Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Jul 1.
Published in final edited form as: J Geriatr Oncol. 2017 May 9;8(4):308–314. doi: 10.1016/j.jgo.2017.04.001

Cardio-Oncology in the Older Adult

Prajwal Reddy 1, Chetan Shenoy 2, Anne H Blaes 3
PMCID: PMC5776715  NIHMSID: NIHMS876939  PMID: 28499724

Abstract

Heart disease and cancer are the leading causes of death in older adults. Many first-line cancer treatments have the potential for cardiotoxicity. Age-related risk factors, pre-existing cardiac disease, and a high prevalence of comorbidities are reasons for increased cardiotoxicity in older adults. Concerns regarding cardiotoxicity may lead to frailty bias and undertreatment, resulting in suboptimal outcomes. There is an urgent need for geriatric-specific evidence and guidelines to help tailor care for this vulnerable group. A multi-disciplinary approach based on close collaboration between oncologists, cardiologists, and geriatricians, among other specialist clinicians is essential.

Keywords: Cardio-oncology, geriatrics, older adult, cardiotoxicity, cardiotoxic treatments

Introduction

Cardio-oncology is an area of growing interest in recent years. Data are lacking on cardio-oncology specific to older adults. This is increasingly becoming important as advances in cancer treatments result in longer patient survival (1). The 5-year survival of patients diagnosed with cancer has steadily increased from 49% to 69% over the past three decades (1). The proportion of cancer survivors that are 65 years or older has increased exponentially. By 2020, it estimated that the number of cancer survivors will increase by 31% to 18 million, and that two-thirds of all cancer survivors will be 65 years or older (24). As the incidence of cancer rises with increasing age, so does the prevalence of cardiovascular disease (CVD). Those older than 65 years account for more than half of CVD hospitalizations and approximately 80% of deaths (5). Even more striking, those 75 years of age or older account for 50% of cardiovascular deaths despite accounting for only 6% of the population (5).

Despite the increased prevalence of older adults living with cancer and associated treatment toxicities, patients 65 years or older are underrepresented in clinical trials (57). Older patients are frequently offered lower doses of chemotherapy due to concerns of toxicity, presumed frailty, and the high likelihood of coexisting comorbidities. Several observational studies have noted poorer outcomes in this group possibly due to undertreatment and age-related factors (8, 9). As a result, optimal care of older adults with cancer necessitates evidence-based and guideline-directed care specific to this vulnerable group. Such care is especially critical for elderly patients at risk for cardiotoxicity.

Risk Factors of Cardiotoxicity

The 2016 American Society of Clinical Oncology (ASCO) Clinical Practice Guideline for the prevention and monitoring of cardiac dysfunction in survivors of adult cancers identifies the following risk factors for cardiac dysfunction: older age (age greater than 60 years), high-dose anthracycline therapy, high-dose radiotherapy, cardiovascular risk factors including smoking, diabetes, dyslipidemia, and obesity, borderline low cardiac function, valvular disease, and history of myocardial infarction (10). An estimated 80% of patients 60 years of age or older have at least one comorbid condition and 50% have two or more (11). In patients older than 80 years, as many as 70% have multiple comorbid conditions (11). In a retrospective analysis of patients receiving treatment for acute myeloid leukemia, a larger proportion of patients older than 60 years had significant comorbidities compared with those younger than 60 years (58.3% vs. 26.3%) (12). Not surprisingly, these comorbidities include diabetes mellitus, chronic obstructive lung disease, chronic kidney disease, and pre-existing heart disease (12). The cumulative effect of these factors, as illustrated in Figure 1 and previously described by Shenoy et al. (13), can be described as a “snowball effect.” The “snowball,” formed of baseline age-related factors is “set in motion” by the cancer diagnosis and is further exacerbated by cancer treatments which cause direct injury to tissues and organs. Polypharmacy and use of potentially inappropriate medications are prevalent in older adults and can exacerbate the toxicity associated with cancer treatments (1416). A multidisciplinary approach, including pharmacist involvement, is vital to preventing polypharmacy and inappropriate medication-related toxicities. Medication therapy management (MTM), consisting a thorough review of medications by a pharmacist, is increasingly being adopted in older patients and those with multiple comorbidities, including those with cancer (17). A growing number of retrospective studies indicate that MTM significantly reduces drug-drug interactions, adverse drug events, and non-adherence in older patients with cancer (17, 18).

Figure 1.

Figure 1

The “snowball effect” resulting in cardiovascular complications of cancer therapy in older adults. See text for details. Adapted with permission from Shenoy et al. (13)

Cancer Treatment-Specific Considerations

Comprehensive reviews exist on the cardiac toxicity of chemotherapy agents (10, 19, 20); in this article, cancer therapies with the highest risk for cardiotoxicity in the elderly population are discussed below including anthracyclines, trastuzumab, tyrosine kinase inhibitors, fluoropyrimidines, and radiation therapy.

Anthracyclines

Anthracyclines remain the first-line therapy for many cancers including breast cancer, hematological cancers, and sarcoma. They are incorporated in more than 50% of treatment regimens for these cancers (21, 22). They are also the most commonly associated cancer drugs known to cause irreversible dose-dependent cardiotoxicity. In a meta-analysis by Smith et al., an anthracycline-based regimen was associated with a higher risk of cardiotoxicity with an odds ratio of 5.43 compared with a non-anthracycline regimen (21). Around 75% of patients with anthracycline-related left ventricular systolic dysfunction do not have full recovery to normal function (23). The incidence of heart failure has been estimated at 2% at 200mg/m2, 5% at 400 mg/m2, 16% at 500 mg/m2, and 26% at 550 mg/m2 (24, 25). ASCO guidelines estimate a 1.6 to 6.8-fold increased risk of cardiac dysfunction in elderly patients, defined as 60 years of age or older, when compared to younger patients with cancer (10). Furthermore, studies limited to patients 65 years of age or older, were noted to have increasing risk of cardiac dysfunction with age (10). Elderly patients may be more susceptible to cardiotoxicity due to their age-related decrease in myocardial volume from cardiomyocyte loss and an increase in interstitial fibrosis. A study using cardiovascular magnetic resonance imaging (CMR) demonstrated that left ventricular mass has an inverse relationship with anthracycline dose and is a predictor of major adverse cardiovascular events (26). Therefore, elderly patients with a lower baseline myocardial volume may especially be vulnerable for cardiotoxicity, further loss of myocardial volume, and consequently, increased adverse cardiovascular outcomes. No specific recommendations regarding dose reductions can be made due to sparse data on cardiac dysfunction and left ventricular volume. A cumulative dose greater than 450 mg/m2 is generally limited due to the risk of cardiac dysfunction, and most oncologists typically attempt to limit doses above 300 mg/m2 if possible in patients of all age groups (27). Although no guidelines are currently set for an upper limit of anthracycline dose in older patients, it may be prudent to have lower limits in elderly patients identified at risk for cardiotoxicity (27). Several randomized control trials have evaluated different formulations of anthracyclines in order to minimize cardiotoxicity (2830). Continuous infusions of epirubicin and doxorubicin are associated with lower cardiotoxicity when compared with a bolus infusion (30). Similarly, studies have demonstrated lower cardiotoxicity with epirubicin when compared with doxorubicin. Liposomal doxorubicin has been shown to have decreased cardiotoxicity while maintaining efficacy in randomized control trials (2830). A single-arm phase II multicenter trial evaluated liposomal doxorubicin for treatment of large B-cell lymphoma in 80 patients aged 60 years or older (28). Cardiac events defined as Common Terminology Criteria for Adverse Events (CTCAE) Grade 3 or higher, were observed in only 4%, with an asymptomatic decline in left ventricular systolic function noted in 20% (28). While this study did not compare liposomal doxorubicin to standard doxorubicin, previous studies have documented significantly higher cardiac toxicity in older patients receiving standard doxorubicin-containing regimens. In the CAPRICE study of elderly or cardiotoxicity-prone patients with high-risk breast cancer, a regimen including liposomal doxorubicin, cyclophosphamide, and paclitaxel resulted in no significant decrease in left ventricular systolic function while achieving a pathological complete response rates comparable to regimens with standard doxorubicin (29).

