Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Nov 14.
Published in final edited form as: J Am Coll Cardiol. 2017 Nov 14;70(20):2536–2551. doi: 10.1016/j.jacc.2017.09.1096

Cardiovascular Complications of Cancer Therapy: Best Practices in Diagnosis, Prevention, and Management—Part 1

Hui-Ming Chang a, Rohit Moudgil b, Tiziano Scarabelli c, Tochi M Okwuosa d, Edward TH Yeh a
PMCID: PMC5825187  NIHMSID: NIHMS913142  PMID: 29145954

Abstract

Modern cancer therapy has successfully cured many cancers and converted a terminal illness to chronic disease. Because cancer patients often have co-existing heart diseases, expert advice from the cardiologists will improve clinical outcome. In addition, cancer therapy can also cause myocardial damage, induce endothelial dysfunction, and alter cardiac conduction. Thus, it is important for practicing cardiologists to be knowledgeable about the diagnosis, prevention, and management of cardiovascular complications of cancer therapy. In this first part of a 2-part review, we will review cancer therapy-induced cardiomyopathy and ischemia. This review is based on MEDLINE literature search, published clinical guidelines, and best practices in major cancer centers. With the number of cancer survivors expanding quickly, the time has come for cardiologists to work closely with cancer specialists to prevent and treat cancer therapy-induced cardiovascular complications.

Keywords: Cardiovascular complication, cancer therapy, cardiomyopathy, ischemia


Heart diseases and cancer are the top two leading causes of death in the United States. Since these maladies share several common risk factors, many of our patients, especially the elderly, are afflicted by both cancer and heart diseases. Furthermore, cancer therapies, either radiation treatment or chemotherapy, can cause cardiovascular complications. Thus, it is important for practicing cardiologists to be familiar with the prevention, diagnosis, and management of cardiovascular complications of cancer therapy. This topic was reviewed in the Journal in 2009 (1). The purpose of this new state of the art review is to provide an update in this emerging discipline that abounds with exciting new developments. Cardiovascular complications covered in this two-part review are heart failure (HF), myocardial ischemia, myocarditis, hypertension (HTN), pulmonary hypertension, pericardial diseases, thromboembolism, QT prolongation and arrhythmias, and radiation-induced cardiovascular diseases. A MEDLINE search for each of these complications was performed; clinically relevant complications were selected based on experiences at the MD Anderson Cancer Center and centers affiliated with authors of this review. Diagnostic and treatment recommendations are based on the best practices developed at MD Anderson Cancer Center and recent guidelines (26).

HF

HF due to chemotherapy has been long recognized as a serious side-effect of daunorubicin, the first anthracycline used clinically (7). While the anthracycline class of chemotherapy agents remains the major cause of chemotherapy-induced cardiomyopathy (CIMP), newer cancer therapy, such as trastuzumab or proteasome inhibitors, can also cause cardiomyopathy (Table 1). It should also be recognized that patients can develop signs and symptoms of clinical heart failure during chemotherapy; however, the cause of cardiac decompensation may be due to fluid over-load, stress-induced cardiomyopathy, or primary cancer, but not chemotherapy (2). CIMP has been described in 1% to 5% of cancer survivors (8,9) and portends one of the worst survivals among cardiomyopathies (10). Early diagnosis and timely intervention has been shown to result in a superior clinical outcome in cancer patients treated with cardiotoxic chemotherapy (11).

Table 1.

Anticancer Agents Associated With HF/Left Ventricular Dysfunction

Chemotherapy Agents Frequency of Use Incidence (%) Prevention/Treatment
Anthracyclines Monitor EF, GLS, troponin
Dexrazoxane, continuous infusion, Liposomal preparation, BB/ACEI
 Doxorubicin (Adriamycin) ++++ 3–26
 Epirubicin (Ellence) + 0.9 – 3.3
 Idarubicin (Idamycin PFS) ++ 5–18
Alkylating agents
 Cyclophosphamide (Cytoxan) ++++ 7–28
 Ifosfamide (Ifex) +++ 17
Antimetabolites
 Decitabine (Dacogen) ++ 5
 Clofarabine (Clobar) + 27
Antimicrotubule agents
 Docetaxel (Taxotere) ++ 2.3–8
Monoclonal antibody-based tyrosine kinase inhibitors
 Trastuzumab (Herceptin) +++ 2–28 Avoid concomitant use with anthracyclines
 Bevacizumab (Avastin) ++ 1–10.9
 Adotrastuzumab emtansine (Kadcyla) + 1.8
 Pertuzumab (Perjeta) + 0.9–16
Small molecule tyrosine kinase inhibitors Treat hypertension aggressively
Ischemia work up and treatment
 Pazopanib (Votrient) ++++ 0.6 – 11
 Ponatinib (Iclusig) + 3–15
 Sorafenib (Nexavar) ++++ 1.9 – 11
 Dabrafenib (Tafinlar) ++++ 8–9
 Sunitinib (sutent) ++++ 1–27
 Dasatinib (Sprycel) ++++ 8–9
 Lapatinib (Tykerb) ++++ 0.9–4.9
 Trametanib (Mekinist) ++++ 7–11
Proteasome Inhibitor
 Carfilzomib (Kyprolis) ++ 7
 Bortezomib (Velcade) ++ 2–5
Miscellaneous
 Tretinoin (Vesanoid) ++++ 6

Frequency of Use: This was determined using inpatient and outpatient doses dispensed at MD Anderson Cancer Center during the time period of January 1, 2014 through December 21, 2014.

+

<1,000 doses dispensed

++

1,000–5,000 doses dispensed

+++

5,000–10,000 doses dispensed

++++

>10,000 doses dispensed

References: (1,2,24,29)

Definition

HF in the initial Von Hoff report was defined as the presence of tachycardia, shortness of breath, neck vein distention, gallop rhythms, ankle edema, hepatomegaly, cardiomegaly, and pleural effusion (12). With the advance of cardiac imaging, echocardiography or multi-gated acquisition (MUGA) radionuclide ventriculography-based evaluation of left ventricular ejection fraction (EF) has recently been included in the diagnostic criteria (4,13). In the trastuzumab clinical trials, drug-associated cardiotoxicity is defined as one or more of the following: 1) cardiomyopathy characterized by a decrease in EF globally or due to regional changes in interventricular septum contraction; 2) symptoms associated with HF; 3) signs associated with HF, such as S3 gallop, tachycardia, or both; 4) decline in initial EF of at least 5% to less than 55% with signs and symptoms of HF or asymptomatic decrease in EF of at least 10% to less than 55% (14). This definition does not include subclinical cardiovascular damage, such as diastolic dysfunction and changes in LV strain, which may occur earlier in response to some of the chemotherapeutic agents. Common Terminology Criteria for Adverse Events (CTCAE) has also defined cardiomyopathy and/or heart failure for the purposes of uniform reporting. In CTCAE 4.03, echocardiography and biomarkers were included to provide a more precise definition of cardiotoxicity.

Incidence and Pathogenesis

Anthracyclines

In a retrospective review of three trials, the incidence of doxorubicin-related HF was found to be 5% at a cumulative dose of 400 mg/m2, 16% at a dose of 500 mg/m2 and 26% at a dose of 550 mg/m2 (15). However, subclinical events occurred in about 30% of the patients, even at the doses of 180–240 mg/m2, about 13 years after the treatments (16). Interestingly, histopathologic changes, such as myofibrillar loss and vacuolization, can be seen in endomyocardial biopsy specimens from patients who have received as little as 240 mg/m2 of doxorubicin (17). These findings suggest that there is no safe dose of anthracycline. Even doses as low as 100 mg/m2 have been associated with reduced cardiac function (16,18). Nonetheless, some patients had no significant cardiac complications despite receiving doses as high as 1000 mg/m2 (19). Individual susceptibility is most likely due to genetic variants in genes that regulate anthracycline cardiotoxicity (20). Other risk factors for anthracycline toxicity include cumulative dose, intravenous bolus administration, higher single doses, history of prior irradiation, use of concomitant agents known to have cardiotoxicity, female gender, underlying CV disease, age (young and elderly), delayed diagnosis, and increase in cardiac biomarkers such as troponins during and after administration (9,2123).

Doxorubicin poisons topoisomerase 2 to cause DNA double strand break and demise of cancer cells. In the cardiomyocytes, doxorubicin targets topoisomerase 2β to induce DNA double-strand breaks and doxorubicin-bound topoisomerase 2β binds promoters of anti-oxidative and electron-transport genes to reduce their transcripts and protein expression (24,25) (Figure 1). Therefore, doxorubicin-treated cells have a marked increase in reactive oxygen species (ROS) and are defective in mitochondria biogenesis. Thus, topoisomerase 2β accounts for the three hallmarks of anthracycline-induced cardiotoxicity: myocyte death, ROS generation, and mitochondriopathy. Reduced topoisomerase 2β expression has been linked to a coding variant in the retinoic receptor γ gene, which predicts susceptibility to anthracycline-induced cardiotoxicity in childhood cancer (26).

Figure 1. Mechanism of Doxorubicin-induced Cardiotoxicity.

Figure 1

Doxorubicin inhibits topoisomerase 2β to induced DNA double strand break, leading to p53 activation and death of cardiomyocytes. Doxorubicin-bound topoisomerase 2β binds to promoters of anti-oxidative genes and PGC-1 that are required for expression of anti-oxidative enzymes and electron transport chains. Thus, topoisomerase 2β is able to account for the three hallmarks of doxorubicin-induced cardiotoxicity: cardiomyocyte death, generation of ROS, and mitochondriopathy (24,25).

Alkylating Agents

Alkylating agents add an alkyl group to the DNA of rapidly dividing cells and in the case of bifunctional alkylating agents, cross-linking the two DNA strands, thereby inhibiting DNA replication and cell proliferation (27). Symptoms may include arrhythmias and conduction disorders, and fulminant HF (28,29). Alkylating agents such as cyclophosphamide induce electrocardiogram (ECG) alterations in the form of low QRS voltage and non-specific T-wave or ST segment abnormalities (28,30,31). Acute symptoms usually occur within 1–2 weeks, last for a few days, and in some patients resolve without any late consequences (28,29,31). The incidence of high-dose cyclophosphamide-induced HF has been reported in as high as 28 % (32). Autopsy of patients with cyclophosphamide-induced cardiotoxicity shows hemorrhagic myocardial necrosis with interstitial edema and fibrin deposition. Complication is higher in elderly patients and in those exposed to anthracycline or mediastinal irradiation (1,29,31).

Ifosfamide is an alkylating nitrogen mustard used for the treatment of lymphomas, sarcomas, and testicular, breast, and lung carcinoma (33). There is a dose-dependent increase in HF associated with ifosfamide administration (33,34). Autopsy studies demonstrated increased heart weight, small pericardial effusions, subendocardial hemorrhage and petechial lesions in the epicardium (29).

Targeted therapies against HER-2 Pathway

Trastuzumab is a monoclonal antibody against the human epidermal growth factor receptor tyrosine kinase (HER2 or ErbB2), which regulates cell growth and repair (35). Overexpression of HER2 occurs in approximately 25% of breast cancers and confers increased proliferative and metastatic potential. HER-2 is expressed in cardiomyocytes and required for survival of cardiomyocytes (36). Mice deficient in HER-2 develop dilated cardiomyopathy and are more sensitive to doxorubicin (36).

Trastuzumab administration in HER2-positive breast cancers led to significant reductions in recurrence rate and overall mortality. A pivotal study in the metastatic setting demonstrated a 33% reduction in mortality at 1 year and an increase in median survival by 5 months (37). HF occurred in 8% of patients who received an anthracycline with cyclophosphamide; however, the incidence of HF increased to 27% with the addition of trastuzumab (37). Subsequently, several large trials confirmed efficacy of trastuzumab in increasing disease-free survival from cancer, but also established trastuzumab’s association with HF (38,39). In these trials, 1.7–4.1% of trastuzumab-treated patients developed HF when anthracycline was not part of the therapeutic regimen (38,39). Trastuzumab-related cardiotoxicity includes various degrees of LV systolic dysfunction, occasionally leading to HF (40). Symptoms are usually mild or moderate and improve following medical management and termination of drug administration (41,42). Improvement is usually seen in about four to six weeks after trastuzumab withdrawal (43). After symptomatic improvement, re-institution of trastuzumab treatment is usually possible (4143).

Other targeted therapies against HER-2 are relatively less cardiotoxic (4446). Perez et al (46) reviewed cardiac safety data in 44 clinical trials that used lapatinib. Of these study patients, only 1.6% experienced a cardiac event and of those only 0.2% was symptomatic. The rate of cardiac events in the lapatinib group was similar to those who did not receive lapatinib (44,46). Similarly, there was no significant increase in left ventricular dysfunction with the addition of pertuzumab to trastuzumab in the NeoSphere study (47,48). In the CLEOPATRA trial, the incidence of cardiac adverse events was 14.5% in the pertuzumab plus trastuzumab plus docetaxel arm compared to 16.4% in the control-placebo plus trastuzumab plus docetaxel arm (49,50). However, in a head-to-head comparison between lapatinib and trastuzumab, patients treated with lapatinib have shorter disease-free survival, but more non-cardiac toxicity, such as rash and diarrhea (51). Thus, one must consider both efficacy and toxicity in choosing drugs.

