Breast Cancer CV Toxicities

Introduction

In women in the United States, breast cancer is the most common cancer and the second leading cause of cancer-related death.1 Improved therapies have increased cure rates and helped patients live longer with incurable disease, such that the 5-year relative survival rate is now 91 percent.1 However, some of these treatments (e.g., radiation and certain chemotherapies) can cause serious side effects, including those that affect the cardiovascular (CV) system.2,3 Acute manifestations of cardiac damage from radiation or chemotherapy include pericarditis and pericardial effusion, and late and long-term manifestations include congestive heart failure (HF), coronary artery disease (CAD), cardiomyopathy, valvular disease, myocardial fibrosis, and conduction abnormalities (sometimes manifesting years after the causal oncologic therapy).2-4 These complications can cause significant morbidity or mortality. Table 1 contains information about the presumed pathophysiologic mechanisms that underlie therapy-induced cardiovascular disease (CVD) in survivors of breast cancer.

Table 1: Pathophysiology of CV Toxicities Observed With Breast Cancer Treatment5-8

Treatment

Pathophysiology of Cardiac Toxicity

Radiation

Free radicals and oxidative stress due to irradiation lead to DNA damage, transcriptional changes, and pro-inflammatory state. This damages the micro-and macro-vasculature and leads to fibrin deposition and fibrosis. This facilitates the development of atherosclerotic disease. Fibrosis can affect the valves and conduction system as well.

Trastuzumab

Not well understood; likely related to the effects of HER2 signaling on cardiac development, myocyte survival, and protection from cardiotoxins. HER2-blockade interferes with the ligand-binding-induced cardioprotective pathways that are obligatory to recover from cardiac injury pathways. It is thought that trastuzumab leads to loss of contractility due to cellular stunning rather than myocyte death.

Anthracycline

Uncertain mechanism. One hypothesis is that the drug-DNA complex triggers cell death. Anthracyclines target topoisomerase II. The drug-topoisomerase II complexes induce DNA double strand breaks, transcriptome changes, and subsequent cell death when the damage is not repaired. Alternatively or in addition, reactive oxygen species and oxidative stress may lead to DNA damage, lipid peroxidation of membranes (especially cardiolipin), and subsequent irreversible damage and fibrosis. The dilated cardiomyopathy observed with anthracycline usage is thought to be due to myocardial necrosis.

Endocrine Therapy

Estrogen's cardioprotective effects are likely due to its influence on lipoproteins, hemostasis, carbohydrate metabolism, and vessel wall tone and reactivity. Selective estrogen receptor modulators act as estrogen agonists in some systems, inhibiting either the oxidation of low-density lipoprotein or lipid peroxidation in cell membranes, which are required for triggering atherosclerosis. Aromatase inhibitors reduce circulating estrogens in postmenopausal women and therefore are associated with slightly increased CV event rates compared with tamoxifen.

Radiation Therapy

Radiation therapy has contributed significantly to the reduction of local recurrence and improved survival of patients with early stage breast cancer.9,10 Breast radiation (often including certain regional nodal fields) has traditionally been recommended routinely after lumpectomy, though recent data suggest that some patients who are older than 70 with low-risk tumors may safely omit radiation.11 After mastectomy, patients with large and/or node-positive disease may also benefit from chest wall and regional nodal radiation. Radiation is also occasionally used to manage symptoms (e.g., pain) in patients with stage 4 (incurable) cancers. When the heart and its structures are included even partially in the radiation field, potential CV side effects can include congestive HF, restrictive cardiomyopathy, ischemic heart disease, valvular heart disease, and constrictive pericarditis and pericardial effusion.12,13 In a recent study comparing CVD risk between the general population and more than 70,000 patients with breast cancer, radiation therapy was associated with an increased risk of the listed manifestations of radiation-induced heart disease.13