Although not specifically in elderly patients, dexrazoxane infusion before anthracycline therapy has been demonstrated to mitigate cardiotoxicity (31, 32). A multicenter, phase III trial of a 164 patients with breast cancer showed a 39% versus 13% fewer cardiac events and 11% versus 1% incidence of heart failure in those treated with dexrazoxane versus no dexrazoxane; treatment efficacy was similar (33). Further studies are needed on the use of dexrazoxane for the prevention of cancer treatment-related cardiotoxicity in the elderly.

Trastuzumab

Trastuzumab is frequently used in the therapy for breast cancer and in a growing number of other cancers which overexpress human epidermal growth factor 2 or HER2 neu. In these tumors, treatment with trastuzumab greatly improves outcomes; trastuzumab, however, also has the potential to cause cardiotoxicity manifested as left ventricular systolic dysfunction. The incidence of cardiotoxicity has been estimated anywhere between 2% to as high as 26% (3436) and has been noted to be independent of the cumulative dose. The broad range in incidence speaks to the varied regimens used including monotherapy, in combination with taxanes, and in combination with anthracyclines, where it has the highest incidence (35). In a large population study of 9,535 patients with a median age of 71 years, those treated with trastuzumab had an overall increased incidence of cardiotoxicity when compared with those not treated with trastuzumab - 29.4% versus 18.6% (35). Furthermore, a competing risk regression model utilizing patients aged 66–70 years as the reference category, noted an incremental increase in risk of cardiac dysfunction with age, noting the highest risk in those 76 years of age or older and those with pre-existing comorbidities (35). Despite the concern for cardiac toxicity, trastuzumab continues to be frequently used due to its high efficacy in treatment of HER2-positive breast cancer (3739). Advancing age is a risk factor for developing cardiac dysfunction; however, shortening therapy is not recommended in this group due to the therapeutic benefit in breast cancer. Screening recommendations are discussed later in this article. In those with evidence of cardiac dysfunction following therapy, following thorough review of benefits and risk of treatment, decision to discontinue or hold treatment should be made by the primary oncologist in close collaboration with a cardiologist.

Fluoropyrimidines

Fluoropyrimidines are associated with the risk cardiotoxicity estimated between 4–7% and consisting of angina, hypertension, arrhythmia (19). The most common toxicity is anginal chest pain, but myocardial infarction, arrhythmias and heart failure have also been reported (19). One small study estimated a 5.5 times higher risk in patients with pre-existing cardiac comorbidities (40). A dose-dependent toxicity is observed, and patients with pre-existing cardiac risk factors are at increased risk (19, 40). Fluoropyrimidine-related cardiotoxicity can also recur with rechallenge. Older patients are at risk for this toxicity due to the increased prevalence of cardiovascular risk factors in this age group.

Tyrosine Kinase Inhibitors

Tyrosine kinase inhibitors (TKI) are small molecule inhibitors that are used in targeted therapy directed at specific cell signaling pathways in several cancers (41). Lapatinib, a TKI inhibits HER2 kinase which is in the same pathway as trastuzumab, but has a decreased risk of cardiotoxicity when compared with trastuzumab (34). In a meta-analysis of 44 clinical studies, 1.6% of 3,689 patients treated with lapatinib were noted to have cardiotoxicity defined as asymptomatic decline in left ventricular systolic function, which was ultimately reversible (19, 34, 42, 43). Other TKIs such as sunitinib and sorafenib have reported cardiotoxicities as high as 11% including an asymptomatic decrease in left ventricular systolic function and a small proportion with heart failure (4446). It is unclear whether potential cardiotoxicities are shared by the class of drugs or are specific to the molecule inhibited. Current evidence, including the studies above, have seldom outlined toxicities by age, and have primarily assessed younger patient groups. More clinical studies are needed to assess cardiotoxicity in older patients receiving these novel agents (10, 19).

Radiation Therapy

Radiation therapy, which is often used in Hodgkin lymphoma and breast cancer, may be followed by cardiotoxicity including radiation-induced coronary artery disease, valvular disorders, constrictive pericarditis, and cardiomyopathy (47). In a population-based, case-control study of 2,138 patients with breast cancer treated with left-sided or right-sided radiation therapy there was an increase in ischemic heart disease and major coronary events with a linear increase of major coronary events with the dose of radiation, within the first five years and at least 20 years after radiation therapy (48). Of note, the proportional increase in major coronary events was similar in women with and without cardiac risk factors at the time of therapy, but was overall higher in patients with pre-existing cardiac disease. Not surprisingly, there was also an increase in major coronary events in older patients especially those with pre-existing cardiac risk (48). In a large registry-based study using the National Cancer Institute’s Surveillance, Epidemiology, and End Results (SEER) program of 27,283 women, patients that received radiation therapy between the years 1973–1979 for left-sided compared with right-sided breast cancer had a statistically significant increase in ischemic heart disease (49). In subsequent years, 15-year mortality from ischemic heart disease decreased from 13% in patients treated in the mid-1970s to 5.8% in the late 1980s and there was no statistically significant difference between left- and right-sided breast cancers (49). The authors concluded these results were secondary to improved shielding and decreased radiation doses over the years (49). While this is encouraging, there are few data specific to the elderly patients on the risk-stratification for, and administration of radiation therapy.