Vascular Endothelial Growth Factor Signaling Pathway Inhibitors

VEGF signaling pathway (VSP) inhibitors include antibody such as bevacizumab that binds to VEGF and small molecule tyrosine kinase inhibitors such as sunitinib and sorafenib, which inhibit the downstream kinase involved in VEGF receptor signaling (52). Cardiovascular side effects of VSP inhibitors include HTN, cardiomyopathy, conduction abnormalities, acute coronary syndromes, and arterial thromboses. Several VSP inhibitors also block receptors that are involved in compensatory response to stress in the cardiomyocytes. When the heart is unable to compensate for HTN-induced by VSP inhibitors, it could lead to HF (53). Therefore, maintaining good blood pressure control during VSP inhibitor therapy can prevent HF (54).

Bevacizumab is the first- or second-line chemotherapy for advanced solid tumors (5561). Its use has been associated with HTN, thromboembolism, and cardiomyopathy (62). Approximately 2%–4% of patients treated with bevacizumab will develop HF (29). Predisposing factors include previous therapy with cardiotoxic chemotherapy drugs, such as anthracyclines (63), capecitabine (64), as well as radiation to the mediastinum (29). In a meta-analysis of VSP inhibitors and its role in cardiovascular complications, the odds ratio for cardiac dysfunction caused by VSP inhibitors was 1.35(65). . More than half (40 out of 77) of the clinical trials included in the analysis utilized bevacizumab as the drug of treatment (65). Another meta-analysis showed that bevacizumab resulted in a significant decrease in EF with relative risk index of 3.4 (66).

In a large trial of sunitinib for the treatment of metastatic renal cell carcinoma, 13% developed LV dysfunction and 3% had severe HF (67). Severe HF was more common in a single-center study of 48 patients treated for metastatic renal cell carcinoma or gastrointestinal stromal tumor (GIST) (68). In a meta-analysis of 6935 patients treated with sunitinib, the incidence of HF was 4.1 % with a relative risk of 1.81 compared to placebo (69). In a retrospective analysis of a phase I/II trial of sunitinib for the treatment of GIST, there was a 2.5 fold increase in cardiotoxicity with higher doses (70). Sorafenib has been associated with a 4 % risk of mild LV dysfunction and 1 % risk of clinical HF (71). HF due to VSP inhibitors is reversible with cessation of therapy (72).

Proteasome Inhibitors

Proteasome is a protein complex present in all cells that degrades other proteins. Inhibition of proteasome blocks cell proliferation and induces apoptosis in tumor cells, especially in multiple myeloma. Bortezomib, a reversible proteasome inhibitor, is not known to cause HF (7375). However, 7% patients treated with carfilzomib developed new onset or worsening of preexisting HF or myocardial ischemia (76). In a phase II trial of 266 patients treated with carfilzomib monotherapy for relapsed myeloma, 10 experienced heart failure (3.8%), four (1.5%) had a cardiac arrest, and two (0.8%) had a MI during the study (77). The risk did not appear to be cumulative, at least through 12 cycles of therapy. Cardiotoxicity appears to be largely reversible with cessation of therapy and the initiation of HF treatment (78).

Screening, risk stratification and early detection strategies

Baseline Risk Assessment

Patients undergoing chemotherapy should have careful clinical evaluation and assessment of CV risk factors, such as coronary artery disease, diabetes, and hypertension (Table 2) (26). These risk factors should be managed according to the ACC/AHA guidelines (79). This is especially important if cancer therapy is known to cause HF. An aggressive HTN management is advised for patients treated with VSP inhibitors. Physical exercise has been shown to reduce cardiotoxicity in mouse models (80).

Table 2.

Best Practices for prevention, monitoring, and treatment of CIMP

Oncologist Cardiologist
Identify high-risk patients1 Modify cardiovascular risk factors2
Order pre-treatment Imaging3 Repeat Imaging studies4
Order biomarkers5
Consider non-cardiotoxic alternatives6 Advise on cardio-protection7
Adjust therapy protocols8 Institute cardio-protective medications9
Monitor during therapy10
Withhold cardiotoxic therapy only as the last resort11
Monitor after completion of therapy12
1

Pre-existing heart disease, diabetes, hyperlipidemia, young or old, female, plan for high-dose anthracycline therapy

2

Optimize cardiac medications, glucose control, diet, weight, exercise

3

If EF<50% or institution low normal, refer to cardiologist

4

Obtain high quality EF. Consider contrast, 3D, strain

5

Troponin and BNP

6

Consider non-cardiotoxic alternatives in high-risk patients

7

Interpret imaging and biomarker results and discuss with oncologist

8

For anthracycline: dose reduction, continuous infusion, liposomal doxorubicin, dexrazoxane; For anti-HER2: Avoid concomitant treatment with anthracyclines; For VSP inhibitors, treat HTN aggressively

9

Start BB or ACEI if EF<50% or EF drop >10% or abnormal GLS (>15% drop) or abnormal troponin

10

Monitor with echo at 3 month interval or symptom-driven; if cardio-protective medications were given, monitor at 1 month interval

11

For anthracycline EF<45%; for anti-HER2 therapy EF<40%

12

Obtain post-therapy EF; repeat echo in 6 months or a year; If EF remains abnormal, follow ACC/AHA HF guidelines

References: (25)

EF assessment is mandatory to establish a baseline cardiac function before cardiotoxic cancer treatment (3). Echocardiography is the preferred modality for assessment of cardiac structure and function. MUGA scan has less inter-observer variability; however, radiation exposure limits its utility as a cardiac monitor. Magnetic resonance imaging (MRI) can be used to obtain a precise EF; however, the use of MRI is limited by its cost. Cardiac biomarkers, such as the troponins and brain natriuretic peptides (BNP), can be used to monitor for the development of cardiac dysfunction (3,5,6). However, it is not known whether routine monitoring of biomarkers is useful in changing clinical outcomes. A number of composite scores have been designed to risk-stratify cancer patients (8183); however, they have not been validated in prospective studies.

Echocardiogram

Echocardiogram is the most important tool for serial evaluation of the heart during cancer therapy. EF should be determined using biplane method of discs according to American Society of Echocardiography guideline. If the endocardial border is not distinct, ultrasonic contrast should aid in endocardial border definition and subsequent volume calculations. However, temporal EF variability may be up to 10% with the confidence interval of 95% in 2D EF readings (84). 3D echocardiography has a temporal variability of 6% and is considered the most accurate EF measurement by echocardiography. Since CIMP is defined as a drop of EF of 10% or more or 5% or more in presence of heart failure symptoms, an accurate measurement of EF is paramount.

Myocardial Strain

Tissue Doppler imaging (TDI) and speckle-tracking strain imaging have emerged as two quantitative techniques for estimating global and regional myocardial mechanical function and have the potential to detect early signs of LV dysfunction (85). However, TDI is both user- and angle-dependent and is unable to differentiate translational motion or tethering effects from myocardial contractility. Speckle-tracking echocardiography (STE) is an angle-independent technique that uses an image-processing algorithm for analyzing motion of “speckles” or “fingerprints” within a 2D echo image, and it has replaced TDI strain as the preferred method for quantitative assessment of cardiac deformation (86,87).

Several studies have evaluated the utility of strain imaging for the detection of chemotherapy-associated cardiotoxicity. Fallah-Rad et al (88) evaluated 42 patients with breast cancer overexpressing HER-2 receiving trastuzumab in the adjuvant setting after anthracycline therapy. Within 3 months, peak global longitudinal and radial strain detected preclinical changes in LV systolic function before a decrease in EF was observed several months later. A recent prospective multicenter study by Sawaya et al demonstrated that global longitudinal strain (GLS) <19% was predictive of subsequent cardiotoxicity and was present in all patients who later developed symptoms of HF (89). Negishi et al similarly showed that a ≥11% relative reduction in GLS was predictive of subsequent trastuzumab-associated cardiotoxicity (90).

Abnormalities in strain parameters can also be seen several years after a cardiotoxic exposure. In 75 asymptomatic breast cancer survivors who received anthracycline with or without adjuvant trastuzumab, GLS was significantly decreased in the chemotherapy group 6 years after therapy compared with control subjects (91). In another meta-analysis, GLS consistently detected early myocardial changes during therapy (92). A 10% to 15% early reduction in GLS during therapy appears to be the most useful parameter for the prediction of cardiotoxicity. In late cancer survivors, global radial and circumferential strain are consistently abnormal, even with normal EF, but their clinical value in predicting subsequent ventricular dysfunction or HF has not been explored (92).

Biomarkers

The use of EF in the diagnosis of CIMP has important limitations. First, the measurement of EF is subject to technique-related variability, which can be higher than the thresholds used to define cardiotoxicity (14,84). Second, the reduction in EF is often a late phenomenon (11,88,93,94). Hence, there is a growing interest in identifying markers of early myocardial damages to predict the development of HF. Biomarkers are an economic and effective way of detecting myocardial dysfunction in apparently asymptomatic patients. The assessment of troponin and brain natriuretic peptide have been shown to be of incremental utility in identifying patients at increased risk for adverse outcomes (9597).

Troponins

Troponin is an important biomarker for acute coronary syndromes (ACS) and other myocardial damage (98100). In an early study, cTnI was elevated in 32% of 204 patients receiving high-dose chemotherapy and increase in cTnI occurred in more than 50% of patients soon after drug administration (101). A follow-up study showed that patients with negative cTnI (<0.08 ng/mL), immediately and 1 month after chemotherapy, showed no EF reduction and had low incidence of cardiac events (9). In contrast, patients with positive cTnI had a higher incidence of adverse cardiac events, including HF and asymptomatic LV dysfunction (9).

Serial troponin measurements in patients with hematologic malignancies treated with anthracycline showed troponin elevation correlated with EF reduction (102). A persistent release of cTnI was associated with a probability of major cardiac events within the first year of followup (9,22). In children treated with high-dose doxorubicin for acute lymphoblastic leukemia, cTnT increased in approximately 30% of cases, and the amount of elevation was predictive of cardiac dysfunction during follow-up (103,104). Thus, most studies showed a good correlation of elevated enzymes with LV dysfunction, especially in patients who were treated with high-dose anthracycline.

Dodos et al performed a series of cTnT measurements on the 3rd to 5th day following the first course of anthracycline and after the last course (105). They did not observe troponin elevation followed by EF deterioration. In their study, cTnT levels did not exceed the upper limit of the normal range in all patients. Only 7% of patients had low-level elevation of cTnT and only 1 of these patients developed decrease in EF. McArthur et al. studied a group of patients treated with bevacizumab, doxorubicin, and cyclophosphamide followed by paclitaxel in early-stage breast cancer (106). Seven patients (9%) experienced either a symptomatic or asymptomatic EF decline. There was no association between EF change and troponin elevation (106). These authors speculate that cTnI release could be missed because samples were drawn prior to chemotherapy (106). Thus, utility of using troponins in predicting EF changes depends on timings of blood drawn relative to chemotherapeutic administration.

In patients with breast cancer receiving anthracycline or trastuzumab, an elevation of high-sensitivity cTnI with a decrease in GLS of at least 19% is highly specific in predicting CIMP (89). Based on these data, an expert panel proposed that cTnI should be measured at baseline and every 3 weeks during trastuzumab therapy accompanied by echocardiography and GLS at baseline and every 3 months (3). A small study concluded that increase in high-sensitivity troponins is a good predictor of LV dysfunction (107). A high baseline level of high-sensitivity troponins is also a predictor of adverse outcomes (107).

Brain Natriuretic Peptide (BNP)

Natriuretic peptides are produced by splitting a prohormone into the amino-terminal inactive form and the carboxy-terminal biologically active hormone. The ventricle secretes biologically active brain natriuretic peptide (BNP) and inactive amino-terminal pro-BNP (NT-pro-BNP) in response to increased ventricular volume and pressure (108). Clinical studies have utilized BNP and NT-pro-BNP as biomarkers of CIMP, and while results are mixed, several studies have indicated that these peptides could be good early indicators of cardiac damage (108111)

In an early anthracycline study, increase in BNP level correlated with E/A ratio increase, suggesting that BNP level may be predictive of diastolic dysfunction. BNP increase during anthracycline treatment is usually transient and in most cases not predictive of clinical outcome. Only patients with persistent elevation of BNP developed overt HF, which also suggests a potential use of BNP in long-term follow-up (112). Nousiainen et al. found no significant correlations between echo parameters and natriuretic peptides until the cumulative doxorubicin dose reached 500 mg/m2 (113). Meinardi et al. showed that during chemotherapy, concentrations of BNP in plasma increase as EF decrease (114). Daugaard et al. also found that neither baseline levels of N-ANP nor BNP nor changes in these variables during therapy were predictive of a change in EF (115). However, persistent elevation of BNP may be an indication of adverse cardiac outcomes (114,115). C-reactive peptide (116118), myeloperoxidase (116), galacetin-3 (116,119,120) and ST-2 (120,121) have been investigated as potential biomakers; however, they cannot be recommended as routine tests at present.

