-ischemic heart failure. J Heart Lung Transplant. 2012; 31:1096–1101. PMID: 22975099.
154. Perrone-Filardi P, Paolillo S, Dellegrottaglie S, Gargiulo P, Savarese G, Marciano C, Casaretti L, Cecere M, Musella F, Pirozzi E, Parente A, Cuocolo A. Assessment of cardiac sympathetic activity by MIBG imaging in patients with heart failure: a clinical appraisal. Heart. 2011; 97:1828–1833. PMID: 21917663.
155. Carrió I, Estorch M, Berná L, López-Pousa J, Tabernero J, Torres G. Indium-111-antimyosin and iodine-123-MIBG studies in early assessment of doxorubicin cardiotoxicity. J Nucl Med. 1995; 36:2044–2049. PMID: 7472595.
156. Carrió I, Lopez-Pousa A, Estorch M, Duncker D, Berná L, Torres G, de Andrés L. Detection of doxorubicin cardiotoxicity in patients with sarcomas by indium-111-antimyosin monoclonal antibody studies. J Nucl Med. 1993; 34:1503–1507. PMID: 8355070.
157. Estorch M, Carrió I, Martínez-Duncker D, Berná L, Torres G, Alonso C, Ojeda B. Myocyte cell damage after administration of doxorubicin or mitoxantrone in breast cancer patients assessed by indium 111 antimyosin monoclonal antibody studies. J Clin Oncol. 1993; 11:1264–1268. PMID: 8315423.
158. Jacobson AF, Senior R, Cerqueira MD, Wong ND, Thomas GS, Lopez VA, Agostini D, Weiland F, Chandna H, Narula J. ADMIRE-HF Investigators. Myocardial iodine-123 meta-iodobenzylguanidine imaging and cardiac events in heart failure. Results of the prospective ADMIRE-HF (AdreView Myocardial Imaging for Risk Evaluation in Heart Failure) study. J Am Coll Cardiol. 2010; 55:2212–2221. PMID: 20188504.
159. Saito K, Takeda K, Imanaka-Yoshida K, Imai H, Sekine T, Kamikura Y. Assessment of fatty acid metabolism in taxan-induced myocardial damage with iodine-123 BMIPP SPECT: comparative study with myocardial perfusion, left ventricular function, and histopathological findings. Ann Nucl Med. 2003; 17:481–488. PMID: 14575384.
160. Giedd KN, Bergmann SR. Fatty acid imaging of the heart. Curr Cardiol Rep. 2011; 13:121–131. PMID: 21240641.
161. Behr TM, Béhé M, Wörmann B. Trastuzumab and breast cancer. N Engl J Med. 2001; 345:995–996. PMID: 11575295.
162. Gaykema SB, de Jong JR, Perik PJ, Brouwers AH, Schröder CP, Oude Munnink TH, Bongaerts AH, de Vries EG, Lub-de Hooge MN. (111)In-trastuzumab scintigraphy in HER2-positive metastatic breast cancer patients remains feasible during trastuzumab treatment. Mol Imaging. 2014; 7. 01. [Epub]. DOI: 10.2310/7290.2014.00011.
163. Perik PJ, Lub-De Hooge MN, Gietema JA, van der Graaf WT, de Korte MA, Jonkman S, Kosterink JG, van Veldhuisen DJ, Sleijfer DT, Jager PL, de Vries EG. Indium-111-labeled trastuzumab scintigraphy in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer. J Clin Oncol. 2006; 24:2276–2282. PMID: 16710024.
164. Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE Jr, Drazner MH, Fonarow GC, Geraci SA, Horwich T, Januzzi JL, Johnson MR, Kasper EK, Levy WC, Masoudi FA, McBride PE, McMurray JJ, Mitchell JE, Peterson PN, Riegel B, Sam F, Stevenson LW, Tang WH, Tsai EJ, Wilkoff BL;. American College of Cardiology Foundation. American Heart Association Task Force on Practice Guidelines. 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2013; 62:e147–e239. PMID: 23747642.
165. Ponikowski P, Voors AA, Anker SD, Bueno H, Cleland JGF, Coats AJS, Falk V, González-Juanatey JR, Harjola VP, Jankowska EA, Jessup M, Linde C, Nihoyannopoulos P, Parissis JT, Pieske B, Riley JP, Rosano GMC, Ruilope LM, Ruschitzka F, Rutten FH, van der Meer P. ESC Scientific Document Group. 2016 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC) developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur Heart J. 2016; 37:2129–2200. PMID: 27206819.
166. Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE Jr, Colvin MM, Drazner MH, Filippatos G, Fonarow GC, Givertz MM, Hollenberg SM, Lindenfeld J, Masoudi FA, McBride PE, Peterson PN, Stevenson LW, Westlake C. 2016 ACC/AHA/HFSA focused update on new pharmacological therapy for heart failure: an update of the 2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology/American Heart Association Task Force on clinical practice guidelines and the Heart Failure Society of America. J Am Coll Cardiol. 2016; 68:1476–1488. PMID: 27216111.
167. Cardinale D, Colombo A, Sandri MT, Lamantia G, Colombo N, Civelli M, Martinelli G, Veglia F, Fiorentini C, Cipolla CM. Prevention of high-dose chemotherapy-induced cardiotoxicity in high-risk patients by angiotensin-converting enzyme inhibition. Circulation. 2006; 114:2474–2481. PMID: 17101852.
168. Ewer MS, Vooletich MT, Durand JB, Woods ML, Davis JR, Valero V, Lenihan DJ. Reversibility of trastuzumab-related cardiotoxicity: new insights based on clinical course and response to medical treatment. J Clin Oncol. 2005; 23:7820–7826. PMID: 16258084.
169. Arteaga CL, Sliwkowski MX, Osborne CK, Perez EA, Puglisi F, Gianni L. Treatment of HER2-positive breast cancer: current status and future perspectives. Nat Rev Clin Oncol. 2011; 9:16–32. PMID: 22124364.
170. Goldhirsch A, Gelber RD, Piccart-Gebhart MJ, de Azambuja E, Procter M, Suter TM, Jackisch C, Cameron D, Weber HA, Heinzmann D, Dal Lago L, McFadden E, Dowsett M, Untch M, Gianni L, Bell R, Köhne CH, Vindevoghel A, Andersson M, Brunt AM, Otero-Reyes D, Song S, Smith I, Leyland-Jones B, Baselga J. Herceptin Adjuvant (HERA) Trial Study Team. HERA) Trial Study Team. 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. PMID: 23871490.
171. Swain SM, Kim SB, Cortés J, Ro J, Semiglazov V, Campone M, Ciruelos E, Ferrero JM, Schneeweiss A, Knott A, Clark E, Ross G, Benyunes MC, Baselga J. Pertuzumab, trastuzumab, and docetaxel for HER2-positive metastatic breast cancer (CLEOPATRA study): overall survival results from a randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2013; 14:461–471. PMID: 23602601.
172. Jensen BV, Skovsgaard T, Nielsen SL. Functional monitoring of anthracycline cardiotoxicity: a prospective, blinded, long-term observational study of outcome in 120 patients. Ann Oncol. 2002; 13:699–709. PMID: 12075737.
173. Cianci G, Morelli MF, Cannita K, Morese R, Ricevuto E, Di Rocco ZC, Porzio G, Lanfiuti Baldi P, Ficorella C. Prophylactic options in patients with 5-fluorouracil-associated cardiotoxicity. Br J Cancer. 2003; 88:1507–1509. PMID: 12771913.
174. Fihn SD, Blankenship JC, Alexander KP, Bittl JA, Byrne JG, Fletcher BJ, Fonarow GC, Lange RA, Levine GN, Maddox TM, Naidu SS, Ohman EM, Smith PK. 2014 ACC/AHA/AATS/PCNA/SCAI/STS focused update of the guideline for the diagnosis and management of patients with stable ischemic heart disease: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines, and the American Association for Thoracic Surgery, Preventive Cardiovascular Nurses Association, Society for Cardiovascular Angiography and Interventions, and Society of Thoracic Surgeons. Circulation. 2014; 130:1749–1767. PMID: 25070666.
175. Amsterdam EA, Wenger NK, Brindis RG, Casey DE Jr, Ganiats TG, Holmes DR Jr, Jaffe AS, Jneid H, Kelly RF, Kontos MC, Levine GN, Liebson PR, Mukherjee D, Peterson ED, Sabatine MS, Smalling RW, Zieman SJ. 2014 AHA/ACC guideline for the management of patients with non-ST-elevation acute coronary syndromes: a report of the American College of Cardiology/ American Heart Association Task Force on Practice Guidelines. Circulation. 2014; 130:e344–e426. PMID: 25249585.