Prevention

Multi-disciplinary Approach to Care of the Older Patients with Cancer

An individualized assessment of patient risk factors is critical in preventing cancer treatment-related complications. Often overlooked, chronologic age is not a reliable indicator of potential comorbidities or frailty (50). Undertreatment and poorer cancer-related outcomes are dangers of using absolute age as a definitive guide for treatment. Use of a comprehensive geriatric assessment (CGA) in the care of elderly patients has the potential to improve outcomes and aid in risk stratification. In recent years, many have advocated for a multi-disciplinary approach including oncologists, cardiologists, pharmacists, and geriatricians (20). Studies evaluating CGAs typically focus on factors such as functional status, cognitive abilities, emotional conditions, comorbid conditions, nutritional status, polypharmacy and environmental situation (51). Standard oncologic evaluations such as the Eastern Cooperative Oncology Group (ECOG) scale and the Karnofsky Performance Scale (KPS) have been noted to poorly predict treatment-related complications in elderly patients (52). In a study by Ghosn et al., an abridged geriatric assessment, incorporating the factors listed above, performed better than the Karnofsky Performance Scale (KPS) and the Physical Performance Test (PPT) in predicting mortality in elderly patients with cancer (52). In a recent consensus statement, the International Society of Geriatric Oncology has recommended incorporating the factors listed above into routine geriatric assessments, but did not find sufficient evidence to recommend any one particular CGA (53). Similarly, the National Comprehensive Cancer Center guidelines have a separate section on the older patient outlining the importance of risk factor assessments in the aging population, particularly related to cardiac toxicity (54). There is a need for evidence on the role of geriatric assessments for risk stratification and on effective ways to incorporate them into routine oncologic practice (55).

Identification of The Higher Risk Older Patient

Risk stratification remains difficult in elderly patients at risk for cardiotoxicity. A comprehensive evaluation of cardiovascular comorbidities such as hypertension, diabetes, dyslipidemia, and smoking need to be evaluated prior to start of therapy (27). Those patients receiving high-dose anthracyclines, high-dose radiation, and history of prior cardiac disease are at greatest risk for cardiac dysfunction are at highest risk for cardiotoxicity (10). Cancer-specific mortality is often higher in older patients, likely due to the impact of age-related factors (35, 56). Evidence from observational studies suggests possible undertreatment, as elderly patients are frequently offered lower doses of chemotherapy due to concern for cardiotoxicity (8, 57). Additionally, alternative treatments exist in some instances, data continue to show significantly better cancer-specific outcomes with first-line treatments such as anthracyclines and trastuzumab in some patient populations (9, 5861). This finding highlights the balance that is needed between minimizing cardiotoxic therapies and improving cancer-specific outcomes by identifying high risk elderly patients.

Pre-treatment evaluation of left ventricular systolic function is a standard part of many treatment protocols and current SIOG recommendations. However, the utility of this approach has been debated due to a low prevalence of asymptomatic left ventricular systolic dysfunction, with this strategy missing most patients that will ultimately have cardiotoxicity (62). Serum biomarkers may be useful in predicting cardiotoxicity and the role of baseline assessment of serum biomarkers prior to cancer treatment in predicting cardiotoxicity is being evaluated (63). Imaging biomarkers are also promising, and the role of pre-existing fibrosis on CMR to predict cardiotoxicity is currently being investigated (64).

A study utilizing the SEER database validated a risk score to help identify a those at risk for heart failure or cardiomyopathy in patients of all ages receiving trastuzumab (65). Patient were stratified into low, intermediate, and high risk; while incorporating readily accessible risk factors such as age, chemotherapy type, coronary artery disease, atrial fibrillation/flutter, diabetes mellitus, hypertension, and renal failure (65). Similar risk score assessments should be studied to aid in a more objective assessment of risk in older patients receiving other cardiotoxic therapies.

Medications

Small randomized controlled trials have evaluated the utility of using mainstays of heart failure therapy, renin angiotensin inhibitors, aldosterone inhibitors and beta-blockers, as preventive therapy for chemotherapy-related cardiotoxicity (31, 6671). None of these studies looked solely at older patients, but neither were they excluded. The OVERCOME trial looked at a small randomized control group of 90 patients (mean age 50 years) diagnosed with hematologic cancers and demonstrated prevention of left ventricular systolic dysfunction in those treated with prophylactic enalapril and carvedilol, compared with placebo (70). The PRADA trial, a randomized controlled trial comparing candesartan and metoprolol in patients treated for early breast cancer with anthracyclines and trastuzumab, indicated a protective effect of candesartan against left ventricular systolic dysfunction without a similar benefit for metoprolol (72). In the MANTICORE 101-Breast trial, perindopril and bisoprolol each attenuated trastuzumab-related declines in left ventricular systolic function, but did not prevent concurrent left ventricular remodeling, which was the primary outcome measure (73). In a double-blind, placebo-controlled study of spironolactone given simultaneously with anthracycline therapy, spironolactone had a protective effect in the development of left ventricular systolic dysfunction (71). Despite the favorable evidence presented above, routine prophylactic use of these therapies for cardiotoxicity is limited by the lack of consensus, as noted in two meta-analysis with differing conclusions on the benefits of these preventive strategies (32, 74). This is likely because of the heterogeneous nature of the studies included in the meta-analyses and the small numbers in each individual trial. There are several ongoing clinical trials with no upper limit for age evaluating the utility of beta blockers, ace inhibitors, and statins in prevention of chemotherapy induced cardiotoxicity (7579).

Although, prophylactic use of these medications has limited evidence currently, treatment of risk factors such as diabetes, hypertension, hyperlipidemia and smoking are important considerations (10). Aggressive treatment of hypertension utilizing beta-blockers, ACE-inhibitors, and ARBs should be considered in patients receiving cardiotoxic therapies.

Exercise

Aerobic exercise has evidence suggesting its use for anthracycline-induced cardiac injury as a preventive and treatment strategy (80). The evidence supporting exercise is well established in pre-clinical animal studies, noting aerobic exercise as an effective prophylactic method in prevention of left ventricular systolic dysfunction (80). Studies in humans have documented improvements of psycho-social parameters in patients receiving cancer treatment (81). Studies investigating the effects of exercise training during breast cancer treatment on prevention of cardiotoxicity are limited. The only study on this topic, by Haykowsky, et al., showed that aerobic training did not prevent trastuzumab-related left ventricular remodeling or decline in left ventricular ejection fraction after four months of treatment in women with HER2-positive breast cancer (82).