Preventive strategies for anthracycline-induced cardiotoxicity

Selecting a non-anthracycline regimen

A randomized study of 3222 women with HER-positive early breast cancer found that a non-anthracycline containing regimen has equal efficacy and less cardiotoxicity (122). With a non-anthracycline regimen containing docetaxel, carboplatin, and trastuzumab (TCH), patients have a 5 year disease-free survival rate of 81% compared to 84% in a anthracycline-containing regimen (ACT or ACT-H). Importantly, the TCH regimen has much lower cardiotoxicity than ACT or ACT-H. Thus, TCH was proposed as an alternative non-anthracycline containing regimen for HER-positive early breast cancers (Table 2).

Substituting doxorubicin with less-cardiotoxic anthracyclines

Over the past five decades, more than 2000 modified anthracycline chemicals have been tested in attempt to reduce cardiotoxicity while retaining tumoricidal efficacy. While several anthracycline derivatives have been evaluated in clinical trials, only epirubicin (123) and idarubicin (124) received approval for clinical use. However, a critical analysis by Cochrane database identified no difference in cardiotoxicity between epirubicin and doxorubicin at equipotent doses (125).

Continuous Infusion

Replacing bolus administration with slow infusions does not significantly affect anthracycline area under the curve but diminishes anthracycline Cmax and anthracycline accumulation in the heart (126). A Cochrane review (127) showed a significantly lower rate of clinical HF with an infusion duration of 6 hours or longer as compared to a shorter infusion duration in the adults. In the pediatric populations, the results of infusion of anthracycline have been disappointing. A randomized trial in children with high-risk acute lymphocytic leukemia found that continuous infusion offered no additional cardiac protection over bolus administration in a median follow-up of 8 years post-diagnosis (128). A follow-up at 10 years also revealed no incremental therapeutic efficacy for infusion (129). Thus, continuous infusion cannot be recommended in the pediatric population (130,131).

PEGylated Liposomal doxorubicin

PEGylated liposomal doxorubicin comprises an aqueous core of doxorubicin hydrochloride encapsulated in liposomes with a protective hydrophilic outer coating of surface-bound methoxypolyethylene glycol (132,133). Delivery of doxorubicin in a PEGylated liposomal form decreases the circulating concentrations of free doxorubicin and results in selective uptake of the agent in tumor cells. In randomized trials, PEGylated liposomal doxorubicin was as effective as doxorubicin or other traditional combination chemotherapies (134136) (137139). Thus, PEGylated liposomal doxorubicin is a useful option in the treatment of various malignancies (140). However, the cost associated with administering this drug has prevented its widespread adoption (141).

Dexrazoxane

Dexrazoxane was originally developed as an anticancer agent (142). Using fibroblasts from topoisomerase2β knockout mice, Lyu et al showed that dexrazoxane is protective against anthracycline-induced toxicity in topoisomerase 2β-dependent manner, linking dexrazoxane to the topisomerase2β theory of anthracycline-cardiotoxicity (25,143) (Figure 1).

The protective effect of dexrazoxane against anthracycline-induced cardiotoxicity has been demonstrated in numerous clinical trials and in adults and children (19,104,144146). Dexrazoxane was approved in Europe and the United States for cardioprotection in patients treated with anthracyclines (Cardioxane and Zinecard) with several generic preparations available (Procard and Cardynax). In addition, dexrazoxane has been also approved for treatment of accidental extravasation of anthracyclines (Savene).

Unfortunately, one phase III trial suggested that dexrazoxane may lower the efficacy of anthracycline in treating breast cancer (19). In this trial, a significant difference in objective response was reported (47% vs. 61%, respectively, p = 0.019). While high response in the placebo group was quite unusual, other endpoints (including survival or time to progression) were not affected by dexrazoxane in this study (19,147). Careful meta-analyses of all available randomized clinical trials found no evidence that dexrazoxane lower doxorubicin’s anti-cancer effect (148,149). However, FDA approves dexrazoxane only in patients who have received more than 300 mg/m2 for metastatic breast cancer and who may benefit from continued doxorubicin treatment (150).

Another controversy about dexrazoxane pertains to an increased risk of second malignancies in the pediatric cancer survivors. This was observed in survivors of Hodgkin lymphoma who had received dexrazoxane in combination with doxorubicin and etoposide. It was postulated that combining these drugs could exceed a threshold above which topoisomerase inhibitors caused genetic instability in normal tissues (151). This report led the European Medicine Agency to disapprove the use of dexrazoxane in children. However, two studies of survivors of childhood acute lymphoblastic leukemia did not detect an increased risk of second malignancies from dexrazoxane (152,153). Thus, risk/benefit analysis supports a wider clinical usage of dexrazoxane with the possible exception in patients receiving etoposide or etoposide-anthracycline combinations.

Preventative Strategies against Trastuzumab-induced cardiotoxicity

The metastatic breast cancer trial showed that concurrent treatment of trastuzumab and anthracycline had detrimental cardiac outcomes (14,37). However, as anthracycline and trastuzumab were not administered at the same time, the incidence of HF drastically reduced. In the N9831 trial, the incidence of NYHA class III/IV HF or cardiac deaths was 0% in the control arm (anthracycline without trastuzumab) and 3.3% in the concurrent anthracycline/trastuzumab arm (154). In the B-31 study, these toxic effects occurred in 0.8% of the control group (anthracycline without trastuzumab) and 4.1% of the anthracycline/trastuzumab arm. HF was not reported in the FinHer trial, in which trastuzumab was omitted during the three cycles of epirubicin-containing regimen (155). These results were in marked distinction from 27% incidence of HF in the original metastatic trials.

Treatment

Symptomatic and asymptomatic HF should be treated according to ACC/AHA guidelines (Table 2) (156,157). We recommend HF treatment when subclinical cardiotoxicity was detected by strain imaging and biomarkers (2,3). Many cancer patients with overt HF or subclinical HF can be treated with ACEI or BB to allow completion of the chemotherapy. Anthracycline-induced cardiotoxicity was considered irreversible, whereas trastuzumab was reversible (43,158). Thus, different EF cutoff threshold for withholding therapy is recommended (Table 2) (2). It should be noted that cessation of cancer therapy should be considered only as the last resort. Every effort should be made to manage HF to allow chemotherapy to continue. Patients with symptomatic and/or overt heart failure should be treated according to ACC/AHA HF guidelines (156,157).

ISCHEMIA

Cancer treatment, including radiation therapy and chemotherapy, are associated with accelerated development of coronary artery disease (CAD) and/or acute coronary syndrome (ACS). Cancer itself can also create a pro-thrombotic state that promotes the development of ACS. Thus, chest pain in cancer patients needs to be investigated promptly. Drugs that commonly associated with ischemia in cancer patients were listed in Table 3.

Table 3.

Anticancer Agents Associated With Myocardial infarction/ischemia

Chemotherapy Agents Frequency of Use Incidence (%) Prevention/Treatment
Antimetabolites Ischemia workup and treatment
 Capecitabine (Xeloda) ++++ 3–9
 Flourouracil (Adrucil) ++++ 1–68
Monoclonal antibody-based tyrosine kinase inhibitors
 Bevacizumab (Avastin) +++ 0.6 – 8.5
Small molecule tyrosine kinase inhibitors
 Nilotinib (Tasigna) ++++ 5–9.4
 Ponatinib (Iclusig) + 12
Angiogenesis Inhibitors
 Lenalidomide (Revlimid) +++ 0–1.9
Antimicrotubule agents
 Paclitaxel (Taxol) ++++ <1.5

Frequency of Use: This was determined using inpatient and outpatient doses dispensed at MD Anderson Cancer Center during the time period of January 1, 2014 through December 21, 2014.

+

<1,000 doses dispensed

++

1,000–5,000 doses dispensed

+++

5,000–10,000 doses dispensed

++++

>10,000 doses dispensed

References: (1,2,24,29)

Incidence

Antimetabolites

Chest pain is the most common symptom associated with 5-fluoracil (5-FU) administration. Less common side effects include myocardial infarction (MI), arrhythmias, HF, cardiogenic shock, and sudden death (159,160). High doses 5-FU (>800 mg/m2) and continuous 5-FU infusions are recognized risk factors of cardiotoxicity (7.6%) as compared to bolus injections (2%) (159,161,162). Other risks factors include preexisting cardiovascular disease, prior mediastinal radiation, and concurrent use of other chemotherapeutic agents (160,161,163169). Cardiac events are typically short-lasting (up to 48 hours) and tend to manifest within 2 to 5 days after initiation of 5-FU (159). Ischemic alterations of the ECG have been reported in 68% of patients and associate with biomarkers elevations in 43% of the cases (162). The overall mortality ranges from 2.2% to 13% (161,165,170).

Selective activation of capecitabine (oral prodrug of 5-FU), which occurs preferentially in cancer cells, explains its lower cardiotoxicity compared to 5-FU. The incidence of capecitabine-induced cardiotoxicity ranged between 3 and 9% (160,171173). One prospective study enrolling 644 patients with no prior history of coronary artery disease reported the occurrence of symptomatic ischemic changes of the ECG in 5.2% of patients receiving capecitabine (166). ECG changes, including ST-segment elevation, were seldom associated with elevation of serum cardiac markers (162,164,174,175).

Anti-microtubule agents

Myocardial ischemia and infarction have been reported in approximately 3% of the patients receiving paclitaxel. In 198 patients treated with paclitaxel for ovarian cancer, 0.5% experienced a MI (176). In the Cancer Therapy Evaluation Program’s Adverse Drug Reaction database, which followed more than 3400 patients, the overall incidence of grade 4 and 5 cardiac events was only 0.29% (176). These events occurred within two weeks from paclitaxel administration (176) and were observed in patients with known cardiac risk factors including HTN and CAD.

Cardiac ischemia has also been reported after administration of docetaxel. In a clinical trial with inoperable, locally advanced squamous cell carcinoma of the head and neck, 355 patients were randomized to receive a standard regimen of cisplatin and 5-FU or the same regimen plus docetaxel. Myocardial ischemia occurred in 1.7% of patients from the docetaxel arm and in 0.6% of patients from the control arm (177). Although not reported in the original study, cardiac ischemia was listed as a side effect of docetaxel the package insert (178).

Antibody-based VEGF inhibitor

In a pooled analysis of 1,745 cancer patients from 5 randomized controlled trials, the overall incidence of arterial thrombotic events (ATEs) was significantly higher in the bevacizumab group (3.8%) as compared to the control group (179). In the Bevacizumab Regimens’ Investigation of Treatment Effects (BRiTE) study, 1,953 patients receiving bevacizumab at 248 U.S. sites were followed for over 20 months (180). The overall incidence of ATEs was 2%. Of these events, >57% were cerebrovascular cardiovascular accidents (CVAs) and transient ischemic attacks (TIAs), while 30% were MI (50% lethal) (180). Independent risk factors for ATEs included age ≥75 years, poor performance status (ECOG performance status score ≥1), preexisting HTN, anticoagulation therapy, and arterial disease (180).

Small molecular tyrosine kinase inhibitors

In a double-blind, international phase III trial enrolling 569 patients with un-resectable, locally advanced, or metastatic pancreatic cancer, CVAs and MI occurred in 4.6% patients randomized to receive gemcitabine plus erlotinib compared with 1.2% patients assigned to gemcitabine plus placebo (181). Although the incidence of CVA/MI was not published in the original study by Moore et al., it was reported by the FDA in the erlotinib package insert (182).

Approximately 3% patients in clinical trials have experienced myocardial ischemia with sorafenib. In an unpublished clinical trial, MI/ischemia occurred among 2.7% of hepatocellular cancer patients treated with sorafenib compared with 1.3% of patients in the placebo group (183). Similarly, sorafenib was associated with a higher incidence of MI/ischemia compared with placebo in patients treated for renal cell carcinoma (3% vs. <1%) (184). The incidence of cardiac ischemia/MI in patients with different types of cancer receiving sorafenib ranges from 2 to 3% (183).

Proteasome inhibitors

The safety profile of carfilzomib was analyzed by Siegel et al. in 526 patients from 4 phase II clinical trials, based on which the use of carfilzomib was approved by the U.S. FDA (185). In this analysis, the overall incidence of myocardial ischemia was reported to be 3.4%. Significantly higher occurrence of ischemic events was reported in two subsequent phase III, randomized, multicenter trials, the ASPIRE and ENDEAVOR studies. In the ASPIRE study, the rate of ischemic heart disease was 5.9% versus 4.6% in the control group (186). Likewise, in the ENDEAVOR study, ischemic events occurred at a rate of 3% as compared to 2% in the control group (187). Hence, high-risk patients should be considered for an ischemic work-up prior to starting carfilzomib treatment (187).