The risk of cardiotoxicity from radiation therapy is related to the total dose of radiation administered, dose per fraction, and volume of heart irradiated2 and, thus, differs depending on whether the left or right breast/chest wall undergoes radiation treatment.14-16 Newer techniques (e.g., three-dimensional planning, conformal blocking, deep-inspiration breath hold, and prone positioning) aim to reduce exposure of healthy tissue to radiation damage,17-21 but most patients who receive left-sided radiation still receive at least 2 Gy to the heart.22 Boekel and colleagues reported that the incidence of ischemic heart disease but not overall CVD was slightly increased after left- versus right-sided radiation therapy for breast cancer.13 The increase is proportional to the mean dose to the heart, begins within a few years after exposure, and continues for at least 20 years.12 In addition, CV side effects are more likely in patients with a known history of cardiac disease.2,23 CAD and cerebrovascular disease are common, as are diabetes, smoking, hypertension, high cholesterol, lack of physical activity, and poor nutrition. Although these comorbidities are typically associated with older age, younger age at treatment actually seems to increase the risk of radiation-induced cardiotoxicity as well.13,24

Trastuzumab

Trastuzumab is a monoclonal antibody that targets the human epidermal growth factor receptor 2 (HER2). Since its discovery, trastuzumab has dramatically improved the survival of the 15-20% of patients with breast cancer whose tumors overexpress HER2.25 However, this antibody is associated with a risk for asymptomatic decrease in left ventricular ejection fraction (LVEF) and, rarely, clinical HF.26,27

Trastuzumab-related cardiotoxicity is more likely in patients who have used anthracyclines previously or concomitantly.28,29 However, concurrent treatment with trastuzumab and radiation is safe.30,31 Pre-existing cardiac dysfunction such as decreased LVEF, older age (age greater than 50 years), obesity, and anti-hypertensive therapy are risk factors.32,33 Diabetes, valvular disease, and CAD are not consistently associated with increased risk,34-36 though diabetes appeared to be a risk factor in one cohort of elderly women.37

Trastuzumab-related cardiotoxicity does not depend on cumulative dose. Moreover, re-challenge is often tolerated after recovery.37 Use of dual HER2 blockade by adding pertuzumab or lapatinib (other drugs that target the same pathway) does not seem to increase the risk of cardiomyopathy over trastuzumab alone.38,39

Anthracycline-Based Chemotherapy

Prolonged treatment with anthracyclines (e.g., doxorubicin and daunorubicin) has been associated with cardiotoxicity in the form of cardiomyopathy and HF.40,41 Acute/subacute cardiotoxicity can be seen any time between treatment initiation and months after treatment cessation, but chronic cardiotoxicity (manifesting as clinical HF or subclinical decline in myocardial function) can take years to develop.42-44 Chronic cardiomyopathy often presents as asymptomatic diastolic or systolic dysfunction and progresses to HF, which may be fatal.

The strongest risk factor for anthracycline-induced cardiotoxicity is cumulative dose.45 Age at the time of exposure, co-administration of other cardiotoxic agents, chest irradiation, preexisting CVD, hypertension, diabetes, and peripheral vascular disease have been associated with anthracycline-related cardiotoxicity. Cardiac function has been shown to decrease up to 30 years after initial treatment,45 though it can be difficult to identify the contribution of anthracycline exposure versus other age-related comorbidities in some very late cases of cardiac dysfunction. As more patients live decades after the diagnosis and treatment of early stage breast cancer, more will have time to develop cardiac compromise. Additional research is needed to better understand the significant individual variability in susceptibility to anthracycline-induced cardiotoxicity. Genetic factors such as polymorphisms in the HAS3 gene may be at play.46

Endocrine Therapies

Endocrine therapy with selective estrogen receptor modulators (e.g., tamoxifen) and aromatase inhibitors improves survival for patients with hormone receptor-positive breast cancer.47,48 Aromatase inhibitors have slightly better antineoplastic efficacy than tamoxifen in postmenopausal patients, but rates of CVD are slightly higher after treatment with aromatase inhibitors than after tamoxifen.49 In a recent study of over 13,000 women, aromatase inhibitor-only users had a similar risk of cardiac ischemia and stroke as tamoxifen-only users, but dysrhythmia, valvular dysfunction, and pericarditis were more prevalent with use of aromatase inhibitors.50 For premenopausal patients, ovarian function suppression is sometimes administered in combination with tamoxifen and always administered if aromatase inhibitor therapy is to be used.51 However, the long-term CV effects of ovarian function suppression remain uncertain.