Surveillance

Imaging

Current recommendations for imaging surveillance include monitoring of left ventricular systolic function during treatment with both anthracyclines and trastuzumab. Current National Comprehensive Cancer Network guidelines suggest cardiac monitoring at baseline, three, six, and nine months after initiating therapy for trastuzumab therapy, upon completion of treatment, and every six months for two years following completion of treatment (83). Additionally, the current International Society of Geriatric Oncology (SIOG) guidelines recommended regular monitoring of left ventricular systolic function with echocardiography or multiple gated acquisition scanning (MUGA) every two to three cycles of anthracyclines in patients 70 years of age or older (19, 27). Further attention is needed in those who have an LVEF drop of 10% even if it remains within the normal range (27). Early identification of left ventricular dysfunction is vital to stop worsening and to identify patients that may benefit from cardioprotective treatments. In a prospective study of 2625 patients with no upper age limit and median age of 51, observed a dose-dependent cardiotoxicity in patients receiving anthracyclines most often within the first year of treatment, along with the observation that close monitoring allowed for early detection and treatment of cardiotoxicity (23). The authors hypothesized, early identification of left ventricular dysfunction, in a potentially reversible phase of injury, allowed for early start of heart failure therapies and recovery of baseline LVEF (23).

Novel echocardiographic techniques are gaining traction as possible early predictors of cardiotoxicity. In cross-sectional studies, tissue Doppler imaging showed promise as an early predictor of adverse cardiac outcomes prior to onset of left ventricular dysfunction (72, 84). Myocardial deformation (strain) has garnered increasing interest as an early predictor of left ventricular systolic dysfunction. In an expert consensus statement, Plana et al., advocate for global longitudinal strain (GLS) as the optimal parameter for early detection of sub-clinical left ventricular dysfunction (85). Cardiac fibrosis identified using CMR also holds promise for the early detection of cardiotoxicity and warrants further study (86). These novel imaging techniques are gaining traction and data are warranted to help in the early or subclinical identification of cardiotoxicity in older patients. Currently, routine screening for left ventricular systolic function remains the standard of care with echocardiography and CMR as the imaging modalities of choice. MUGA is less preferable as the imaging modality of choice for surveillance due to its use of ionizing radiation in a patient population that requires serial studies. With 12 months of adjuvant trastuzumab as the standard of care, this translates into a minimum of nine studies, associated with a significant dose of ionizing radiation and the risk of radiation-related secondary cancers. A recent publication highlighted this issue through the case of a patient with multiple myeloma who received 17 MUGAs, corresponding to an effective radiation dose of 113 mSv, over a span of three years (87).

Serum Biomarkers

High sensitivity troponin has been evaluated as a possible early predictor for cardiotoxicity during cancer treatment (8891). A study of 703 patients treated with high-dose anthracyclines, evaluated the prognostic value of serial high-sensitivity cardiac troponin at preset time points on start, 12, 24, 36, and 72 hours, and one month after each dose of high-dose anthracycline therapy (91). The incidence of cardiovascular events, defined as heart failure, asymptomatic decline of left ventricular systolic function, and arrhythmias, was noted at 87% in the group with both early and late troponin elevation, 37% with only early elevated troponin and 1% with negative troponin values (91). A similar study in patients receiving trastuzumab with troponin elevation identified those at risk for major adverse cardiac events and irreversible cardiotoxicity (90, 92). The use of NT-proBNP has been limited to clinically appropriate settings such as patients presenting with signs and symptoms of heart failure (93). The evidence on the use of biomarkers in predicting patients at risk for cardiotoxicity is limited and more studies are needed before recommendations are made regarding routine use of biomarkers for surveillance including in older adults.

Treatment of Cardiotoxicity

Heart failure and cardiomyopathy from cancer treatments should be treated according to the standard ACC/AHA guidelines for the evaluation and management of heart failure (94). This generally includes the traditional mainstays of heart failure treatment: beta blockers, ACE-inhibitors, angiotensin II receptor blockers, aldosterone antagonists, and diuretics. Exercise training and cardiac rehabilitation likely have similar efficacy in this group as well.

Summary

In conclusion, significant evidence suggests that older patients are more susceptible to cancer-treatment related cardiotoxicity compared to their younger counterparts. Barriers such as pre-existing comorbidities, the potential for undertreatment and under-representation in clinical trials remain. There is a need for close collaboration between clinicians in a multi-disciplinary approach. Clinical studies are urgently needed on the prevention, early detection, and treatment of cardiotoxicity in older cancer survivors.

Acknowledgments

This article was supported by National Institutes of Health grant K23HL132011-01 and the University of Minnesota Clinical and Translational Science Institute KL2 Scholars Career Development Program Award (National Institutes of Health grant KL2TR000113-05), both to Dr. Chetan Shenoy.

Footnotes

Disclosures and Conflict of Interest Statements

The authors have no conflicts of interest to disclose.

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.