Pathophysiology

Persistent coronary spasm occurring at the level of a preexisting plaque was observed during cardiac catheterization in a patient receiving continuous 5-FU infusion (188). Experimental work in rabbit aortic rings showed that incremental doses of 5-FU induced endothelium-independent vasoconstriction, secondary to protein kinase C-mediated vasoconstriction of vascular smooth muscle (189). An alternative mechanism of cardiotoxicity postulates a direct toxic effect of 5-FU on the coronary endothelium (190192), with ensuing endothelial injury leading to micro-thrombotic occlusions (190). Although undetectable by coronary angiography, endothelial injury and small vessel thrombosis could be key ultrastructural findings (191,192). 5-FU-induced endovascular injury could be reduced by use of anticoagulants (193,194). Little is known regarding the cardiotoxic effect of paclitaxel. Myocardial ischemia in association with paclitaxel is thought to be due to concurrent use of other drugs and preexisting cardiac conditions (195). The polyoxyethylated castor oil, known as Cremophor EL, which is used as a vehicle for paclitaxel in the injectable formulation may contribute to the overall cardiotoxicity, by inducing histamine release (195).

VEGF stimulates endothelial cell proliferation to maintain endothelial viability and vascular integrity (196). Consequently, bevacizumab administration may impair the regenerative potential of endothelial cells in response to stress. Exposed sub-endothelial collagen can trigger tissue factor activation resulting in thromboembolism (196,197). VEGF inhibition also impairs the production of nitric oxide and prostacyclin, as well as increases hematocrit and blood viscosity via overproduction of erythropoietin, all of which heighten the thromboembolic risk (196). Carfilzomib was found to increase coronary perfusion pressure and resting vasoconstriction tone in isolated rabbit heart and aorta (198200). In addition, carfilzomib amplified the spasmogenic effect of noradrenaline and angiotensin II, curbed the anti-spasmogenic activity of nifedipine and nitroglycerin, and reduced the vasodilating effect of acetylcholine.

Screening, diagnosis, and treatment

Since pre-existing CAD is a known risk factor for the development of chemotherapy-induced ACS, ischemic work up should be initiated in all high-risk patients before administration of drugs known to cause cardiac ischemia (Table 3). Patients with suspected ACS should be treated according to ACC/AHA guidelines (201,202). Besides statin and beta blockers, the cornerstones ACS treatment include percutaneous coronary intervention, as well as antiplatelet and anticoagulant therapy, all of which pose an incremental bleeding risk in cancer patients with thrombocytopenia. Although prospective studies in this specific population are currently lacking, a retrospective analysis carried out in cancer patients with thrombocytopenia and ACS showed that aspirin improved 7-day survival rate without increasing the bleeding risk (203). Although case-specific considerations are warranted, life-saving interventions should not be denied to cancer patients with ongoing ACS because of thrombocytopenia (204). The response to anticoagulants and antiplatelet agents in patients with platelet counts >50,000/L seems to be comparable to that observed in patients with normal platelet counts (204). However, reduced heparin doses, ranging from 30 to 50 units/kg, may be required for patients whose platelet counts is <50,000/µL (204). Dual anti-platelet therapy with aspirin and clopidogrel can be used for patients with platelet counts >30,000/µL, while aspirin as a single agent should be given to those with platelet counts >10,000/µL. With platelet count below 10,000/µL, bleeding risk against the risk of leaving the thrombotic event untreated should be carefully evaluated (203). In cancer patients with ACS and thrombocytopenia, revascularization can still proceed with radial access, micro-puncture kits, and closure devices for the artery entry site. When the femoral access is chosen, prolonged groin pressure of at least 30 minutes should be instituted to obtain hemostasis (205).

Patients treated with 5-FU or capecitabine should be closely monitored for myocardial ischemia with serial ECGs. Preemptive use of coronary vasodilators, such as nitrates and calcium-channel blockers, should be considered. In cancer patients who develop acute chest pain while receiving 5-FU or capecitabine, offending drugs should be withheld until diagnostic workup is completed and anti-anginal therapy is instituted. It is possible to re-challenge the patients with close monitoring; however, an alternative regimen that does not contain the offending drug is a better option (159,163,165,172).

Temporary or permanent discontinuation of sorafenib is also advised in the management of patients developing cardiac ischemia during or following treatment (183). There is scarcity of data about re-challenge. Since patients who suffered a stroke or MI within 12 months from enrollment have been excluded from the trials evaluating bevacizumab, the safety of the drug in this high-risk population is unknown (206). Treatment with bevacizumab should be promptly discontinued in patients who develop severe ATEs during treatment. The safety of restarting bevacizumab after resolution of an ATE has not been evaluated (206). As cardiac complications caused by carfilzomib are serious, high-risk patients, including those ≥75 years, should undergo an ischemic work-up prior to starting carfilzomib treatment (203). Prompt discontinuation of carfilzomib is warranted when chest pain developed during infusion.

Central Illustration. Management of cancer therapy-induced cardiovascular complications.

Central Illustration

Best practices in the management of cancer therapy-induced cardiomyopathy and ischemia. EF=ejection fraction; GLS=global longitudinal strain; VSP=VEGF signaling pathway; BB=beta blocker; ACEI= angiotensin converting enzyme inhibitor; CAD=coronary artery disease; DAT=dual anti-platelet therapy; DES=drug-eluting stent.

Condensed Abstract.

The anthracycline class of chemotherapy drugs is known to cause cardiotoxicity in a dose-dependent manner. Recent understanding of the molecular mechanism of anthracycline cardiotoxicity suggests an approach to prevent this dreaded complication. Trastuzumab when used in conjunction with anthracycline usually resulted in significant cardiotoxicity, which can be prevented by avoiding concurrent use of these drugs. 5-FU and VSP inhibitors can cause cardiac ischemia, especially in patients with pre-existing CAD. Ischemia workup is indicated before therapy in high-risk patients. Life-saving interventions for ACS should not be denied to cancer patients with chemotherapy-induced thrombocytopenia.

Acknowledgments

Funding/Support: National Institutes of Health (HL126916) to ETHY. ETHY is the Frances T. McAndrew Chair in the University of Missouri, School of Medicine.

Abbreviation

ACEI

Angiotensin-Converting Enzyme Inhibitor

ACS

Acute Coronary Syndrome

ATE

Arterial Thrombotic Event

BB

Beta Blocker

CAD

Coronary Artery Disease

CIMP

Chemotherapy-induced Cardiomyopathy

CTCAE

Common Terminology Criteria for Adverse Events

CVA

Cerebral Vascular Accident

ECG

Electrocardiogram

EF

Ejection Fraction

FDA

Food and Drug Administration

GLS

Global Longitudinal Strain

HF

Heart Failure

5-FU

5-flurouracil

HTN

Hypertension

MI

Myocardial Infarction

MUGA

Multi-gated Acquisition Radionuclide Ventriculography

ROS

Reactive Oxygen Species

STE

Speckle Tracking Echocardiography

TDI

Tissue Doppler Imaging

TIA

Transient Ischemic Attack

Tn

Troponin

VEGF

Vascular Endothelial Growth Factor

VSP

VEGF Signaling Pathway

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Disclosures: No conflict of interest disclosures for all authors.