176. Iannaccone M, D Ascenzo F, De Filippo O, Gagliardi M, Southern DA, Raposeiras-Roubín S, Abu-Assi E, Henriques JPS, Saucedo J, González-Juanatey JR, Wilton SB, Kikkert WJ, Nuñez-Gil I, Ariza-Sole A, Song X, Alexopoulos D, Liebetrau C, Kawaji T, Huczek Z, Nie SP, Fujii T, Correia L, Kawashiri MA, García-Acuña JM, Alfonso E, Terol B, Garay A, Zhang D, Chen Y, Xanthopoulou I, Osman N, Möllmann H, Shiomi H, Kowara M, Filipiak K, Wang X, Yan Y, Fan JY, Ikari Y, Nakahashi T, Sakata K, Yamagishi M, Moretti C, Gaita F, Kalpak O, Kedev S. Optimal medical therapy in patients with malignancy undergoing percutaneous coronary intervention for acute coronary syndrome: a BleeMACS sub-study. Am J Cardiovasc Drugs. 2017; 17:61–71. PMID: 27738920.
177. Vieira RD, Pereira AC, Lima EG, Garzillo CL, Rezende PC, Favarato D, Hueb AC, Gersh BJ, Ramires JA, Hueb W. Cancer-related deaths among different treatment options in chronic coronary artery disease: results of a 6-year follow-up of the MASS II study. Coron Artery Dis. 2012; 23:79–84. PMID: 22157410.
178. Iliescu CA, Grines CL, Herrmann J, Yang EH, Cilingiroglu M, Charitakis K, Hakeem A, Toutouzas KP, Leesar MA, Marmagkiolis K. SCAI expert consensus statement: evaluation, management, and special considerations of cardio-oncology patients in the cardiac catheterization laboratory (endorsed by the cardiological society of india, and sociedad Latino Americana de Cardiologia intervencionista). Catheter Cardiovasc Interv. 2016; 87:E202–E223. PMID: 26756277.
179. Strevel EL, Ing DJ, Siu LL. Molecularly targeted oncology therapeutics and prolongation of the QT interval. J Clin Oncol. 2007; 25:3362–3371. PMID: 17664484.
180. Farmakis D, Parissis J, Filippatos G. Insights into onco-cardiology: atrial fibrillation in cancer. J Am Coll Cardiol. 2014; 63:945–953. PMID: 24361314.
181. Bates SE, Rosing DR, Fojo T, Piekarz RL. Challenges of evaluating the cardiac effects of anticancer agents. Clin Cancer Res. 2006; 12:3871–3874. PMID: 16818679.
182. January CT, Wann LS, Alpert JS, Calkins H, Cigarroa JE, Cleveland JC Jr, Conti JB, Ellinor PT, Ezekowitz MD, Field ME, Murray KT, Sacco RL, Stevenson WG, Tchou PJ, Tracy CM, Yancy CW. American College of Cardiology/American Heart Association Task Force on Practice Guidelines. 2014 AHA/ACC/HRS guideline for the management of patients with atrial fibrillation: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines and the Heart Rhythm Society. J Am Coll Cardiol. 2014; 64:e1–e76. PMID: 24685669.
183. Lip GY, Ponikowski P, Andreotti F, Anker SD, Filippatos G, Homma S, Morais J, Pullicino P, Rasmussen LH, Marin F, Lane DA. ESC Task Force. Thrombo-embolism and antithrombotic therapy for heart failure in sinus rhythm. a joint consensus document from the ESC heart failure association and the ESC working group on thrombosis. Eur J Heart Fail. 2012; 14:681–695. PMID: 22611046.
184. Kirchhof P, Benussi S, Kotecha D, Ahlsson A, Atar D, Casadei B, Castella M, Diener HC, Heidbuchel H, Hendriks J, Hindricks G, Manolis AS, Oldgren J, Popescu BA, Schotten U, Van Putte B, Vardas P. ESC Scientific Document Group. 2016 ESC Guidelines for the management of atrial fibrillation developed in collaboration with EACTS. Eur Heart J. 2016; 37:2893–2962. PMID: 27567408.
185. Mathur P, Paydak H, Thanendrarajan S, van Rhee F. Atrial fibrillation in hematologic malignancies, especially after autologous hematopoietic stem cell transplantation: review of risk factors, current management, and future directions. Clin Lymphoma Myeloma Leuk. 2016; 16:70–75. PMID: 26776085.