Detection of CV Toxicity

Baseline history and physical exam with attention to signs and symptoms of CV compromise is recommended for all patients who will undergo potentially cardiotoxic breast cancer treatment. Echocardiography (preferably three dimensional or two dimensional with contrast) is typically used to monitor cardiac function before and during trastuzumab (to identify reversible subclinical declines in ejection fraction) in patients with early-stage breast cancer. Whether there is value to echocardiographic monitoring in patients with metastatic disease (who may receive many years of treatment), and at what frequency if so, is uncertain.52,53 The utility of repeated echocardiography is also controversial in patients with early-stage cancer who are at relatively low risk of chronic HF due to young age and no receipt of anthracycline. Patients who receive anthracycline have traditionally been assessed by echocardiography only at baseline, but the 2015 National Comprehensive Cancer Network Survivorship guideline added a recommendation to consider post-anthracycline echocardiography in certain groups at high risk of cardiotoxicity (Table 2).54 The American Society of Echocardiography issued two consensus statements on cardiac monitoring during chemotherapy and after radiation therapy, which are meant to serve as a reference for cardiology and cardio-oncology providers.55,56 More research is warranted to identify which patients do and do not benefit from intensive monitoring and preventative strategies.

Table 2: Criteria for Consideration of Post-Anthracycline Echocardiography According to National Comprehensive Cancer Network Guidelines54

Age over 65

Underlying CV risk factors such as hypertension, dyslipidemia, diabetes mellitus, and family history of cardiomyopathy

History of other CV comorbidities (e.g., atrial fibrillation, known CAD, or structural heart disease)

Treatment with a high cumulative anthracycline dose (>300 mg/m2 doxorubicin)

Low-normal LVEF (50-54%) at baseline

Symptoms of HF

How Can We Prevent Cardiotoxicity?

It is important to take cardiac risks into account during the decision-making process about chemotherapy. For some patients who are at increased risk of cardiotoxicity due to age or comorbidities, the estimated risk/benefit ratio may weigh in favor of a non-anthracycline-containing regimen or no chemotherapy at all. For others, there may be a role for heart-protective medications concurrent with anthracycline use.57,58 A number of medications have been tested and tried as reviewed elsewhere.58 The most recent of the randomized controlled cohort studies is the PRADA (Prevention of Cardiac Dysfunction During Adjuvant Breast Cancer Therapy) trial, which used a 2 x 2 randomization to assign 130 women with early-stage breast cancer and no serious comorbidity to the angiotensin receptor blocker candesartan cilexetil, the beta-blocker metoprolol succinate, or matching placebos in parallel with adjuvant anthracycline-based chemotherapy. The overall decline in LVEF was 2.6% in the placebo group and 0.8% in the candesartan group in the intention-to-treat analysis. Ejection fraction declines did not differ between metoprolol and placebo.59 Indeed, there does not seem to be a class effect for beta-blockers, and only carvedilol and nebivolol have documented cardioprotective properties in the setting of anthracycline therapy. On the contrary, there does seem to be a class effect for angiotensin-converting enzyme (ACE) inhibitors and possibly angiotensin-receptor blockers; of note, spironolactone seems to be protective against anthracycline cardiotoxicity as well. One small-sized randomized controlled study outlined the cardioprotective properties of atorvastatin. Currently, the PREVENT (Preventing Anthracycline Cardiovascular Toxicity With Statins) study is enrolling patients with early-stage breast cancer and patients with lymphoma to receive a statin medication or placebo before and during anthracycline therapy to assess if cardiac function is protected by simvastatin. Dexrazoxane is an expensive and potentially toxic medication that is proven to decrease CV toxicity from anthracyclines, but due to adverse effects such as leukemia, the Food and Drug Administration has limited the use of dexrazoxane to women with metastatic breast cancer who have received a cumulative dose of 300 mg/m2 doxorubicin and who will continue to receive doxorubicin to maintain tumor control.60

For radiation-induced heart disease, no preventive effort other than reduction of exposure is effective. Patients with CV risk factors including age >65 years and history of CAD and myocardial infarction should be appropriately counseled on the risk of coronary/cardiac events.12