References

  • 1.Yeh ET, Bickford CL. Cardiovascular complications of cancer therapy: incidence, pathogenesis, diagnosis, and management. J Am Coll Cardiol. 2009;53:2231–47. doi: 10.1016/j.jacc.2009.02.050. [DOI] [PubMed] [Google Scholar]
  • 2.Yeh ET. MD Anderson Practices in Onco-Cardiology. 2016 [Google Scholar]
  • 3.Plana JC, Galderisi M, Barac A, et al. Expert consensus for multimodality imaging evaluation of adult patients during and after cancer therapy: a report from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. Eur Heart J Cardiovasc Imaging. 2014;15:1063–93. doi: 10.1093/ehjci/jeu192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Zamorano JL, Lancellotti P, Rodriguez Munoz D, et al. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines: The Task Force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC) Eur Heart J. 2016;37:2768–2801. doi: 10.1093/eurheartj/ehw211. [DOI] [PubMed] [Google Scholar]
  • 5.Armenian SH, Lacchetti C, Barac A, et al. Prevention and Monitoring of Cardiac Dysfunction in Survivors of Adult Cancers: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2017;35:893–911. doi: 10.1200/JCO.2016.70.5400. [DOI] [PubMed] [Google Scholar]
  • 6.Curigliano G, Cardinale D, Suter T, et al. Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO Clinical Practice Guidelines. Ann Oncol. 2012;23(Suppl 7):vii155–66. doi: 10.1093/annonc/mds293. [DOI] [PubMed] [Google Scholar]
  • 7.Tan C, Tasaka H, Yu KP, Murphy ML, Karnofsky DA. Daunomycin, an antitumor antibiotic, in the treatment of neoplastic disease. Clinical evaluation with special reference to childhood leukemia. Cancer. 1967;20:333–353. doi: 10.1002/1097-0142(1967)20:3<333::aid-cncr2820200302>3.0.co;2-k. [DOI] [PubMed] [Google Scholar]
  • 8.Hequet O, Le QH, Moullet I, et al. Subclinical late cardiomyopathy after doxorubicin therapy for lymphoma in adults. J Clin Oncol. 2004;22:1864–71. doi: 10.1200/JCO.2004.06.033. [DOI] [PubMed] [Google Scholar]
  • 9.Cardinale D, Sandri MT, Colombo A, et al. Prognostic value of troponin I in cardiac risk stratification of cancer patients undergoing high-dose chemotherapy. Circulation. 2004;109:2749–54. doi: 10.1161/01.CIR.0000130926.51766.CC. [DOI] [PubMed] [Google Scholar]
  • 10.Felker GM, Thompson RE, Hare JM, et al. Underlying causes and long-term survival in patients with initially unexplained cardiomyopathy. N Engl J Med. 2000;342:1077–84. doi: 10.1056/NEJM200004133421502. [DOI] [PubMed] [Google Scholar]
  • 11.Cardinale D, Colombo A, Lamantia G, et al. Anthracycline-induced cardiomyopathy: clinical relevance and response to pharmacologic therapy. J Am Coll Cardiol. 2010;55:213–20. doi: 10.1016/j.jacc.2009.03.095. [DOI] [PubMed] [Google Scholar]
  • 12.Von Hoff DD, Layard MW, Basa P, et al. Risk factors for doxorubicin-induced congestiveheart failure. Annals of internal medicine. 1979;91:710–7. doi: 10.7326/0003-4819-91-5-710. [DOI] [PubMed] [Google Scholar]
  • 13.Virani SA, Dent S, Brezden-Masley C, et al. Canadian Cardiovascular Society Guidelines for Evaluation and Management of Cardiovascular Complications of Cancer Therapy. Can J Cardiol. 2016;32:831–41. doi: 10.1016/j.cjca.2016.02.078. [DOI] [PubMed] [Google Scholar]
  • 14.Seidman A, Hudis C, Pierri MK, et al. Cardiac dysfunction in the trastuzumab clinical trials experience. J Clin Oncol. 2002;20:1215–21. doi: 10.1200/JCO.2002.20.5.1215. [DOI] [PubMed] [Google Scholar]
  • 15.Swain SM, Whaley FS, Ewer MS. Congestive heart failure in patients treated with doxorubicin: a retrospective analysis of three trials. Cancer. 2003;97:2869–79. doi: 10.1002/cncr.11407. [DOI] [PubMed] [Google Scholar]
  • 16.Vandecruys E, Mondelaers V, De Wolf D, Benoit Y, Suys B. Late cardiotoxicity after low dose of anthracycline therapy for acute lymphoblastic leukemia in childhood. Journal of cancer survivorship : research and practice. 2012;6:95–101. doi: 10.1007/s11764-011-0186-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Nysom K, Holm K, Lipsitz SR, et al. Relationship between cumulative anthracycline dose and late cardiotoxicity in childhood acute lymphoblastic leukemia. J Clin Oncol. 1998;16:545–50. doi: 10.1200/JCO.1998.16.2.545. [DOI] [PubMed] [Google Scholar]
  • 18.van der Pal HJ, van Dalen EC, Hauptmann M, et al. Cardiac function in 5-year survivors of childhood cancer: a long-term follow-up study. Archives of internal medicine. 2010;170:1247–55. doi: 10.1001/archinternmed.2010.233. [DOI] [PubMed] [Google Scholar]
  • 19.Swain SM, Whaley FS, Gerber MC, et al. Cardioprotection with dexrazoxane for doxorubicin-containing therapy in advanced breast cancer. J Clin Oncol. 1997;15:1318–32. doi: 10.1200/JCO.1997.15.4.1318. [DOI] [PubMed] [Google Scholar]
  • 20.Aminkeng F, Ross CJ, Rassekh SR, et al. Recommendations for genetic testing to reduce the incidence of anthracycline-induced cardiotoxicity. Br J Clin Pharmacol. 2016;82:683–95. doi: 10.1111/bcp.13008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.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;9:1215–23. doi: 10.1200/JCO.1991.9.7.1215. [DOI] [PubMed] [Google Scholar]
  • 22.Cardinale D, Sandri MT, Martinoni A, et al. Myocardial injury revealed by plasma troponin I in breast cancer treated with high-dose chemotherapy. Ann Oncol. 2002;13:710–5. doi: 10.1093/annonc/mdf170. [DOI] [PubMed] [Google Scholar]
  • 23.Pichon MF, Cvitkovic F, Hacene K, et al. Drug-induced cardiotoxicity studied by longitudinal B-type natriuretic peptide assays and radionuclide ventriculography. In Vivo. 2005;19:567–76. [PubMed] [Google Scholar]
  • 24.Yeh ET, Chang HM. Oncocardiology-Past, Present, and Future: A Review. JAMA Cardiol. 2016;1:1066–1072. doi: 10.1001/jamacardio.2016.2132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zhang S, Liu X, Bawa-Khalfe T, et al. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med. 2012;18:1639–42. doi: 10.1038/nm.2919. [DOI] [PubMed] [Google Scholar]
  • 26.Aminkeng F, Bhavsar AP, Visscher H, et al. A coding variant in RARG confers susceptibility to anthracycline-induced cardiotoxicity in childhood cancer. Nat Genet. 2015;47:1079–84. doi: 10.1038/ng.3374. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Fu D, Calvo JA, Samson LD. Balancing repair and tolerance of DNA damage caused by alkylating agents. Nat Rev Cancer. 2012;12:104–20. doi: 10.1038/nrc3185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Floyd JD, Nguyen DT, Lobins RL, Bashir Q, Doll DC, Perry MC. Cardiotoxicity of cancer therapy. J Clin Oncol. 2005;23:7685–96. doi: 10.1200/JCO.2005.08.789. [DOI] [PubMed] [Google Scholar]
  • 29.Yeh ET, Tong AT, Lenihan DJ, et al. Cardiovascular complications of cancer therapy: diagnosis, pathogenesis, and management. Circulation. 2004;109:3122–3131. doi: 10.1161/01.CIR.0000133187.74800.B9. [DOI] [PubMed] [Google Scholar]
  • 30.Schimmel KJ, Richel DJ, van den Brink RB, Guchelaar HJ. Cardiotoxicity of cytotoxic drugs. Cancer Treat Rev. 2004;30:181–91. doi: 10.1016/j.ctrv.2003.07.003. [DOI] [PubMed] [Google Scholar]
  • 31.Slordal L, Spigset O. Heart failure induced by non-cardiac drugs. Drug Saf. 2006;29:567–586. doi: 10.2165/00002018-200629070-00003. [DOI] [PubMed] [Google Scholar]
  • 32.Gottdiener JS, Appelbaum FR, Ferrans VJ, Deisseroth A, Ziegler J. Cardiotoxicity associated with high-dose cyclophosphamide therapy. Archives of internal medicine. 1981;141:758–63. [PubMed] [Google Scholar]
  • 33.Quezado ZM, Wilson WH, Cunnion RE, et al. High-dose ifosfamide is associated with severe, reversible cardiac dysfunction. Annals of internal medicine. 1993;118:31–6. doi: 10.7326/0003-4819-118-1-199301010-00006. [DOI] [PubMed] [Google Scholar]
  • 34.Kandylis K, Vassilomanolakis M, Tsoussis S, Efremidis AP. Ifosfamide cardiotoxicity in humans. Cancer Chemother Pharmacol. 1989;24:395–6. doi: 10.1007/BF00257451. [DOI] [PubMed] [Google Scholar]
  • 35.Yarden Y. The EGFR family and its ligands in human cancer. signalling mechanisms and therapeutic opportunities. Eur J Cancer. 2001;37(Suppl 4):S3–8. doi: 10.1016/s0959-8049(01)00230-1. [DOI] [PubMed] [Google Scholar]
  • 36.Crone SA, Zhao YY, Fan L, et al. ErbB2 is essential in the prevention of dilated cardiomyopathy. Nat Med. 2002;8:459–65. doi: 10.1038/nm0502-459. [DOI] [PubMed] [Google Scholar]
  • 37.Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–92. doi: 10.1056/NEJM200103153441101. [DOI] [PubMed] [Google Scholar]
  • 38.Bird BR, Swain SM. Cardiac toxicity in breast cancer survivors: review of potential cardiac problems. Clin Cancer Res. 2008;14:14–24. doi: 10.1158/1078-0432.CCR-07-1033. [DOI] [PubMed] [Google Scholar]
  • 39.Hudis CA. Trastuzumab–mechanism of action and use in clinical practice. N Engl J Med. 2007;357:39–51. doi: 10.1056/NEJMra043186. [DOI] [PubMed] [Google Scholar]
  • 40.Yavas O, Yazici M, Eren O, Oyan B. The acute effect of trastuzumab infusion on ECG parameters in metastatic breast cancer patients. Swiss Med Wkly. 2007;137:556–8. doi: 10.4414/smw.2007.11899. [DOI] [PubMed] [Google Scholar]
  • 41.Keefe DL. Trastuzumab-associated cardiotoxicity. Cancer. 2002;95:1592–600. doi: 10.1002/cncr.10854. [DOI] [PubMed] [Google Scholar]
  • 42.Perez EA, Rodeheffer R. Clinical cardiac tolerability of trastuzumab. J Clin Oncol. 2004;22:322–9. doi: 10.1200/JCO.2004.01.120. [DOI] [PubMed] [Google Scholar]
  • 43.Ewer MS, Vooletich MT, Durand JB, et al. Reversibility of trastuzumab-related cardiotoxicity: new insights based on clinical course and response to medical treatment. J Clin Oncol. 2005;23:7820–6. doi: 10.1200/JCO.2005.13.300. [DOI] [PubMed] [Google Scholar]
  • 44.Azim H, Azim HA, Jr, Escudier B. Trastuzumab versus lapatinib: the cardiac side of the story. Cancer Treat Rev. 2009;35:633–8. doi: 10.1016/j.ctrv.2009.06.007. [DOI] [PubMed] [Google Scholar]
  • 45.Lenihan D, Suter T, Brammer M, Neate C, Ross G, Baselga J. Pooled analysis of cardiac safety in patients with cancer treated with pertuzumab. Ann Oncol. 2012;23:791–800. doi: 10.1093/annonc/mdr294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Perez EA, Koehler M, Byrne J, Preston AJ, Rappold E, Ewer MS. Cardiac safety of lapatinib: pooled analysis of 3689 patients enrolled in clinical trials. Mayo Clin Proc. 2008;83:679–86. doi: 10.4065/83.6.679. [DOI] [PubMed] [Google Scholar]
  • 47.Baselga J, Gelmon KA, Verma S, et al. Phase II trial of pertuzumab and trastuzumab in patients with human epidermal growth factor receptor 2-positive metastatic breast cancer that progressed during prior trastuzumab therapy. J Clin Oncol. 2010;28:1138–44. doi: 10.1200/JCO.2009.24.2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Gianni L, Pienkowski T, Im YH, et al. Efficacy and safety of neoadjuvant pertuzumab and trastuzumab in women with locally advanced, inflammatory, or early HER2-positive breast cancer (NeoSphere): a randomised multicentre, open-label, phase 2 trial. Lancet Oncol. 2012;13:25–32. doi: 10.1016/S1470-2045(11)70336-9. [DOI] [PubMed] [Google Scholar]
  • 49.Baselga J, Cortes J, Kim SB, et al. Pertuzumab plus trastuzumab plus docetaxel for metastatic breast cancer. N Engl J Med. 2012;366:109–19. doi: 10.1056/NEJMoa1113216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Swain SM, Ewer MS, Cortes J, et al. Cardiac tolerability of pertuzumab plus trastuzumab plus docetaxel in patients with HER2-positive metastatic breast cancer in CLEOPATRA: a randomized, double-blind, placebo-controlled phase III study. Oncologist. 2013;18:257–64. doi: 10.1634/theoncologist.2012-0448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gelmon KA, Boyle FM, Kaufman B, et al. Lapatinib or Trastuzumab Plus Taxane Therapy for Human Epidermal Growth Factor Receptor 2-Positive Advanced Breast Cancer: Final Results of NCIC CTG MA.31. J Clin Uncool. 2015;33:1574–83. doi: 10.1200/JCO.2014.56.9590. [DOI] [PubMed] [Google Scholar]
  • 52.Moslehi JJ. Cardiovascular Toxic Effects of Targeted Cancer Therapies. N Engl J Med. 2016;375:1457–1467. doi: 10.1056/NEJMra1100265. [DOI] [PubMed] [Google Scholar]
  • 53.Chintalgattu V, Ai D, Langley RR, et al. Cardiomyocyte PDGFR-beta signaling is an essential component of the mouse cardiac response to load-induced stress. J Clin Invest. 2010;120:472–84. doi: 10.1172/JCI39434. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Chen MH, Kerkela R, Force T. Mechanisms of cardiac dysfunction associated with tyrosine kinase inhibitor cancer therapeutics. Circulation. 2008;118:84–95. doi: 10.1161/CIRCULATIONAHA.108.776831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Aghajanian C, Blank SV, Goff BA, et al. OCEANS: a randomized, double-blind, placebo-controlled phase III trial of chemotherapy with or without bevacizumab in patients with platinum-sensitive recurrent epithelial ovarian, primary peritoneal, or fallopian tube cancer. J Clin Oncol. 2012;30:2039–45. doi: 10.1200/JCO.2012.42.0505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Friedman HS, Prados MD, Wen PY, et al. Bevacizumab alone and in combination with irinotecan in recurrent glioblastoma. J Clin Oncol. 2009;27:4733–40. doi: 10.1200/JCO.2008.19.8721. [DOI] [PubMed] [Google Scholar]