186. Xiang E, Ahuja T, Raco V, Cirrone F, Green D, Papadopoulos J. Anticoagulation prescribing patterns in patients with cancer. J Thromb Thrombolysis. 2018; 45:89–98. PMID: 29052104.
187. Laube ES, Yu A, Gupta D, Miao Y, Samedy P, Wills J, Harnicar S, Soff GA, Mantha S. Rivaroxaban for stroke prevention in patients with nonvalvular atrial fibrillation and active cancer. Am J Cardiol. 2017; 120:213–217. PMID: 28549819.
188. James PA, Oparil S, Carter BL, Cushman WC, Dennison-Himmelfarb C, Handler J, Lackland DT, LeFevre ML, MacKenzie TD, Ogedegbe O, Smith SC Jr, Svetkey LP, Taler SJ, Townsend RR, Wright JT Jr, Narva AS, Ortiz E. 2014 evidence-based guideline for the management of high blood pressure in adults: report from the panel members appointed to the Eighth Joint National Committee (JNC 8). JAMA. 2014; 311:507–520. PMID: 24352797.
189. Lackland DT. Hypertension: joint national committee on detection, evaluation, and treatment of high blood pressure guidelines. Curr Opin Neurol. 2013; 26:8–12. PMID: 23241566.
190. Lyman GH, Bohlke K, Khorana AA, Kuderer NM, Lee AY, Arcelus JI, Balaban EP, Clarke JM, Flowers CR, Francis CW, Gates LE, Kakkar AK, Key NS, Levine MN, Liebman HA, Tempero MA, Wong SL, Somerfield MR, Falanga A;. American Society of Clinical Oncology. Venous thromboembolism prophylaxis and treatment in patients with cancer: American Society of Clinical Oncology clinical practice guideline update 2014. J Clin Oncol. 2015; 33:654–656. PMID: 25605844.
191. Khorana AA. Risk assessment and prophylaxis for VTE in cancer patients. J Natl Compr Canc Netw. 2011; 9:789–797. PMID: 21715725.
192. Akl EA, Kahale L, Barba M, Neumann I, Labedi N, Terrenato I, Sperati F, Muti P, Schünemann H. Anticoagulation for the long-term treatment of venous thromboembolism in patients with cancer. Cochrane Database Syst Rev. 2014; 7. 08. [Epub]. DOI: 10.1002/14651858.CD006650.
193. Prins MH, Lensing AW, Bauersachs R, van Bellen B, Bounameaux H, Brighton TA, Cohen AT, Davidson BL, Decousus H, Raskob GE, Berkowitz SD, Wells PS. EINSTEIN Investigators. Oral rivaroxaban versus standard therapy for the treatment of symptomatic venous thromboembolism: a pooled analysis of the EINSTEIN-DVT and PE randomized studies. Thromb J. 2013; 11:21. PMID: 24053656.
194. Schulman S, Goldhaber SZ, Kearon C, Kakkar AK, Schellong S, Eriksson H, Hantel S, Feuring M, Kreuzer J. Treatment with dabigatran or warfarin in patients with venous thromboembolism and cancer. Thromb Haemost. 2015; 114:150–157. PMID: 25739680.
195. Farge D, Debourdeau P, Beckers M, Baglin C, Bauersachs RM, Brenner B, Brilhante D, Falanga A, Gerotzafias GT, Haim N, Kakkar AK, Khorana AA, Lecumberri R, Mandala M, Marty M, Monreal M, Mousa SA, Noble S, Pabinger I, Prandoni P, Prins MH, Qari MH, Streiff MB, Syrigos K, Bounameaux H, Büller HR. International clinical practice guidelines for the treatment and prophylaxis of venous thromboembolism in patients with cancer. J Thromb Haemost. 2013; 11:56–70. PMID: 23217107.
196. Papanikolaou J, Spathoulas K, Makris D, Zakynthinos E. Thrombolysis for massive pulmonary embolism in a patient with hemorrhagic shock. Am J Respir Crit Care Med. 2016; 194:e15–e16. PMID: 27529567.
197. O'Connell CL, Liebman HA. Approach to the management of incidental venous thromboembolic events in patients with cancer. J Natl Compr Canc Netw. 2014; 12:1557–1560. PMID: 25361802.