Treatment Options

Patients demonstrating signs of cardiac dysfunction should be referred for a cardiology consultation prior to receiving any additional cardiotoxic therapy. Cessation of a cardiotoxic drug can reverse or limit toxicity in some cases. When acute cardiotoxicity is identified in a patient receiving anthracycline, a change in regimen to a non-anthracycline-containing combination (or cessation of chemotherapy entirely depending on the clinical situation) is warranted. Cardinale and colleagues found that 42% of patients with asymptomatic ejection fracture declines after anthracycline therapy experienced complete ejection fracture recovery after treatment with a beta-blocker and ACE inhibitor, but there was no placebo in this study to clarify if there would have been improvement with anthracycline cessation alone.61 Randomized clinical trial data regarding optimal drugs for treatment of cardiotoxicity in patients with breast cancer are needed. Current best practices include use of beta-blockers and ACE inhibitors, lifestyle modifications (smoking cessation and exercise), and management of other comorbidities (e.g., diabetes, hypertension, and hypercholesterolemia).

References

  1. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin 2016;66:7-30.
  2. Bovelli D, Plataniotis G, Roila F; ESMO Guidelines Working Group. Cardiotoxicity of chemotherapeutic agents and radiotherapy-related heart disease: ESMO Clinical Practice Guidelines. Ann Oncol 2010;21(Suppl 5):v277-82.
  3. Yeh ET, Tong AT, Lenihan DJ, et al. Cardiovascular complications of cancer therapy: diagnosis, pathogenesis, and management. Circulation 2004;109:3122-31.
  4. Hooning MJ, Aleman BM, van Rosmalen AJ, Kuenen MA, Klijn JG, van Leeuwen FE. Cause-specific mortality in long-term survivors of breast cancer: A 25-year follow-up study. Int J Radiat Oncol Biol Phys 2006;64:1081-91.
  5. Bartelink H, Horiot JC, Poortmans PM, et al. Impact of a higher radiation dose on local control and survival in breast-conserving therapy of early breast cancer: 10-year results of the randomized boost versus no boost EORTC 22881-10882 trial. J Clin Oncol 2007;25:3259-65.
  6. Taunk NK, Haffty BG, Kostis JB, Goyal S. Radiation-induced heart disease: pathologic abnormalities and putative mechanisms. Front Oncol 2015;5:39.
  7. Zeglinski M, Ludke A, Jassal DS, Singal PK. Trastuzumab-induced cardiac dysfunction: A 'dual-hit'. Exp Clin Cardiol 2011;16:70-4.
  8. Volkova M, Russell R 3rd. Anthracycline cardiotoxicity: prevalence, pathogenesis and treatment. Curr Cardiol Rev 2011;7:214-20.
  9. Smith IE, Dowsett M. Aromatase inhibitors in breast cancer. N Engl J Med 2003;348:2431-42.
  10. Clarke M, Collins R, Darby S, et al. Effects of radiotherapy and of differences in the extent of surgery for early breast cancer on local recurrence and 15-year survival: an overview of the randomised trials. Lancet 2005;366:2087-106.
  11. Hughes KS, Schnaper LA, Bellon JR, et al. Lumpectomy plus tamoxifen with or without irradiation in women age 70 years or older with early breast cancer: long-term follow-up of CALGB 9343. J Clin Oncol 2013;31:2382-7.
  12. Darby SC, Ewertz M, McGale P, et al. Risk of ischemic heart disease in women after radiotherapy for breast cancer. N Engl J Med 2013;368:987-98.
  13. Boekel NB, Schaapveld M, Gietema JA, et al. Cardiovascular Disease Risk in a Large, Population-Based Cohort of Breast Cancer Survivors. Int J Radiat Oncol Biol Phys 2016;94:1061-72.
  14. Correa CR, Litt HI, Hwang WT, Ferrari VA, Solin LJ, Harris EE. Coronary artery findings after left-sided compared with right-sided radiation treatment for early-stage breast cancer. J Clin Oncol 2007;25:3031-7.
  15. Vallis KA, Pintilie M, Chong N, et al. Assessment of coronary heart disease morbidity and mortality after radiation therapy for early breast cancer. J Clin Oncol 2002;20:1036-42.