References

  • 1.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin. 2016;66(1):7–30. doi: 10.3322/caac.21332. [DOI] [PubMed] [Google Scholar]
  • 2.Parry C, Kent EE, Mariotto AB, Alfano CM, Rowland JH. Cancer survivors: a booming population. Cancer Epidemiol Biomarkers Prev. 2011;20(10):1996–2005. doi: 10.1158/1055-9965.EPI-11-0729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.de Moor JS, Mariotto AB, Parry C, Alfano CM, Padgett L, Kent EE, et al. Cancer survivors in the United States: prevalence across the survivorship trajectory and implications for care. Cancer Epidemiol Biomarkers Prev. 2013;22(4):561–70. doi: 10.1158/1055-9965.EPI-12-1356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Weir HK, Anderson RN, Coleman King SM, Soman A, Thompson TD, Hong Y, et al. Heart Disease and Cancer Deaths - Trends and Projections in the United States, 1969–2020. Prev Chronic Dis. 2016;13:E157. doi: 10.5888/pcd13.160211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Rich MW, Chyun DA, Skolnick AH, Alexander KP, Forman DE, Kitzman DW, et al. Knowledge Gaps in Cardiovascular Care of the Older Adult Population: A Scientific Statement From the American Heart Association, American College of Cardiology, and American Geriatrics Society. Circulation. 2016;133(21):2103–22. doi: 10.1161/CIR.0000000000000380. [DOI] [PubMed] [Google Scholar]
  • 6.Hutchins LF, Unger JM, Crowley JJ, Coltman CA, Jr, Albain KS. Underrepresentation of patients 65 years of age or older in cancer-treatment trials. N Engl J Med. 1999;341(27):2061–7. doi: 10.1056/NEJM199912303412706. [DOI] [PubMed] [Google Scholar]
  • 7.Javid SH, Unger JM, Gralow JR, Moinpour CM, Wozniak AJ, Goodwin JW, et al. A prospective analysis of the influence of older age on physician and patient decision-making when considering enrollment in breast cancer clinical trials (SWOG S0316) Oncologist. 2012;17(9):1180–90. doi: 10.1634/theoncologist.2011-0384. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Fourcadier E, Tretarre B, Gras-Aygon C, Ecarnot F, Daures JP, Bessaoud F. Under-treatment of elderly patients with ovarian cancer: a population based study. BMC Cancer. 2015;15:937. doi: 10.1186/s12885-015-1947-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Muss HB, Berry DA, Cirrincione CT, Theodoulou M, Mauer AM, Kornblith AB, et al. Adjuvant chemotherapy in older women with early-stage breast cancer. N Engl J Med. 2009;360(20):2055–65. doi: 10.1056/NEJMoa0810266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Armenian SH, Lacchetti C, Barac A, Carver J, Constine LS, Denduluri N, et al. Prevention and Monitoring of Cardiac Dysfunction in Survivors of Adult Cancers: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2016 JCO2016705400. [Google Scholar]
  • 11.Centers for Disease Control and Prevention; 2013. https://www.cdc.gov/aging/pdf/state-aging-health-in-america-2013.pdf. [Google Scholar]
  • 12.Tawfik B, Pardee TS, Isom S, Sliesoraitis S, Winter A, Lawrence J, et al. Comorbidity, age, and mortality among adults treated intensively for acute myeloid leukemia (AML) J Geriatr Oncol. 2016;7(1):24–31. doi: 10.1016/j.jgo.2015.10.182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Shenoy C, Klem I, Crowley AL, Patel MR, Winchester MA, Owusu C, et al. Cardiovascular complications of breast cancer therapy in older adults. Oncologist. 2011;16(8):1138–43. doi: 10.1634/theoncologist.2010-0348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Turner JP, Shakib S, Singhal N, Hogan-Doran J, Prowse R, Johns S, et al. Prevalence and factors associated with polypharmacy in older people with cancer. Support Care Cancer. 2014;22(7):1727–34. doi: 10.1007/s00520-014-2171-x. [DOI] [PubMed] [Google Scholar]
  • 15.Lopez-Martin C, Garrido Siles M, Alcaide-Garcia J, Faus Felipe V. Role of clinical pharmacists to prevent drug interactions in cancer outpatients: a single-centre experience. Int J Clin Pharm. 2014;36(6):1251–9. doi: 10.1007/s11096-014-0029-4. [DOI] [PubMed] [Google Scholar]
  • 16.Saarelainen LK, Turner JP, Shakib S, Singhal N, Hogan-Doran J, Prowse R, et al. Potentially inappropriate medication use in older people with cancer: prevalence and correlates. J Geriatr Oncol. 2014;5(4):439–46. doi: 10.1016/j.jgo.2014.07.001. [DOI] [PubMed] [Google Scholar]
  • 17.Yeoh TT, Si P, Chew L. The impact of medication therapy management in older oncology patients. Support Care Cancer. 2013;21(5):1287–93. doi: 10.1007/s00520-012-1661-y. [DOI] [PubMed] [Google Scholar]
  • 18.Yeoh TT, Tay XY, Si P, Chew L. Drug-related problems in elderly patients with cancer receiving outpatient chemotherapy. J Geriatr Oncol. 2015;6(4):280–7. doi: 10.1016/j.jgo.2015.05.001. [DOI] [PubMed] [Google Scholar]
  • 19.Accordino MK, Neugut AI, Hershman DL. Cardiac effects of anticancer therapy in the elderly. J Clin Oncol. 2014;32(24):2654–61. doi: 10.1200/JCO.2013.55.0459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Nguyen K-L, Alrezk R, Mansourian PG, Naeim A, Rettig MB, Lee CC. The Crossroads of Geriatric Cardiology and Cardio-Oncology. Current Geriatrics Reports. 2015;4(4):327–37. doi: 10.1007/s13670-015-0147-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Smith LA, Cornelius Vr, Plummer CJ, Levitt G, Verrill M, Canney P, Jones A, et al. Cardiotoxicity of anthracycline agents for the treatment of cancer: systematic review and meta-analysis of randomised controlled trials. doi: 10.1186/1471-2407-10-337. (1471–2407 (Electronic)) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Fisher RI, Gaynor ER, Dahlberg S, Oken MM, Grogan TM, Mize EM, et al. Comparison of a standard regimen (CHOP) with three intensive chemotherapy regimens for advanced non-Hodgkin’s lymphoma. N Engl J Med. 1993;328(14):1002–6. doi: 10.1056/NEJM199304083281404. [DOI] [PubMed] [Google Scholar]
  • 23.Cardinale D, Colombo A, Bacchiani G, Tedeschi I, Meroni CA, Veglia F, et al. Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy. Circulation. 2015;131(22):1981–8. doi: 10.1161/CIRCULATIONAHA.114.013777. [DOI] [PubMed] [Google Scholar]
  • 24.Singal PK, Iliskovic N. Doxorubicin-induced cardiomyopathy. N Engl J Med. 1998;339(13):900–5. doi: 10.1056/NEJM199809243391307. [DOI] [PubMed] [Google Scholar]