198. Galiè N, Humbert M, Vachiery JL, Gibbs S, Lang I, Torbicki A, Simonneau G, Peacock A, Vonk Noordegraaf A, Beghetti M, Ghofrani A, Gomez Sanchez MA, Hansmann G, Klepetko W, Lancellotti P, Matucci M, McDonagh T, Pierard LA, Trindade PT, Zompatori M, Hoeper M, Aboyans V, Vaz Carneiro A, Achenbach S, Agewall S, Allanore Y, Asteggiano R, Paolo Badano L, Albert Barberà J, Bouvaist H, Bueno H, Byrne RA, Carerj S, Castro G, Erol Ç, Falk V, Funck-Brentano C, Gorenflo M, Granton J, Iung B, Kiely DG, Kirchhof P, Kjellstrom B, Landmesser U, Lekakis J, Lionis C, Lip GY, Orfanos SE, Park MH, Piepoli MF, Ponikowski P, Revel MP, Rigau D, Rosenkranz S, Völler H, Luis Zamorano J. 2015 ESC/ERS Guidelines for the diagnosis and treatment of pulmonary hypertension: The Joint Task Force for the Diagnosis and Treatment of Pulmonary Hypertension of the European Society of Cardiology (ESC) and the European Respiratory Society (ERS): Endorsed by: Association for European Paediatric and Congenital Cardiology (AEPC), International Society for Heart and Lung Transplantation (ISHLT). Eur Heart J. 2016; 37:67–119. PMID: 26320113.
199. Ranchoux B, Günther S, Quarck R, Chaumais MC, Dorfmüller P, Antigny F, Dumas SJ, Raymond N, Lau E, Savale L, Jaïs X, Sitbon O, Simonneau G, Stenmark K, Cohen-Kaminsky S, Humbert M, Montani D, Perros F. Chemotherapy-induced pulmonary hypertension: role of alkylating agents. Am J Pathol. 2015; 185:356–371. PMID: 25497573.
200. Adler Y, Charron P, Imazio M, Badano L, Barón-Esquivias G, Bogaert J, Brucato A, Gueret P, Klingel K, Lionis C, Maisch B, Mayosi B, Pavie A, Ristić AD, Sabaté Tenas M, Seferovic P, Swedberg K, Tomkowski W, Achenbach S, Agewall S, Al-Attar N, Angel Ferrer J, Arad M, Asteggiano R, Bueno H, Caforio AL, Carerj S, Ceconi C, Evangelista A, Flachskampf F, Giannakoulas G, Gielen S, Habib G, Kolh P, Lambrinou E, Lancellotti P, Lazaros G, Linhart A, Meurin P, Nieman K, Piepoli MF, Price S, Roos-Hesselink J, Roubille F, Ruschitzka F, Sagristà Sauleda J, Sousa-Uva M, Uwe Voigt J, Luis Zamorano. ESC). 2015 ESC guidelines for the diagnosis and management of pericardial diseases: The Task Force for the diagnosis and management of pericardial diseases of the European Society of Cardiology (ESC) endorsed by: The European Association for Cardio-Thoracic Surgery (EACTS). Eur Heart J. 2015; 36:2921–2964. PMID: 26320112.
201. Lestuzzi C, Berretta M, Tomkowski W. 2015 update on the diagnosis and management of neoplastic pericardial disease. Expert Rev Cardiovasc Ther. 2015; 13:377–389. PMID: 25797903.
202. Raval J, Nagaraja V, Eslick GD, Denniss AR. The role of colchicine in pericarditis--a systematic review and meta-analysis of randomised trials. Heart Lung Circ. 2015; 24:660–666. PMID: 25766664.
203. El Haddad D, Iliescu C, Yusuf SW, William WN Jr, Khair TH, Song J, Mouhayar EN. Outcomes of cancer patients undergoing percutaneous pericardiocentesis for pericardial effusion. J Am Coll Cardiol. 2015; 66:1119–1128. PMID: 26337990.
204. Feldman D, Pamboukian SV, Teuteberg JJ, Birks E, Lietz K, Moore SA, Morgan JA, Arabia F, Bauman ME, Buchholz HW, Deng M, Dickstein ML, El-Banayosy A, Elliot T, Goldstein DJ, Grady KL, Jones K, Hryniewicz K, John R, Kaan A, Kusne S, Loebe M, Massicotte MP, Moazami N, Mohacsi P, Mooney M, Nelson T, Pagani F, Perry W, Potapov EV, Eduardo Rame J, Russell SD, Sorensen EN, Sun B, Strueber M, Mangi AA, Petty MG, Rogers J. International Society for Heart and Lung Transplantation. The 2013 International Society for heart and lung transplantation guidelines for mechanical circulatory support: executive summary. J Heart Lung Transplant. 2013; 32:157–187. PMID: 23352391.
Fig. 1