  • 57.Giantonio BJ, Catalano PJ, Meropol NJ, et al. Bevacizumab in combination with oxaliplatin, fluorouracil, and leucovorin (FOLFOX4) for previously treated metastatic colorectal cancer: results from the Eastern Cooperative Oncology Group Study E3200. J Clin Oncol. 2007;25:1539–44. doi: 10.1200/JCO.2006.09.6305. [DOI] [PubMed] [Google Scholar]
  • 58.Hurwitz H, Fehrenbacher L, Novotny W, et al. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med. 2004;350:2335–42. doi: 10.1056/NEJMoa032691. [DOI] [PubMed] [Google Scholar]
  • 59.Miller K, Wang M, Gralow J, et al. Paclitaxel plus bevacizumab versus paclitaxel alone for metastatic breast cancer. N Engl J Med. 2007;357:2666–76. doi: 10.1056/NEJMoa072113. [DOI] [PubMed] [Google Scholar]
  • 60.Rini BI, Halabi S, Rosenberg JE, et al. Phase III trial of bevacizumab plus interferon alfa versus interferon alfa monotherapy in patients with metastatic renal cell carcinoma: final results of CALGB 90206. J Clin Oncol. 2010;28:2137–43. doi: 10.1200/JCO.2009.26.5561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Tewari KS, Sill MW, Long HJ, 3rd, et al. Improved survival with bevacizumab in advanced cervical cancer. N Engl J Med. 2014;370:734–43. doi: 10.1056/NEJMoa1309748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Galfrascoli E, Piva S, Cinquini M, et al. Risk/benefit profile of bevacizumab in metastatic colon cancer: a systematic review and meta-analysis. Dig Liver Dis. 2011;43:286–94. doi: 10.1016/j.dld.2010.10.010. [DOI] [PubMed] [Google Scholar]
  • 63.D’Adamo DR, Anderson SE, Albritton K, et al. Phase II study of doxorubicin and bevacizumab for patients with metastatic soft-tissue sarcomas. J Clin Oncol. 2005;23:7135–42. doi: 10.1200/JCO.2005.16.139. [DOI] [PubMed] [Google Scholar]
  • 64.Miller KD, Chap LI, Holmes FA, et al. Randomized phase III trial of capecitabine compared with bevacizumab plus capecitabine in patients with previously treated metastatic breast cancer. J Clin Oncol. 2005;23:792–9. doi: 10.1200/JCO.2005.05.098. [DOI] [PubMed] [Google Scholar]
  • 65.Abdel-Qadir H, Ethier JL, Lee DS, Thavendiranathan P, Amir E. Cardiovascular toxicity of angiogenesis inhibitors in treatment of malignancy: A systematic review and meta-analysis. Cancer Treat Rev. 2017;53:120–127. doi: 10.1016/j.ctrv.2016.12.002. [DOI] [PubMed] [Google Scholar]
  • 66.Escalante CP, Chang YC, Liao K, et al. Meta-analysis of cardiovascular toxicity risks in cancer patients on selected targeted agents. Support Care Cancer. 2016;24:4057–74. doi: 10.1007/s00520-016-3310-3. [DOI] [PubMed] [Google Scholar]
  • 67.Motzer RJ, Hutson TE, Tomczak P, et al. Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma. J Clin Oncol. 2009;27:3584–90. doi: 10.1200/JCO.2008.20.1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Telli ML, Witteles RM, Fisher GA, Srinivas S. Cardiotoxicity associated with the cancer therapeutic agent sunitinib malate. Ann Oncol. 2008;19:1613–8. doi: 10.1093/annonc/mdn168. [DOI] [PubMed] [Google Scholar]
  • 69.Richards CJ, Je Y, Schutz FA, et al. Incidence and risk of congestive heart failure in patients with renal and nonrenal cell carcinoma treated with sunitinib. J Clin Oncol. 2011;29:3450–6. doi: 10.1200/JCO.2010.34.4309. [DOI] [PubMed] [Google Scholar]
  • 70.Chu TF, Rupnick MA, Kerkela R, et al. Cardiotoxicity associated with tyrosine kinase inhibitor sunitinib. Lancet. 2007;370:2011–9. doi: 10.1016/S0140-6736(07)61865-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Hall PS, Harshman LC, Srinivas S, Witteles RM. The frequency and severity of cardiovascular toxicity from targeted therapy in advanced renal cell carcinoma patients. JACC Heart Fail. 2013;1:72–8. doi: 10.1016/j.jchf.2012.09.001. [DOI] [PubMed] [Google Scholar]
  • 72.Uraizee I, Cheng S, Moslehi J. Reversible cardiomyopathy associated with sunitinib and sorafenib. N Engl J Med. 2011;365:1649–50. doi: 10.1056/NEJMc1108849. [DOI] [PubMed] [Google Scholar]
  • 73.Gupta A, Pandey A, Sethi S. Bortezomib-induced congestive cardiac failure in a patient with multiple myeloma. Cardiovasc Toxicol. 2012;12:184–7. doi: 10.1007/s12012-011-9146-7. [DOI] [PubMed] [Google Scholar]
  • 74.Hacihanefioglu A, Tarkun P, Gonullu E. Acute severe cardiac failure in a myeloma patient due to proteasome inhibitor bortezomib. Int J Hematol. 2008;88:219–22. doi: 10.1007/s12185-008-0139-7. [DOI] [PubMed] [Google Scholar]
  • 75.Jerkins JH, Suciu A, Mazimba S, Calvo A. Bortezomib-induced Severe Congestive Heart Failure. Cardiol Res. 2010;1:20–23. doi: 10.4021/cr105e. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Maurea N, Spallarossa P, Cadeddu C, et al. 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):e93–e104. doi: 10.2459/JCM.0000000000000383. Special issue on Cardiotoxicity from Antiblastic Drugs and Cardioprotection. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Siegel DS, Martin T, Wang M, et al. A phase 2 study of single-agent carfilzomib (PX-171-003-A1) in patients with relapsed and refractory multiple myeloma. Blood. 2012;120:2817–25. doi: 10.1182/blood-2012-05-425934. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Grandin EW, Ky B, Cornell RF, Carver J, Lenihan DJ. Patterns of cardiac toxicity associated with irreversible proteasome inhibition in the treatment of multiple myeloma. J Card Fail. 2015;21:138–44. doi: 10.1016/j.cardfail.2014.11.008. [DOI] [PubMed] [Google Scholar]
  • 79.Goff DC, Jr, Lloyd-Jones DM, Bennett G, et al. 2013 ACC/AHA guideline on the assessment of cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2014;63:2935–59. doi: 10.1016/j.jacc.2013.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Chicco AJ, Hydock DS, Schneider CM, Hayward R. Low-intensity exercise training during doxorubicin treatment protects against cardiotoxicity. J Appl Physiol (1985) 2006;100:519–27. doi: 10.1152/japplphysiol.00148.2005. [DOI] [PubMed] [Google Scholar]
  • 81.Dranitsaris G, Rayson D, Vincent M, et al. The development of a predictive model to estimate cardiotoxic risk for patients with metastatic breast cancer receiving anthracyclines. Breast Cancer Res Treat. 2008;107:443–50. doi: 10.1007/s10549-007-9803-5. [DOI] [PubMed] [Google Scholar]
  • 82.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. doi: 10.1161/JAHA.113.000472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Herrmann J, Lerman A, Sandhu NP, Villarraga HR, Mulvagh SL, Kohli M. Evaluation and management of patients with heart disease and cancer: cardio-oncology. Mayo Clin Proc. 2014;89:1287–306. doi: 10.1016/j.mayocp.2014.05.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Thavendiranathan P, Grant AD, Negishi T, Plana JC, Popovic 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. doi: 10.1016/j.jacc.2012.09.035. [DOI] [PubMed] [Google Scholar]
  • 85.Gorcsan J, 3rd, Tanaka H. Echocardiographic assessment of myocardial strain. J Am Coll Cardiol. 2011;58:1401–13. doi: 10.1016/j.jacc.2011.06.038. [DOI] [PubMed] [Google Scholar]
  • 86.Geyer H, Caracciolo G, Abe H, et al. Assessment of myocardial mechanics using speckle tracking echocardiography: fundamentals and clinical applications. J Am Soc Echocardiogr. 2010;23:351–69. doi: 10.1016/j.echo.2010.02.015. quiz 453–5. [DOI] [PubMed] [Google Scholar]
  • 87.Perk G, Tunick PA, Kronzon I. Non-Doppler two-dimensional strain imaging by echocardiography–from technical considerations to clinical applications. J Am Soc Echocardiogr. 2007;20:234–43. doi: 10.1016/j.echo.2006.08.023. [DOI] [PubMed] [Google Scholar]
  • 88.Fallah-Rad N, Walker JR, Wassef A, et al. The utility of cardiac biomarkers, tissue velocity and strain imaging, and cardiac magnetic resonance imaging in predicting early left ventricular dysfunction in patients with human epidermal growth factor receptor II-positive breast cancer treated with adjuvant trastuzumab therapy. J Am Coll Cardiol. 2011;57:2263–70. doi: 10.1016/j.jacc.2010.11.063. [DOI] [PubMed] [Google Scholar]
  • 89.Sawaya H, Sebag IA, Plana JC, et al. 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. doi: 10.1161/CIRCIMAGING.112.973321. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.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;26:493–8. doi: 10.1016/j.echo.2013.02.008. [DOI] [PubMed] [Google Scholar]
  • 91.Ho E, Brown A, Barrett P, et al. Subclinical anthracycline- and trastuzumab-induced cardiotoxicity in the long-term follow-up of asymptomatic breast cancer survivors: a speckle tracking echocardiographic study. Heart. 2010;96:701–7. doi: 10.1136/hrt.2009.173997. [DOI] [PubMed] [Google Scholar]
  • 92.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;63:2751–68. doi: 10.1016/j.jacc.2014.01.073. [DOI] [PubMed] [Google Scholar]
  • 93.Cardinale D, Colombo A, Torrisi R, et al. Trastuzumab-induced cardiotoxicity: clinical and prognostic implications of troponin I evaluation. J Clin Oncol. 2010;28:3910–6. doi: 10.1200/JCO.2009.27.3615. [DOI] [PubMed] [Google Scholar]
  • 94.Telli ML, Hunt SA, Carlson RW, Guardino AE. Trastuzumab-related cardiotoxicity: calling into question the concept of reversibility. J Clin Oncol. 2007;25:3525–33. doi: 10.1200/JCO.2007.11.0106. [DOI] [PubMed] [Google Scholar]
  • 95.Braunwald E. Biomarkers in heart failure. N Engl J Med. 2008;358:2148–59. doi: 10.1056/NEJMra0800239. [DOI] [PubMed] [Google Scholar]
  • 96.Ky B, French B, Levy WC, et al. Multiple biomarkers for risk prediction in chronic heart failure. Circ Heart Fail. 2012;5:183–90. doi: 10.1161/CIRCHEARTFAILURE.111.965020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Zethelius B, Berglund L, Sundstrom J, et al. Use of multiple biomarkers to improve the prediction of death from cardiovascular causes. N Engl J Med. 2008;358:2107–16. doi: 10.1056/NEJMoa0707064. [DOI] [PubMed] [Google Scholar]
  • 98.Katus HA, Remppis A, Looser S, Hallermeier K, Scheffold T, Kubler W. Enzyme linked immuno assay of cardiac troponin T for the detection of acute myocardial infarction in patients. J Mol Cell Cardiol. 1989;21:1349–53. doi: 10.1016/0022-2828(89)90680-9. [DOI] [PubMed] [Google Scholar]
  • 99.Balk EM, Ioannidis JP, Salem D, Chew PW, Lau J. Accuracy of biomarkers to diagnose acute cardiac ischemia in the emergency department: a meta-analysis. Ann Emerg Med. 2001;37:478–94. doi: 10.1067/mem.2001.114905. [DOI] [PubMed] [Google Scholar]
  • 100.Jaffe AS, Landt Y, Parvin CA, Abendschein DR, Geltman EM, Ladenson JH. Comparative sensitivity of cardiac troponin I and lactate dehydrogenase isoenzymes for diagnosing acute myocardial infarction. Clin Chem. 1996;42:1770–6. [PubMed] [Google Scholar]
  • 101.Cardinale D, Sandri MT, Martinoni A, et al. Left ventricular dysfunction predicted by early troponin I release after high-dose chemotherapy. J Am Coll Cardiol. 2000;36:517–22. doi: 10.1016/s0735-1097(00)00748-8. [DOI] [PubMed] [Google Scholar]
  • 102.Auner HW, Tinchon C, Linkesch W, et al. Prolonged monitoring of troponin T for the detection of anthracycline cardiotoxicity in adults with hematological malignancies. Ann Hematol. 2003;82:218–22. doi: 10.1007/s00277-003-0615-3. [DOI] [PubMed] [Google Scholar]
  • 103.Lipshultz SE, Miller TL, Scully RE, et al. Changes in cardiac biomarkers during doxorubicin treatment of pediatric patients with high-risk acute lymphoblastic leukemia: associations with long-term echocardiographic outcomes. J Clin Oncol. 2012;30:1042–9. doi: 10.1200/JCO.2010.30.3404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Lipshultz SE, Scully RE, Lipsitz SR, et al. Assessment of dexrazoxane as a cardioprotectant in doxorubicin-treated children with high-risk acute lymphoblastic leukaemia: long-term follow-up of a prospective, randomised, multicentre trial. Lancet Oncol. 2010;11:950–61. doi: 10.1016/S1470-2045(10)70204-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Dodos F, Halbsguth T, Erdmann E, Hoppe UC. Usefulness of myocardial performance index and biochemical markers for early detection of anthracycline-induced cardiotoxicity in adults. Clin Res Cardiol. 2008;97:318–26. doi: 10.1007/s00392-007-0633-6. [DOI] [PubMed] [Google Scholar]