  • 25.Von Hoff DD, Layard MW, Basa P, Davis HL, Jr, Von Hoff AL, Rozencweig M, et al. Risk factors for doxorubicin-induced congestive heart failure. Ann Intern Med. 1979;91(5):710–7. doi: 10.7326/0003-4819-91-5-710. [DOI] [PubMed] [Google Scholar]
  • 26.Neilan TG, Coelho-Filho OR, Pena-Herrera D, Shah RV, Jerosch-Herold M, Francis SA, et al. Left ventricular mass in patients with a cardiomyopathy after treatment with anthracyclines. Am J Cardiol. 2012;110(11):1679–86. doi: 10.1016/j.amjcard.2012.07.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Aapro M, Bernard-Marty C, Brain EGC, Batist G, Erdkamp F, Krzemieniecki K, Leonard R, et al. Anthracycline cardiotoxicity in the elderly cancer patient: a SIOG expert position paper. doi: 10.1093/annonc/mdq609. (1569–8041 (Electronic)) [DOI] [PubMed] [Google Scholar]
  • 28.Oki Y, Ewer MS, Lenihan DJ, Fisch MJ, Hagemeister FB, Fanale M, et al. Pegylated liposomal doxorubicin replacing conventional doxorubicin in standard R-CHOP chemotherapy for elderly patients with diffuse large B-cell lymphoma: an open label, single arm, phase II trial. Clin Lymphoma Myeloma Leuk. 2015;15(3):152–8. doi: 10.1016/j.clml.2014.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Gil-Gil MJ, Bellet M, Morales S, Ojeda B, Manso L, Mesia C, et al. Pegylated liposomal doxorubicin plus cyclophosphamide followed by paclitaxel as primary chemotherapy in elderly or cardiotoxicity-prone patients with high-risk breast cancer: results of the phase II CAPRICE study. Breast Cancer Res Treat. 2015;151(3):597–606. doi: 10.1007/s10549-015-3415-2. [DOI] [PubMed] [Google Scholar]
  • 30.Hunault-Berger M, Leguay T, Thomas X, Legrand O, Huguet F, Bonmati C, et al. A randomized study of pegylated liposomal doxorubicin versus continuous-infusion doxorubicin in elderly patients with acute lymphoblastic leukemia: the GRAALL-SA1 study. Haematologica. 2011;96(2):245–52. doi: 10.3324/haematol.2010.027862. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.van Dalen EC, Caron HN, Dickinson HO, Kremer LC. Cardioprotective interventions for cancer patients receiving anthracyclines. Cochrane Database Syst Rev. 2011;(6):CD003917. doi: 10.1002/14651858.CD003917.pub4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Abdel-Qadir H, Ong G, Fazelzad R, Amir E, Lee DS, Thavendiranathan P, et al. Interventions for preventing cardiomyopathy due to anthracyclines: a Bayesian network meta-analysis. Ann Oncol. 2016 doi: 10.1093/annonc/mdw671. [DOI] [PubMed] [Google Scholar]
  • 33.Marty M, Espie M, Llombart A, Monnier A, Rapoport BL, Stahalova V, 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(4):614–22. doi: 10.1093/annonc/mdj134. [DOI] [PubMed] [Google Scholar]
  • 34.Azim H, Azim HA, Jr, Escudier B. Trastuzumab versus lapatinib: the cardiac side of the story. Cancer Treat Rev. 2009;35(7):633–8. doi: 10.1016/j.ctrv.2009.06.007. [DOI] [PubMed] [Google Scholar]
  • 35.Chavez-MacGregor M, Zhang N, Buchholz TA, Zhang Y, Niu J, Elting L, et al. Trastuzumab-related cardiotoxicity among older patients with breast cancer. J Clin Oncol. 2013;31(33):4222–8. doi: 10.1200/JCO.2013.48.7884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Leung HW, Chan AL. Trastuzumab-induced cardiotoxicity in elderly women with HER-2-positive breast cancer: a meta-analysis of real-world data. Expert Opin Drug Saf. 2015;14(11):1661–71. doi: 10.1517/14740338.2015.1089231. [DOI] [PubMed] [Google Scholar]
  • 37.Advani PP, Ballman KV, Dockter TJ, Colon-Otero G, Perez EA. Long-Term Cardiac Safety Analysis of NCCTG N9831 (Alliance) Adjuvant Trastuzumab Trial. J Clin Oncol. 2016;34(6):581–7. doi: 10.1200/JCO.2015.61.8413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Mavroudis D, Saloustros E, Malamos N, Kakolyris S, Boukovinas I, Papakotoulas P, et al. Six versus 12 months of adjuvant trastuzumab in combination with dose-dense chemotherapy for women with HER2-positive breast cancer: a multicenter randomized study by the Hellenic Oncology Research Group (HORG) Ann Oncol. 2015;26(7):1333–40. doi: 10.1093/annonc/mdv213. [DOI] [PubMed] [Google Scholar]
  • 39.Pivot X, Romieu G, Debled M, Pierga JY, Kerbrat P, Bachelot T, et al. 6 months versus 12 months of adjuvant trastuzumab for patients with HER2-positive early breast cancer (PHARE): a randomised phase 3 trial. Lancet Oncol. 2013;14(8):741–8. doi: 10.1016/S1470-2045(13)70225-0. [DOI] [PubMed] [Google Scholar]
  • 40.Polk A, Shahmarvand N, Vistisen K, Vaage-Nilsen M, Larsen FO, Schou M, et al. Incidence and risk factors for capecitabine-induced symptomatic cardiotoxicity: a retrospective study of 452 consecutive patients with metastatic breast cancer. BMJ Open. 2016;6(10):e012798. doi: 10.1136/bmjopen-2016-012798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Arora A, Scholar EM. Role of tyrosine kinase inhibitors in cancer therapy. J Pharmacol Exp Ther. 2005;315(3):971–9. doi: 10.1124/jpet.105.084145. [DOI] [PubMed] [Google Scholar]
  • 42.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(6):679–86. doi: 10.4065/83.6.679. [DOI] [PubMed] [Google Scholar]
  • 43.Sendur MA, Aksoy S, Altundag K. Cardiotoxicity of novel HER2-targeted therapies. Curr Med Res Opin. 2013;29(8):1015–24. doi: 10.1185/03007995.2013.807232. [DOI] [PubMed] [Google Scholar]
  • 44.Sternberg CN, Calabro F, Bracarda S, Carteni G, Lo Re G, Ruggeri EM, et al. Safety and efficacy of sunitinib in patients from Italy with metastatic renal cell carcinoma: final results from an expanded-access trial. Oncology. 2015;88(5):273–80. doi: 10.1159/000369256. [DOI] [PubMed] [Google Scholar]
  • 45.Rosa GM, Gigli L, Tagliasacchi MI, Di Iorio C, Carbone F, Nencioni A, et al. Update on cardiotoxicity of anti-cancer treatments. Eur J Clin Invest. 2016;46(3):264–84. doi: 10.1111/eci.12589. [DOI] [PubMed] [Google Scholar]