Flow chart of surveillance and diagnosis of cardiac toxicity in breast cancer treatment. For pretreatment evaluation of breast cancer, conventional and CV risk factors should be assessed. In case of absence of risk factors, anti-cancer treatment could be proceeded. For surveillance of cardiac toxicity, cardiac biomarkers such as NT-proBNP or Tn-I may be useful at each chemotherapeutic cycle, and echocardiography is also required regularly according to the drugs. However, if risk factor is present, echocardiography (or CMR imaging) would be necessary for cardiac function. Overall, LV dysfunction or myocardial injury with elevated Tn-I can be managed with BB, ACEIs/ARBs, or statin. CV: cardiovascular, NT-proBNP: N-terminal pro-B-type natriuretic peptide, Tn-I: troponin-I, LV: left ventricle, CMR: cardiac magnetic resonance, BB: beta-blocker, ACEIs: angiotensin converting enzyme inhibitors, ARBs: angiotensin receptor blockers.

jcu-26-1-g001
Fig. 2

Approach of LV dysfunction in diagnostic or therapeutic modalities. Baseline cardiac imaging for LV function can be performed using 2D-echocardiography (or contrast-/3D-echocardigraphy) or strain. In case of LV dysfunction, cardio-oncologic consultation is required, and the etiologies of LV dysfunction should be evaluated using appropriate imaging modalities, including CMR, CAG, SPECT, and cardiac CT. ECG: electrocardiography, BNP: B-type natriuretic peptide, NT-proBNP: N-terminal pro-B-type natriuretic peptide, Tn-I: troponin-I, 2D: two-dimensional, 3D: three-dimensional, CMR: cardiac magnetic resonance, LV: left ventricle, CT: computed tomography, CAG: coronary angiography, SPECT: singlephoton emission computed tomography.

jcu-26-1-g002
Fig. 3

Flow chart of approach for cardiac dysfunction during breast cancer treatment. Treatment with BB, ACEIs/ARBs, or statin is required for LV dysfunction. According to the strain and LVEF using echocardiography, the risk-and-benefit for breast cancer treatment should be evaluated. If necessary, cancer treatment could be stopped for 4 weeks and re-evaluation of LV function is required using repeated echocardiography. Tn-I: troponin-I, BB: beta-blocker, ACEIs: angiotensin converting enzyme inhibitor, ARBs: angiotensin receptor blockers, LV: left ventricle, EF: ejection fraction, GLS: global longitudinal strain.

jcu-26-1-g003
Table 1

Common chemotherapeutic agents related with cardiomyopathy

jcu-26-1-i001
Group Agent
Anthracyclines Doxorubicin*
Epirubicin
Idarubicin*
Alkylating agents Cyclophosphamide*
Ifosfamide*
Antimetabolites Clofarabine*
Antimicrotuble agents Docetaxel*
Monoclonal antibody-based tyrosine kinase inhibitors Bevacizumab*
Trastuzumab*
Proteasome inhibitors Bortezomib
Small molecular tyrosine kinase inhibitors Sunitinib*
Imatinib mesylate
Lapatinib
Dasatinib

*Drugs are considered frequent and important for cardiomyopathy

Table 2

Common chemotherapeutic agents related with ischemic heart disease

jcu-26-1-i002
Group Agent
Antimetabolites Capecitabine*
Fluorouracil*
Antimicrotuble agents Docetaxel
Paclitaxel
Monoclonal antibody-based tyrosine kinase inhibitors Bevacizumab*
Small molecular tyrosine kinase inhibitors Sorafenib*
Eriotinib