  • 106.McArthur HL, Rugo H, Nulsen B, et al. A feasibility study of bevacizumab plus dose-dense doxorubicin-cyclophosphamide (AC) followed by nanoparticle albumin-bound paclitaxel in early-stage breast cancer. Clin Cancer Res. 2011;17:3398–407. doi: 10.1158/1078-0432.CCR-10-1969. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Blaes AH, Rehman A, Vock DM, et al. Utility of high-sensitivity cardiac troponin T in patients receiving anthracycline chemotherapy. Vasc Health Risk Manag. 2015;11:591–4. doi: 10.2147/VHRM.S89842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Germanakis I, Anagnostatou N, Kalmanti M. Troponins and natriuretic peptides in the monitoring of anthracycline cardiotoxicity. Pediatr Blood Cancer. 2008;51:327–33. doi: 10.1002/pbc.21633. [DOI] [PubMed] [Google Scholar]
  • 109.Broeyer FJ, Osanto S, Ritsema van Eck HJ, et al. Evaluation of biomarkers for cardiotoxicity of anthracyclin-based chemotherapy. J Cancer Res Clin Oncol. 2008;134:961–8. doi: 10.1007/s00432-008-0372-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Lee HS, Son CB, Shin SH, Kim YS. Clinical correlation between brain natriutetic peptide and anthracyclin-induced cardiac toxicity. Cancer Res Treat. 2008;40:121–6. doi: 10.4143/crt.2008.40.3.121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Mavinkurve-Groothuis AM, Kapusta L, Nir A, Groot-Loonen J. The role of biomarkers in the early detection of anthracycline-induced cardiotoxicity in children: a review of the literature. Pediatr Hematol Oncol. 2008;25:655–64. doi: 10.1080/08880010802244001. [DOI] [PubMed] [Google Scholar]
  • 112.Suzuki T, Hayashi D, Yamazaki T, et al. Elevated B-type natriuretic peptide levels after anthracycline administration. Am Heart J. 1998;136:362–3. doi: 10.1053/hj.1998.v136.89908. [DOI] [PubMed] [Google Scholar]
  • 113.Nousiainen T, Vanninen E, Jantunen E, et al. Natriuretic peptides during the development of doxorubicin-induced left ventricular diastolic dysfunction. J Intern Med. 2002;251:228–34. doi: 10.1046/j.1365-2796.2002.00951.x. [DOI] [PubMed] [Google Scholar]
  • 114.Meinardi MT, van Veldhuisen DJ, Gietema JA, et al. Prospective evaluation of early cardiac damage induced by epirubicin-containing adjuvant chemotherapy and locoregional radiotherapy in breast cancer patients. J Clin Oncol. 2001;19:2746–53. doi: 10.1200/JCO.2001.19.10.2746. [DOI] [PubMed] [Google Scholar]
  • 115.Daugaard G, Lassen U, Bie P, et al. Natriuretic peptides in the monitoring of anthracycline induced reduction in left ventricular ejection fraction. Eur J Heart Fail. 2005;7:87–93. doi: 10.1016/j.ejheart.2004.03.009. [DOI] [PubMed] [Google Scholar]
  • 116.Ky B, Putt M, Sawaya H, et al. Early increases in multiple biomarkers predict subsequent cardiotoxicity in patients with breast cancer treated with doxorubicin, taxanes, and trastuzumab. J Am Coll Cardiol. 2014;63:809–16. doi: 10.1016/j.jacc.2013.10.061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Onitilo AA, Engel JM, Stankowski RV, Liang H, Berg RL, Doi SA. High-sensitivity C-reactive protein (hs-CRP) as a biomarker for trastuzumab-induced cardiotoxicity in HER2-positive early-stage breast cancer: a pilot study. Breast Cancer Res Treat. 2012;134:291–8. doi: 10.1007/s10549-012-2039-z. [DOI] [PubMed] [Google Scholar]
  • 118.Sendur MA, Aksoy S, Yorgun H, et al. Comparison of the long term cardiac effects associated with 9 and 52 weeks of trastuzumab in HER2-positive early breast cancer. Current medical research and opinion. 2015;31:547–56. doi: 10.1185/03007995.2015.1005834. [DOI] [PubMed] [Google Scholar]
  • 119.de Boer RA, Voors AA, Muntendam P, van Gilst WH, van Veldhuisen DJ. Galectin-3: a novel mediator of heart failure development and progression. Eur J Heart Fail. 2009;11:811–7. doi: 10.1093/eurjhf/hfp097. [DOI] [PubMed] [Google Scholar]
  • 120.Feola M, Garrone O, Occelli M, et al. Cardiotoxicity after anthracycline chemotherapy in breast carcinoma: effects on left ventricular ejection fraction, troponin I and brain natriuretic peptide. Int J Cardiol. 2011;148:194–8. doi: 10.1016/j.ijcard.2009.09.564. [DOI] [PubMed] [Google Scholar]
  • 121.Armenian SH, Gelehrter SK, Vase T, et al. Screening for cardiac dysfunction in anthracycline-exposed childhood cancer survivors. Clin Cancer Res. 2014;20:6314–23. doi: 10.1158/1078-0432.CCR-13-3490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Slamon D, Eiermann W, Robert N, et al. Adjuvant trastuzumab in HER2-positive breast cancer. N Engl J Med. 2011;365:1273–83. doi: 10.1056/NEJMoa0910383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Poole CJ, Earl HM, Hiller L, et al. Epirubicin and cyclophosphamide, methotrexate, and fluorouracil as adjuvant therapy for early breast cancer. N Engl J Med. 2006;355:1851–62. doi: 10.1056/NEJMoa052084. [DOI] [PubMed] [Google Scholar]
  • 124.Li X, Xu S, Tan Y, Chen J. The effects of idarubicin versus other anthracyclines for induction therapy of patients with newly diagnosed leukaemia. Cochrane Database Syst Rev. 2015:CD010432. doi: 10.1002/14651858.CD010432.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.van Dalen EC, Michiels EM, Caron HN, Kremer LC. Different anthracycline derivates for reducing cardiotoxicity in cancer patients. Cochrane Database Syst Rev. 2010:CD005006. doi: 10.1002/14651858.CD005006.pub2. [DOI] [PubMed] [Google Scholar]
  • 126.Minotti G, Menna P, Salvatorelli E, Cairo G, Gianni L. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. PharmacolRev. 2004;56:185–229. doi: 10.1124/pr.56.2.6. [DOI] [PubMed] [Google Scholar]
  • 127.van Dalen EC, van der Pal HJ, Kremer LC. Different dosage schedules for reducing cardiotoxicity in people with cancer receiving anthracycline chemotherapy. Cochrane Database Syst Rev. 2016;3:CD005008. doi: 10.1002/14651858.CD005008.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Lipshultz SE, Miller TL, Lipsitz SR, et al. Continuous Versus Bolus Infusion of Doxorubicin in Children With ALL: Long-term Cardiac Outcomes. Pediatrics. 2012;130:1003–11. doi: 10.1542/peds.2012-0727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Lipshultz SE, Cochran TR, Franco VI, Miller TL. Treatment-related cardiotoxicity in survivors of childhood cancer. Nat Rev Clin Oncol. 2013;10:697–710. doi: 10.1038/nrclinonc.2013.195. [DOI] [PubMed] [Google Scholar]
  • 130.Gupta M, Steinherz PG, Cheung NK, Steinherz L. Late cardiotoxicity after bolus versus infusion anthracycline therapy for childhood cancers. Med Pediatr Oncol. 2003;40:343–7. doi: 10.1002/mpo.10298. [DOI] [PubMed] [Google Scholar]
  • 131.Levitt GA, Dorup I, Sorensen K, Sullivan I. Does anthracycline administration by infusion in children affect late cardiotoxicity? Br J Haematol. 2004;124:463–8. doi: 10.1111/j.1365-2141.2004.04803.x. [DOI] [PubMed] [Google Scholar]
  • 132.Pignata S, Scambia G, Ferrandina G, et al. Carboplatin plus paclitaxel versus carboplatin plus pegylated liposomal doxorubicin as first-line treatment for patients with ovarian cancer: the MITO-2 randomized phase III trial. J Clin Oncol. 2011;29:3628–35. doi: 10.1200/JCO.2010.33.8566. [DOI] [PubMed] [Google Scholar]
  • 133.Sharpe M, Easthope SE, Keating GM, Lamb HM. Polyethylene glycol-liposomal doxorubicin: a review of its use in the management of solid and haematological malignancies and AIDS-related Kaposi’s sarcoma. Drugs. 2002;62:2089–126. doi: 10.2165/00003495-200262140-00012. [DOI] [PubMed] [Google Scholar]
  • 134.Al-Batran SE, Guntner M, Pauligk C, et al. Anthracycline rechallenge using pegylated liposomal doxorubicin in patients with metastatic breast cancer: a pooled analysis using individual data from four prospective trials. Br J Cancer. 2010;103:1518–23. doi: 10.1038/sj.bjc.6605961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Keller AM, Mennel RG, Georgoulias VA, et al. Randomized phase III trial of pegylated liposomal doxorubicin versus vinorelbine or mitomycin C plus vinblastine in women with taxane-refractory advanced breast cancer. J Clin Oncol. 2004;22:3893–901. doi: 10.1200/JCO.2004.08.157. [DOI] [PubMed] [Google Scholar]
  • 136.O’Brien ME, Wigler N, Inbar M, et al. Reduced cardiotoxicity and comparable efficacy in a phase III trial of pegylated liposomal doxorubicin HCl (CAELYX/Doxil) versus conventional doxorubicin for first-line treatment of metastatic breast cancer. Ann Oncol. 2004;15:440–9. doi: 10.1093/annonc/mdh097. [DOI] [PubMed] [Google Scholar]
  • 137.Gordon AN, Fleagle JT, Guthrie D, Parkin DE, Gore ME, Lacave AJ. Recurrent epithelial ovarian carcinoma: a randomized phase III study of pegylated liposomal doxorubicin versus topotecan. J Clin Oncol. 2001;19:3312–22. doi: 10.1200/JCO.2001.19.14.3312. [DOI] [PubMed] [Google Scholar]
  • 138.Mutch DG, Orlando M, Goss T, et al. Randomized phase III trial of gemcitabine compared with pegylated liposomal doxorubicin in patients with platinum-resistant ovarian cancer. J Clin Oncol. 2007;25:2811–8. doi: 10.1200/JCO.2006.09.6735. [DOI] [PubMed] [Google Scholar]
  • 139.Pujade-Lauraine E, Wagner U, Aavall-Lundqvist E, et al. Pegylated liposomal Doxorubicin and Carboplatin compared with Paclitaxel and Carboplatin for patients with platinum-sensitive ovarian cancer in late relapse. J Clin Oncol. 2010;28:3323–9. doi: 10.1200/JCO.2009.25.7519. [DOI] [PubMed] [Google Scholar]
  • 140.Duggan ST, Keating GM. Pegylated liposomal doxorubicin: a review of its use in metastatic breast cancer, ovarian cancer, multiple myeloma and AIDS-related Kaposi’s sarcoma. Drugs. 2011;71:2531–58. doi: 10.2165/11207510-000000000-00000. [DOI] [PubMed] [Google Scholar]
  • 141.Smith DH, Adams JR, Johnston SR, Gordon A, Drummond MF, Bennett CL. A comparative economic analysis of pegylated liposomal doxorubicin versus topotecan in ovarian cancer in the USA and the UK. Ann Oncol. 2002;13:1590–7. doi: 10.1093/annonc/mdf275. [DOI] [PubMed] [Google Scholar]
  • 142.Creighton AM, Birnie GD. The effect of bisdioxopiperazines on the synthesis of deoxyribonucleic acid, ribonucleic acid and protein in growing mouse-embryo fibroblasts. The Biochemical journal. 1969;114:58P. doi: 10.1042/bj1140058pa. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Lyu YL, Kerrigan JE, Lin CP, et al. Topoisomerase IIbeta mediated DNA double-strand breaks: implications in doxorubicin cardiotoxicity and prevention by dexrazoxane. Cancer research. 2007;67:8839–46. doi: 10.1158/0008-5472.CAN-07-1649. [DOI] [PubMed] [Google Scholar]
  • 144.Marty M, Espie M, Llombart A, et al. Multicenter randomized phase III study of the cardioprotective effect of dexrazoxane (Cardioxane) in advanced/metastatic breast cancer patients treated with anthracycline-based chemotherapy. Ann Oncol. 2006;17:614–22. doi: 10.1093/annonc/mdj134. [DOI] [PubMed] [Google Scholar]
  • 145.Speyer JL, Green MD, Zeleniuch-Jacquotte A, et al. ICRF-187 permits longer treatment with doxorubicin in women with breast cancer. J Clin Oncol. 1992;10:117–27. doi: 10.1200/JCO.1992.10.1.117. [DOI] [PubMed] [Google Scholar]
  • 146.Yu Y, Kalinowski DS, Kovacevic Z, et al. Thiosemicarbazones from the old to new: iron chelators that are more than just ribonucleotide reductase inhibitors. Journal of medicinal chemistry. 2009;52:5271–94. doi: 10.1021/jm900552r. [DOI] [PubMed] [Google Scholar]
  • 147.Swain SM, Vici P. The current and future role of dexrazoxane as a cardioprotectant in anthracycline treatment: expert panel review. J Cancer Res Clin Oncol. 2004;130:1–7. doi: 10.1007/s00432-003-0498-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Seymour L, Bramwell V, Moran LA. Use of dexrazoxane as a cardioprotectant in patients receiving doxorubicin or epirubicin chemotherapy for the treatment of cancer. The Provincial Systemic Treatment Disease Site Group. Cancer prevention & control : CPC = Prevention & controle en cancerologie : PCC. 1999;3:145–59. [PubMed] [Google Scholar]
  • 149.van Dalen EC, Caron HN, Dickinson HO, Kremer LC. Cardioprotective interventions for cancer patients receiving anthracyclines. Cochrane Database Syst Rev. 2008:CD003917. doi: 10.1002/14651858.CD003917.pub3. [DOI] [PubMed] [Google Scholar]
  • 150.Schuchter LM, Hensley ML, Meropol NJ, Winer EP, American Society of Clinical Oncology C, Radiotherapy Expert P 2002 update of recommendations for the use of chemotherapy and radiotherapy protectants: clinical practice guidelines of the American Society of Clinical Oncology. J Clin Oncol. 2002;20:2895–903. doi: 10.1200/JCO.2002.04.178. [DOI] [PubMed] [Google Scholar]
  • 151.Tebbi CK, London WB, Friedman D, et al. Dexrazoxane-associated risk for acute myeloid leukemia/myelodysplastic syndrome and other secondary malignancies in pediatric Hodgkin’s disease. J Clin Oncol. 2007;25:493–500. doi: 10.1200/JCO.2005.02.3879. [DOI] [PubMed] [Google Scholar]