  • 46.Chu TF, Rupnick MA, Kerkela R, Dallabrida SM, Zurakowski D, Nguyen L, et al. Cardiotoxicity associated with tyrosine kinase inhibitor sunitinib. Lancet. 2007;370(9604):2011–9. doi: 10.1016/S0140-6736(07)61865-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Cuomo JR, Sharma GK, Conger PD, Weintraub NL. Novel concepts in radiation-induced cardiovascular disease. World J Cardiol. 2016;8(9):504–19. doi: 10.4330/wjc.v8.i9.504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Darby SC, Ewertz M, McGale P, Bennet AM, Blom-Goldman U, Bronnum D, et al. Risk of ischemic heart disease in women after radiotherapy for breast cancer. N Engl J Med. 2013;368(11):987–98. doi: 10.1056/NEJMoa1209825. [DOI] [PubMed] [Google Scholar]
  • 49.Giordano SH, Kuo YF, Freeman JL, Buchholz TA, Hortobagyi GN, Goodwin JS. Risk of cardiac death after adjuvant radiotherapy for breast cancer. J Natl Cancer Inst. 2005;97(6):419–24. doi: 10.1093/jnci/dji067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Basso U, Tonti S, Bassi C, Brunello A, Pasetto LM, Scaglione D, et al. Management of Frail and Not-Frail elderly cancer patients in a hospital-based geriatric oncology program. Crit Rev Oncol Hematol. 2008;66(2):163–70. doi: 10.1016/j.critrevonc.2007.12.006. [DOI] [PubMed] [Google Scholar]
  • 51.Terret C, Zulian GB, Naiem A, Albrand G. Multidisciplinary approach to the geriatric oncology patient. J Clin Oncol. 2007;25(14):1876–81. doi: 10.1200/JCO.2006.10.3291. [DOI] [PubMed] [Google Scholar]
  • 52.Ghosn M, Ibrahim T, El Rassy E, Nassani N, Ghanem S, Assi T. Abridged geriatric assessment is a better predictor of overall survival than the Karnofski Performance Scale and Physical Performance Test in elderly cancer patients. J Geriatr Oncol. 2016 doi: 10.1016/j.jgo.2016.11.002. [DOI] [PubMed] [Google Scholar]
  • 53.Wildiers H, Heeren P, Puts M, Topinkova E, Janssen-Heijnen ML, Extermann M, et al. International Society of Geriatric Oncology consensus on geriatric assessment in older patients with cancer. J Clin Oncol. 2014;32(24):2595–603. doi: 10.1200/JCO.2013.54.8347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.National Comprehensive Cancer Network 2016 [Version 2.2016. [Available from: NCCN.org.
  • 55.Angarita FA, Chesney T, Elser C, Mulligan AM, McCready DR, Escallon J. Treatment patterns of elderly breast cancer patients at two Canadian cancer centres. Eur J Surg Oncol. 2015;41(5):625–34. doi: 10.1016/j.ejso.2015.01.028. [DOI] [PubMed] [Google Scholar]
  • 56.Denegri A, Moccetti T, Moccetti M, Spallarossa P, Brunelli C, Ameri P. Cardiac toxicity of trastuzumab in elderly patients with breast cancer. J Geriatr Cardiol. 2016;13(4):355–63. doi: 10.11909/j.issn.1671-5411.2016.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Morrison VA, Hamlin P, Soubeyran P, Stauder R, Wadhwa P, Aapro M, et al. Approach to therapy of diffuse large B-cell lymphoma in the elderly: the International Society of Geriatric Oncology (SIOG) expert position commentary. Ann Oncol. 2015;26(6):1058–68. doi: 10.1093/annonc/mdv018. [DOI] [PubMed] [Google Scholar]
  • 58.Peters FP, Fickers MM, Erdkamp FL, Wals J, Wils JA, Schouten HC. The effect of optimal treatment on elderly patients with aggressive non-Hodgkin’s lymphoma: more patients treated with unaffected response rates. Ann Hematol. 2001;80(7):406–10. doi: 10.1007/s002770100315. [DOI] [PubMed] [Google Scholar]
  • 59.Liang R, Todd D, Chan TK, Chiu E, Lie A, Ho F. COPP chemotherapy for elderly patients with intermediate and high grade non-Hodgkin’s lymphoma. Hematol Oncol. 1993;11(1):43–50. doi: 10.1002/hon.2900110106. [DOI] [PubMed] [Google Scholar]
  • 60.Sonneveld P, Huijgens PC, Hagenbeek A. Dose reduction is not recommended for elderly patients undergoing chemotherapy for non-Hodgkin lymphoma. Ned Tijdschr Geneeskd. 1999;143(8):418–9. [PubMed] [Google Scholar]
  • 61.Sonneveld P, de Ridder M, van der Lelie H, Nieuwenhuis K, Schouten H, Mulder A, et al. Comparison of doxorubicin and mitoxantrone in the treatment of elderly patients with advanced diffuse non-Hodgkin’s lymphoma using CHOP versus CNOP chemotherapy. J Clin Oncol. 1995;13(10):2530–9. doi: 10.1200/JCO.1995.13.10.2530. [DOI] [PubMed] [Google Scholar]
  • 62.Steuter J, Bociek R, Loberiza F, Mathers D, Armitage J, Vose J, et al. Utility of prechemotherapy evaluation of left ventricular function for patients with lymphoma. Clin Lymphoma Myeloma Leuk. 2015;15(1):29–34. doi: 10.1016/j.clml.2014.06.026. [DOI] [PubMed] [Google Scholar]
  • 63.Yu AF, Ky B. Roadmap for biomarkers of cancer therapy cardiotoxicity. Heart. 2016;102(6):425–30. doi: 10.1136/heartjnl-2015-307894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Cardiac Fibrosis and Risk Prediction in Cancer Treatment-Related Cardiotoxicity. 2017 Jan 19; Available from: https://www.clinicaltrials.gov/ct2/show/NCT02798679?term=NCT02798679&rank=1.
  • 65.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(1):e000472. doi: 10.1161/JAHA.113.000472. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Cardinale D, Colombo A, Sandri MT, Lamantia G, Colombo N, Civelli M, et al. Prevention of high-dose chemotherapy-induced cardiotoxicity in high-risk patients by angiotensin-converting enzyme inhibition. Circulation. 2006;114(23):2474–81. doi: 10.1161/CIRCULATIONAHA.106.635144. [DOI] [PubMed] [Google Scholar]
  • 67.Tashakori Beheshti A, Mostafavi Toroghi H, Hosseini G, Zarifian A, Homaei Shandiz F, Fazlinezhad A. Carvedilol Administration Can Prevent Doxorubicin-Induced Cardiotoxicity: A Double-Blind Randomized Trial. Cardiology. 2016;134(1):47–53. doi: 10.1159/000442722. [DOI] [PubMed] [Google Scholar]
  • 68.Di Lisi D, Leggio G, Vitale G, Arrotti S, Iacona R, Inciardi RM, et al. Chemotherapy cardiotoxicity: cardioprotective drugs and early identification of cardiac dysfunction. J Cardiovasc Med (Hagerstown) 2016;17(4):270–5. doi: 10.2459/JCM.0000000000000232. [DOI] [PubMed] [Google Scholar]
  • 69.Elitok A, Oz F, Cizgici AY, Kilic L, Ciftci R, Sen F, et al. Effect of carvedilol on silent anthracycline-induced cardiotoxicity assessed by strain imaging: A prospective randomized controlled study with six-month follow-up. Cardiol J. 2014;21(5):509–15. doi: 10.5603/CJ.a2013.0150. [DOI] [PubMed] [Google Scholar]