  16. Nilsson G, Holmberg L, Garmo H, et al. Distribution of coronary artery stenosis after radiation for breast cancer. J Clin Oncol 2012;30:380-6.
  17. Giraud P, Djadi-Prat J, Morelle M, et al. Contribution of respiratory gating techniques for optimization of breast cancer radiotherapy. Cancer Invest 2012;30:323-30.
  18. Zellars R, Bravo PE, Tryggestad E, et al. SPECT analysis of cardiac perfusion changes after whole-breast/chest wall radiation therapy with or without active breathing coordinator: results of a randomized phase 3 trial. Int J Radiat Oncol Biol Phys 2014;88:778-85.
  19. Raza S, Lymberis SC, Ciervide R, et al. Comparison of Acute and Late Toxicity of Two Regimens of 3- and 5-Week Concomitant Boost Prone IMRT to Standard 6-Week Breast Radiotherapy. Front Oncol 2012;2:44.
  20. Mulliez T, Veldeman L, Speleers B, et al. Heart dose reduction by prone deep inspiration breath hold in left-sided breast irradiation. Radiother Oncol 2015;114:79-84.
  21. Mulliez T, Speleers B, Mahjoubi K, et al. Prone left-sided whole-breast irradiation: significant heart dose reduction using end-inspiratory versus end-expiratory gating. Cancer Radiother 2014;18:672-7.
  22. Taylor CW, Povall JM, McGale P, et al. Cardiac dose from tangential breast cancer radiotherapy in the year 2006. Int J Radiat Oncol Biol Phys 2008;72:501-7.
  23. Valachis A, Nilsson C. Cardiac risk in the treatment of breast cancer: assessment and management. Breast Cancer (Dove Med Press) 2015;7:21-35.
  24. Antonini N, Jones H, Horiot JC, et al. Effect of age and radiation dose on local control after breast conserving treatment: EORTC trial 22881-10882. Radiother Oncol 2007;82:265-71.
  25. Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med 2005;353:1673-84.
  26. Thavendiranathan P, Abdel-Qadir H, Fischer HD, et al. Breast Cancer Therapy-Related Cardiac Dysfunction in Adult Women Treated in Routine Clinical Practice: A Population-Based Cohort Study. J Clin Oncol 2016;34:2239-46.
  27. Hansel TT, Kropshofer H, Singer T, Mitchell JA, George AJ. The safety and side effects of monoclonal antibodies. Nat Rev Drug Discov 2010;9:325-38.
  28. Bowles EJ, Wellman R, Feigelson HS, et al. Risk of heart failure in breast cancer patients after anthracycline and trastuzumab treatment: a retrospective cohort study. J Natl Cancer Inst 2012;104:1293-305.
  29. 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.
  30. Romond EH, Perez EA, Bryant J, et al. Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer. N Engl J Med 2005;353:1673-84.
  31. Piccart-Gebhart MJ, Procter M, Leyland-Jones B, et al. Trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med 2005;353:1659-72.
  32. Feldman AM, Lorell BH, Reis SE. Trastuzumab in the treatment of metastatic breast cancer : anticancer therapy versus cardiotoxicity. Circulation 2000;102:272-4.
  33. Perik PJ, de Korte MA, van Veldhuisen DJ, Gietema JA, Sleijfer DT, de Vries EG. Cardiotoxicity associated with the use of trastuzumab in breast cancer patients. Expert Rev Anticancer Ther 2007;7:1763-71.
  34. Perez EA, Suman VJ, Davidson NE, et al. Cardiac safety analysis of doxorubicin and cyclophosphamide followed by paclitaxel with or without trastuzumab in the North Central Cancer Treatment Group N9831 adjuvant breast cancer trial. J Clin Oncol 2008;26:1231-8.
  35. Ewer SM, Ewer MS. Cardiotoxicity profile of trastuzumab. Drug Saf 2008;31:459-67.
  36. 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.
  37. Keefe DL. Trastuzumab-associated cardiotoxicity. Cancer 2002;95:1592-600.
  38. 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.
  39. Blackwell KL, Burstein HJ, Storniolo AM, et al. Randomized study of Lapatinib alone or in combination with trastuzumab in women with ErbB2-positive, trastuzumab-refractory metastatic breast cancer. J Clin Oncol 2010;28:1124-30.