  • 152.Salzer WL, Devidas M, Carroll WL, et al. Long-term results of the pediatric oncology group studies for childhood acute lymphoblastic leukemia 1984–2001: a report from the children’s oncology group. Leukemia. 2010;24:355–70. doi: 10.1038/leu.2009.261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Vrooman LM, Neuberg DS, Stevenson KE, et al. The low incidence of secondary acute myelogenous leukaemia in children and adolescents treated with dexrazoxane for acute lymphoblastic leukaemia: a report from the Dana-Farber Cancer Institute ALL Consortium. Eur J Cancer. 2011;47:1373–9. doi: 10.1016/j.ejca.2011.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Romond EH, Jeong JH, Rastogi P, et al. Seven-year follow-up assessment of cardiac function in NSABP B-31, a randomized trial comparing doxorubicin and cyclophosphamide followed by paclitaxel (ACP) with ACP plus trastuzumab as adjuvant therapy for patients with node-positive, human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol. 2012;30:3792–9. doi: 10.1200/JCO.2011.40.0010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Joensuu H, Kellokumpu-Lehtinen PL, Bono P, et al. Adjuvant docetaxel or vinorelbine with or without trastuzumab for breast cancer. N Engl J Med. 2006;354:809–20. doi: 10.1056/NEJMoa053028. [DOI] [PubMed] [Google Scholar]
  • 156.Yancy CW, Jessup M, Bozkurt B, et al. 2017 ACC/AHA/HFSA Focused 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. Circulation. 2017 doi: 10.1161/CIR.0000000000000509. [DOI] [PubMed] [Google Scholar]
  • 157.Yancy CW, Jessup M, Bozkurt B, et al. 2013 ACCF/AHA guideline for the management of heart failure: executive summary: a report of the American College of Cardiology Foundation/American Heart Association Task Force on practice guidelines. Circulation. 2013;128:1810–52. doi: 10.1161/CIR.0b013e31829e8807. [DOI] [PubMed] [Google Scholar]
  • 158.Ewer MS, Lippman SM. Type II chemotherapy-related cardiac dysfunction: time to recognize a new entity. J Clin Oncol. 2005;23:2900–2. doi: 10.1200/JCO.2005.05.827. [DOI] [PubMed] [Google Scholar]
  • 159.Meyer CC, Calis KA, Burke LB, Walawander CA, Grasela TH. Symptomatic cardiotoxicity associated with 5-fluorouracil. Pharmacotherapy. 1997;17:729–36. [PubMed] [Google Scholar]
  • 160.Van Cutsem E, Hoff PM, Blum JL, Abt M, Osterwalder B. Incidence of cardiotoxicity with the oral fluoropyrimidine capecitabine is typical of that reported with 5-fluorouracil. Ann Oncol. 2002;13:484–5. doi: 10.1093/annonc/mdf108. [DOI] [PubMed] [Google Scholar]
  • 161.de Forni M, Malet-Martino MC, Jaillais P, et al. Cardiotoxicity of high-dose continuous infusion fluorouracil: a prospective clinical study. J Clin Oncol. 1992;10:1795–801. doi: 10.1200/JCO.1992.10.11.1795. [DOI] [PubMed] [Google Scholar]
  • 162.Bertolini A, Flumano M, Fusco O, et al. Acute cardiotoxicity during capecitabine treatment: a case report. Tumori. 2001;87:200–6. doi: 10.1177/030089160108700317. [DOI] [PubMed] [Google Scholar]
  • 163.Anand AJ. Fluorouracil cardiotoxicity. Ann Pharmacother. 1994;28:374–8. doi: 10.1177/106002809402800314. [DOI] [PubMed] [Google Scholar]
  • 164.Cardinale D, Colombo A, Colombo N. Acute coronary syndrome induced by oral capecitabine. Can J Cardiol. 2006;22:251–3. doi: 10.1016/s0828-282x(06)70905-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Jensen SA, Sorensen JB. Risk factors and prevention of cardiotoxicity induced by 5-fluorouracil or capecitabine. Cancer Chemother Pharmacol. 2006;58:487–93. doi: 10.1007/s00280-005-0178-1. [DOI] [PubMed] [Google Scholar]
  • 166.Kosmas C, Kallistratos MS, Kopterides P, et al. Cardiotoxicity of fluoropyrimidines in different schedules of administration: a prospective study. J Cancer Res Clin Oncol. 2008;134:75–82. doi: 10.1007/s00432-007-0250-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Labianca R, Beretta G, Clerici M, Fraschini P, Luporini G. Cardiac toxicity of 5-fluorouracil: a study on 1083 patients. Tumori. 1982;68:505–10. doi: 10.1177/030089168206800609. [DOI] [PubMed] [Google Scholar]
  • 168.Rezkalla S, Kloner RA, Ensley J, et al. Continuous ambulatory ECG monitoring during fluorouracil therapy: a prospective study. J Clin Oncol. 1989;7:509–14. doi: 10.1200/JCO.1989.7.4.509. [DOI] [PubMed] [Google Scholar]
  • 169.Schnetzler B, Popova N, Collao Lamb C, Sappino AP. Coronary spasm induced by capecitabine. Ann Oncol. 2001;12:723–4. doi: 10.1023/a:1011152931300. [DOI] [PubMed] [Google Scholar]
  • 170.Robben NC, Pippas AW, Moore JO. The syndrome of 5-fluorouracil cardiotoxicity. An elusive cardiopathy. Cancer. 1993;71:493–509. doi: 10.1002/1097-0142(19930115)71:2<493::aid-cncr2820710235>3.0.co;2-c. [DOI] [PubMed] [Google Scholar]
  • 171.Ng M, Cunningham D, Norman AR. The frequency and pattern of cardiotoxicity observed with capecitabine used in conjunction with oxaliplatin in patients treated for advanced colorectal cancer (CRC) Eur J Cancer. 2005;41:1542–6. doi: 10.1016/j.ejca.2005.03.027. [DOI] [PubMed] [Google Scholar]
  • 172.Saif MW, Tomita M, Ledbetter L, Diasio RB. Capecitabine-related cardiotoxicity: recognition and management. J Support Oncol. 2008;6:41–8. [PubMed] [Google Scholar]
  • 173.Walko CM, Lindley C. Capecitabine: a review. Clin Ther. 2005;27:23–44. doi: 10.1016/j.clinthera.2005.01.005. [DOI] [PubMed] [Google Scholar]
  • 174.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. doi: 10.1093/annonc/mdf035. [DOI] [PubMed] [Google Scholar]
  • 175.Papadopoulos CA, Wilson H. Capecitabine-associated coronary vasospasm: a case report. Emerg Med J. 2008;25:307–9. doi: 10.1136/emj.2007.055681. [DOI] [PubMed] [Google Scholar]
  • 176.Arbuck SG, Strauss H, Rowinsky E, et al. A reassessment of cardiac toxicity associated with Taxol. J Natl Cancer Inst Monogr. 1993:117–30. [PubMed] [Google Scholar]
  • 177.Vermorken JB, Remenar E, van Herpen C, et al. Cisplatin, fluorouracil, and docetaxel in unresectable head and neck cancer. N Engl J Med. 2007;357:1695–704. doi: 10.1056/NEJMoa071028. [DOI] [PubMed] [Google Scholar]
  • 178.Sanofi. Taxotere Prescribing Information. 2015 Accessed at: http://products.sanofi.us/Taxotere/taxotere.pdf.
  • 179.Scappaticci FA, Skillings JR, Holden SN, et al. Arterial thromboembolic events in patients with metastatic carcinoma treated with chemotherapy and bevacizumab. J Natl Cancer Inst. 2007;99:1232–9. doi: 10.1093/jnci/djm086. [DOI] [PubMed] [Google Scholar]
  • 180.Kozloff M, Yood MU, Berlin J, et al. Clinical outcomes associated with bevacizumab-containing treatment of metastatic colorectal cancer: the BRiTE observational cohort study. Oncologist. 2009;14:862–70. doi: 10.1634/theoncologist.2009-0071. [DOI] [PubMed] [Google Scholar]
  • 181.Moore MJ, Goldstein D, Hamm J, et al. Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: a phase III trial of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol. 2007;25:1960–6. doi: 10.1200/JCO.2006.07.9525. [DOI] [PubMed] [Google Scholar]
  • 182.Administration. FaD. Tarceva (erlotinib) label. 2010 Accessed at https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/021743s14s16lbl.pdf.
  • 183.Pharma. B. Sorafenib (Nexavar) package insert. 2013 Accessed at http://labeling.bayerhealthcare.com/html/products/pi/Nexavar_PI.pdf.
  • 184.Escudier B, Eisen T, Stadler WM, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356:125–34. doi: 10.1056/NEJMoa060655. [DOI] [PubMed] [Google Scholar]
  • 185.Siegel D, Martin T, Nooka A, et al. Integrated safety profile of single-agent carfilzomib: experience from 526 patients enrolled in 4 phase II clinical studies. Haematologica. 2013;98:1753–61. doi: 10.3324/haematol.2013.089334. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Stewart AK, Rajkumar SV, Dimopoulos MA, et al. Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma. N Engl J Med. 2015;372:142–52. doi: 10.1056/NEJMoa1411321. [DOI] [PubMed] [Google Scholar]
  • 187.Amgen. Kyprolis (carfilzomib). Prescribing information. 2017 Accessed at: http://pi.amgen.com/~/media/amgen/repositorysites/pi-amgen-com/kyprolis/kyprolis_pi.ashx.
  • 188.Shoemaker LK, Arora U, Rocha Lima CM. 5-fluorouracil-induced coronary vasospasm. Cancer Control. 2004;11:46–9. doi: 10.1177/107327480401100207. [DOI] [PubMed] [Google Scholar]
  • 189.Mosseri M, Fingert HJ, Varticovski L, Chokshi S, Isner JM. In vitro evidence that myocardial ischemia resulting from 5-fluorouracil chemotherapy is due to protein kinase C-mediated vasoconstriction of vascular smooth muscle. Cancer research. 1993;53:3028–33. [PubMed] [Google Scholar]
  • 190.Cwikiel M, Eskilsson J, Albertsson M, Stavenow L. The influence of 5-fluorouracil and methotrexate on vascular endothelium. An experimental study using endothelial cells in the culture. Ann Oncol. 1996;7:731–7. doi: 10.1093/oxfordjournals.annonc.a010723. [DOI] [PubMed] [Google Scholar]
  • 191.Cwikiel M, Eskilsson J, Wieslander JB, Stjernquist U, Albertsson M. The appearance of endothelium in small arteries after treatment with 5-fluorouracil. An electron microscopic study of late effects in rabbits. Scanning Microsc. 1996;10:805–18. discussion 819. [PubMed] [Google Scholar]
  • 192.Cwikiel M, Zhang B, Eskilsson J, Wieslander JB, Albertsson M. The influence of 5-fluorouracil on the endothelium in small arteries. An electron microscopic study in rabbits. Scanning Microsc. 1995;9:561–76. [PubMed] [Google Scholar]
  • 193.Jensen SA, Sorensen JB. 5-fluorouracil-based therapy induces endovascular injury having potential significance to development of clinically overt cardiotoxicity. Cancer Chemother Pharmacol. 2012;69:57–64. doi: 10.1007/s00280-011-1669-x. [DOI] [PubMed] [Google Scholar]
  • 194.Kinhult S, Albertsson M, Eskilsson J, Cwikiel M. Antithrombotic treatment in protection against thrombogenic effects of 5-fluorouracil on vascular endothelium: a scanning microscopy evaluation. Scanning. 2001;23:1–8. doi: 10.1002/sca.4950230101. [DOI] [PubMed] [Google Scholar]
  • 195.Rowinsky EK, McGuire WP, Guarnieri T, Fisherman JS, Christian MC, Donehower RC. Cardiac disturbances during the administration of taxol. J Clin Oncol. 1991;9:1704–12. doi: 10.1200/JCO.1991.9.9.1704. [DOI] [PubMed] [Google Scholar]
  • 196.Kamba T, McDonald DM. Mechanisms of adverse effects of anti-VEGF therapy for cancer. Br J Cancer. 2007;96:1788–95. doi: 10.1038/sj.bjc.6603813. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Kilickap S, Abali H, Celik I. Bevacizumab, bleeding, thrombosis, and warfarin. J Clin Oncol. 2003;21:3542. doi: 10.1200/JCO.2003.99.046. author reply 3543. [DOI] [PubMed] [Google Scholar]
  • 198.Chen-Scarabelli C, Corsetti G, Pasini E, et al. Spasmogenic Effects of the Proteasome Inhibitor Carfilzomib on Coronary Resistance, Vascular Tone and Reactivity. EBioMedicine. 2017 doi: 10.1016/j.ebiom.2017.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 199.Scarabelli TMC-SC, Gavazzoni M, Sahni G, Saravolatz L, Raddino R, Narula J. Cardiovascular effects of carfilzomib, a new proteasome inhibitor, on coronary resistances, vascular tone and vascular reactivity. 2015 doi: 10.1016/j.ebiom.2017.05.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Scarabelli TMGM, Chen-Scarabelli C, Sahni G, Saravolatz L, Narula J, Raddino R. Cardiovascular Toxicity of Carfilzomib on Vascular Tone, Vascular Reactivity and Endothelial function. 2014 [Google Scholar]
  • 201.Amsterdam EA, Wenger NK, Brindis RG, et al. 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. J Am Coll Cardiol. 2014;64:e139–228. doi: 10.1016/j.jacc.2014.09.017. [DOI] [PubMed] [Google Scholar]
  • 202.O’Gara PT, Kushner FG, Ascheim DD, et al. 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. Circulation. 2013;127:e362–425. doi: 10.1161/CIR.0b013e3182742cf6. [DOI] [PubMed] [Google Scholar]
  • 203.Sarkiss MG, Yusuf SW, Warneke CL, et al. Impact of aspirin therapy in cancer patients with thrombocytopenia and acute coronary syndromes. Cancer. 2007;109:621–7. doi: 10.1002/cncr.22434. [DOI] [PubMed] [Google Scholar]
  • 204.Iliescu C, Durand JB, Kroll M. Cardiovascular interventions in thrombocytopenic cancer patients. Tex Heart Inst J. 2011;38:259–60. [PMC free article] [PubMed] [Google Scholar]
  • 205.Iliescu CA, Grines CL, Herrmann J, et al. 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:E202–23. doi: 10.1002/ccd.26379. [DOI] [PubMed] [Google Scholar]
  • 206.Genentech I. Avastin (bavacizumab) label – Prescribing information. 2016 Accessed at: https://www.gene.com/download/pdf/avastin_prescribing.pdf.

RESOURCES