*Drugs are considered frequent and important for cardiomyopathy

Table 3

Chemotherapeutic agents associated with cardiac arrhythmias

jcu-26-1-i003
Arrhythmia Agent
Sinus bradycardia Taxane, thalidomide
Atrioventricular block Taxane, anthracyclines, cyclophosphamide
5-FU, rituximab
Atrial fibrillation Alkylating agents, anthracyclines, taxane
5-FU, gemcitabine
Ventricular tachycardia Doxorubicin, alkylating agents

5-FU: 5-fluorouracil

Table 4

Chemotherapeutic agents associated with QT prolongation

jcu-26-1-i004
Group Agent
Tyrosine kinase inhibitors Sunitinib, sorafenib, vandetanib, nilotinib
Histone deacetylase inhibitors Vorinostat, despipeptide (FK-228, romidepsin), panobinostat
Anthracyclines Doxorubicin
Arsenic trioxide
Table 5

Chemotherapeutic agents associated with pericardial effusion

jcu-26-1-i005
Group Agent
Anthracycline Doxorubicin, daunorubicin
Tyrosine kinase inhibitors Dastanib, imatinib, nilotinib
Antimetabolites 5-FU, cytarabine
Alkylating agents Cyclophosphamide, ifosfamide, busulphan
All-trans retinoic acid

5-FU: 5-fluorouracil

Table 6

Comparison of relative cardiotoxicities according to type of anti-cancer treatment and schedules

jcu-26-1-i006
Drug Schedule Cardiotoxicity index Recommended max. dose (mg/m2)
Doxorubicin Rapid infusion 1 400
Doxorubicin Weekly 0.7 550
Doxorubicin 24-h infusion 0.62 550
Doxorubicin 48-h infusion 0.57 625
Doxorubicin 96-h infusion 0.5 800–1000
Epirubicin Rapid infusion 0.44 900
Mitoxantrone Rapid infusion 2.5 160
Daunorubicin Rapid infusion 0.5 800
Idarubicin Rapid infusion 2.67 150
Doxorubicin + dexrazoxane Rapid infusion 0.5 800–1000
Doxorubicin, 300 mg/m2 + dexrazoxane Rapid infusion 0.73 500
Table 7

Conventional risk factors of chemotherapy induced cardiotoxicity

jcu-26-1-i007
Female sex
Age (< 18 years old, > 75 years old)
Uncontrolled hypertension or diabetes mellitus
Renal failure
Previous history of cardiotoxicity
Pre-existing heart disease: LV hypertrophy, coronary artery disease
Cardiomyopathy: reduced LV ejection fraction
Concomitant or previous radiation therapy involving heart
Concomitant chemotherapy: anthracyclines/trastuzumab

LV: left ventricular

Table 8

Summary for management of cardiovascular toxicity

jcu-26-1-i008
Toxicity Managemen
Heart failure Avoid risk factors
Diuretics, BB, MRA, ACEIs or ARBs
CRT or ICD
Ischemic heart disease Nitrate or nitroglycerine
Antiplatelet agent, anticoagulation
BB, CCB, ACEIs or ARBs
Lipid-lowering agents
Coronary revascularization (intervention or surgery)
Hypertension Diuretics, DHP-CCB, BB, ACEIs or ARBs
Arrhythmia
 QT prolongation Avoid risk factors, ICD in cases of VT or VF
 Atrial fibrillation Rhythm control: cardioversion
Rate control: BB, digoxin, non-DHP-CCB
Acute pericarditis Aspirin, NSAID, colchicine
Pericardial effusion Pericardiocentesis, pericardiotomy
Pulmonary arterial hypertension Ambrisentan, iloprost
Venous thromboembolism Anticoagulation

BB: beta blocker, MRA: mineralocorticoid receptor antagonist, ACEIs: angiotensin converting enzyme inhibitors, ARBs: angiotensin receptor blockers, CRT: cardiac resynchronization therapy, ICD: implantable cardioverter defibrillator, CCB: calcium channel blocker, DHP: dihydropyridine, VT: ventricular tachycardia, VF: ventricular fibrillation, NSAID: nonsteroidal anti-inflammatory drug

TOOLS
Similar articles