  • 70.Bosch X, Rovira M, Sitges M, Domenech A, Ortiz-Perez JT, de Caralt TM, et al. Enalapril and carvedilol for preventing chemotherapy-induced left ventricular systolic dysfunction in patients with malignant hemopathies: the OVERCOME trial (preventiOn of left Ventricular dysfunction with Enalapril and caRvedilol in patients submitted to intensive ChemOtherapy for the treatment of Malignant hEmopathies) J Am Coll Cardiol. 2013;61(23):2355–62. doi: 10.1016/j.jacc.2013.02.072. [DOI] [PubMed] [Google Scholar]
  • 71.Akpek M, Ozdogru I, Sahin O, Inanc M, Dogan A, Yazici C, et al. Protective effects of spironolactone against anthracycline-induced cardiomyopathy. Eur J Heart Fail. 2015;17(1):81–9. doi: 10.1002/ejhf.196. [DOI] [PubMed] [Google Scholar]
  • 72.Gulati G, Zhang KW, Scherrer-Crosbie M, Ky B. Cancer and cardiovascular disease: the use of novel echocardiography measures to predict subsequent cardiotoxicity in breast cancer treated with anthracyclines and trastuzumab. Curr Heart Fail Rep. 2014;11(4):366–73. doi: 10.1007/s11897-014-0214-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Pituskin E, Mackey JR, Koshman S, Jassal D, Pitz M, Haykowsky MJ, et al. Multidisciplinary Approach to Novel Therapies in Cardio-Oncology Research (MANTICORE 101-Breast): A Randomized Trial for the Prevention of Trastuzumab-Associated Cardiotoxicity. J Clin Oncol. 2016 doi: 10.1200/JCO.2016.68.7830. JCO2016687830. [DOI] [PubMed] [Google Scholar]
  • 74.Kalam K, Marwick TH. Role of cardioprotective therapy for prevention of cardiotoxicity with chemotherapy: a systematic review and meta-analysis. Eur J Cancer. 2013;49(13):2900–9. doi: 10.1016/j.ejca.2013.04.030. [DOI] [PubMed] [Google Scholar]
  • 75.SAFE trial. 2017 Jan 19; Available from: https://www.clinicaltrials.gov/ct2/show/NCT02236806?term=prevention+of+chemotherapy+cardiotoxicity&rank=7.
  • 76.CECCY trial. 2017 Jan 19; Available from: https://www.clinicaltrials.gov/ct2/show/NCT01724450?term=prevention+of+chemotherapy+cardiotoxicity&rank=2.
  • 77.PREVENT trial. 2017 Jan 19; Available from: https://www.clinicaltrials.gov/ct2/results?term=NCT01988571&Search=Search.
  • 78.Study of the Effect of Eplerenone on Heart Function in Women Receiving Anthracycline Chemotherapy for Breast Cancer. 2017 Jan 19; Available from: https://www.clinicaltrials.gov/ct2/show/NCT01708798?term=NCT01708798&rank=1.
  • 79.ICOS-ONE. 2017 Jan 19; Available from: https://www.clinicaltrials.gov/ct2/show/NCT01968200?term=NCT01968200&rank=1.
  • 80.Scott JM, Khakoo A, Mackey JR, Haykowsky MJ, Douglas PS, Jones LW. Modulation of anthracycline-induced cardiotoxicity by aerobic exercise in breast cancer: current evidence and underlying mechanisms. Circulation. 2011;124(5):642–50. doi: 10.1161/CIRCULATIONAHA.111.021774. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Sturgeon KM, Ky B, Libonati JR, Schmitz KH. The effects of exercise on cardiovascular outcomes before, during, and after treatment for breast cancer. Breast Cancer Res Treat. 2014;143(2):219–26. doi: 10.1007/s10549-013-2808-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Haykowsky MJ, Mackey JR, Thompson RB, Jones LW, Paterson DI. Adjuvant trastuzumab induces ventricular remodeling despite aerobic exercise training. Clin Cancer Res. 2009;15(15):4963–7. doi: 10.1158/1078-0432.CCR-09-0628. [DOI] [PubMed] [Google Scholar]
  • 83.Carlson RW, Brown E, Burstein HJ, Gradishar WJ, Hudis CA, Loprinzi C, et al. NCCN Task Force Report: Adjuvant Therapy for Breast Cancer. J Natl Compr Canc Netw. 2006;4(Suppl 1):S1–26. [PubMed] [Google Scholar]
  • 84.Ho E, Brown A, Barrett P, Morgan RB, King G, Kennedy MJ, 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(9):701–7. doi: 10.1136/hrt.2009.173997. [DOI] [PubMed] [Google Scholar]
  • 85.Plana JC, Galderisi M, Barac A, Ewer MS, Ky B, Scherrer-Crosbie M, 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(10):1063–93. doi: 10.1093/ehjci/jeu192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Melendez GC, Jordan JH, D’Agostino RB, Jr, Vasu S, Hamilton CA, Hundley WG. Progressive 3-Month Increase in LV Myocardial ECV After Anthracycline-Based Chemotherapy. JACC Cardiovasc Imaging. 2016 doi: 10.1016/j.jcmg.2016.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Bhatti S, Hendel RC, Lopez-Mattei J, Schwartz RG, Raff G, Einstein AJ. Frequent MUGA testing in a myeloma patient: A case-based ethics discussion. J Nucl Cardiol. 2016 doi: 10.1007/s12350-016-0540-1. [DOI] [PubMed] [Google Scholar]
  • 88.Pistillucci G, Ciorra AA, Sciacca V, Raponi M, Rossi R, Veltri E. Troponin I and B-type Natriuretic Peptide (BNP) as biomarkers for the prediction of cardiotoxicity in patients with breast cancer treated with adjuvant anthracyclines and trastuzumab. Clin Ter. 2015;166(1):e67–71. doi: 10.7417/CT.2015.1812. [DOI] [PubMed] [Google Scholar]
  • 89.Lipshultz SE, Miller TL, Scully RE, Lipsitz SR, Rifai N, Silverman LB, 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(10):1042–9. doi: 10.1200/JCO.2010.30.3404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Cardinale D, Sandri MT. Role of biomarkers in chemotherapy-induced cardiotoxicity. Prog Cardiovasc Dis. 2010;53(2):121–9. doi: 10.1016/j.pcad.2010.04.002. [DOI] [PubMed] [Google Scholar]
  • 91.Cardinale D, Sandri MT, Colombo A, Colombo N, Boeri M, Lamantia G, et al. Prognostic value of troponin I in cardiac risk stratification of cancer patients undergoing high-dose chemotherapy. Circulation. 2004;109(22):2749–54. doi: 10.1161/01.CIR.0000130926.51766.CC. [DOI] [PubMed] [Google Scholar]
  • 92.Cardinale D, Colombo A, Torrisi R, Sandri MT, Civelli M, Salvatici M, et al. Trastuzumab-induced cardiotoxicity: clinical and prognostic implications of troponin I evaluation. J Clin Oncol. 2010;28(25):3910–6. doi: 10.1200/JCO.2009.27.3615. [DOI] [PubMed] [Google Scholar]
  • 93.Skovgaard D, Hasbak P, Kjaer A. BNP predicts chemotherapy-related cardiotoxicity and death: comparison with gated equilibrium radionuclide ventriculography. PLoS One. 2014;9(5):e96736. doi: 10.1371/journal.pone.0096736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Hunt SA, Baker DW, Chin MH, Cinquegrani MP, Feldman AM, Francis GS, et al. ACC/AHA guidelines for the evaluation and management of chronic heart failure in the adult: executive summary. J Heart Lung Transplant. 2002;21(2):189–203. doi: 10.1016/s1053-2498(01)00776-8. [DOI] [PubMed] [Google Scholar]

RESOURCES