  40. Zambetti M, Moliterni A, Materazzo C, et al. Long-term cardiac sequelae in operable breast cancer patients given adjuvant chemotherapy with or without doxorubicin and breast irradiation. J Clin Oncol 2001;19:37-43.
  41. Shan K, Lincoff AM, Young JB. Anthracycline-induced cardiotoxicity. Ann Intern Med 1996;125:47-58.
  42. Bowles EJ, Wellman R, Feigelson HS, et al. Risk of heart failure in breast cancer patients after anthracycline and trastuzumab treatment: a retrospective cohort study. J Natl Cancer Inst 2012;104:1293-305.
  43. Doyle JJ, Neugut AI, Jacobson JS, Grann VR, Hershman DL. Chemotherapy and cardiotoxicity in older breast cancer patients: a population-based study. J Clin Oncol 2005;23:8597-605.
  44. Krischer JP, Epstein S, Cuthbertson DD, Goorin AM, Epstein ML, Lipshultz SE. Clinical cardiotoxicity following anthracycline treatment for childhood cancer: the Pediatric Oncology Group experience. J Clin Oncol 1997;15:1544-52.
  45. Singal PK, Iliskovic N. Doxorubicin-induced cardiomyopathy. N Engl J Med 1998;339:900-5.
  46. Wang X, Liu W, Sun CL, et al. Hyaluronan synthase 3 variant and anthracycline-related cardiomyopathy: a report from the children's oncology group. J Clin Oncol 2014;32:647-53.
  47. Fisher B, Costantino J, Redmond C, et al. A randomized clinical trial evaluating tamoxifen in the treatment of patients with node-negative breast cancer who have estrogen-receptor-positive tumors. N Engl J Med 1989;320:479-84.
  48. Howell A, Cuzick J, Baum M, et al. Results of the ATAC (Arimidex, Tamoxifen, Alone or in Combination) trial after completion of 5 years' adjuvant treatment for breast cancer. Lancet 2005;365:60-2.
  49. Cuppone F, Bria E, Verma S, et al. Do adjuvant aromatase inhibitors increase the cardiovascular risk in postmenopausal women with early breast cancer? Meta-analysis of randomized trials. Cancer 2008;112:260-7.
  50. Haque R, Shi J, Schottinger JE, et al. Cardiovascular Disease After Aromatase Inhibitor Use. JAMA Oncol 2016 Apr 21 [Epub ahead of print].
  51. Francis PA, Regan MM, Fleming GF, et al. Adjuvant ovarian suppression in premenopausal breast cancer. N Engl J Med 2015;372:436-46.
  52. Davis CC, Zelnak A, Eley JW, Goldstein DA, Switchenko JM, McKibbin T. Clinical Utility of Routine Cardiac Monitoring in Breast Cancer Patients Receiving Trastuzumab. Ann Pharmacother 2016;50:712-7.
  53. Yu AF, Manrique C, Pun S, et al. Cardiac Safety of Paclitaxel Plus Trastuzumab and Pertuzumab in Patients With HER2-Positive Metastatic Breast Cancer. Oncologist 2016;21:418-24.
  54. Clinical Practice Guidelines in Oncology Survivorship (Version 1.2016) (National Comprehensive Cancer Network website). 2016. Available at: https://www.nccn.org/professionals/physician_gls/pdf/survivorship.pdf. Accessed 08/02/2016.
  55. Lancellotti P, Nkomo VT, Badano LP, et al. Expert consensus for multi-modality imaging evaluation of cardiovascular complications of radiotherapy in adults: a report from the European Association of Cardiovascular Imaging and the American Society of Echocardiography. J Am Soc Echocardiogr 2013;26:1013-32.
  56. 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.
  57. Yeh ET, Vejpongsa P. Subclinical Cardiotoxicity Associated With Cancer Therapy: Early Detection and Future Directions. J Am Coll Cardiol 2015;65:2523-5.
  58. 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.
  59. Gulati G, Heck SL, Ree AH, et al. Prevention of cardiac dysfunction during adjuvant breast cancer therapy (PRADA): a 2 × 2 factorial, randomized, placebo-controlled, double-blind clinical trial of candesartan and metoprolol. Eur Heart J 2016;37:1671-80.
  60. 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.
  61. 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.

Keywords: Cardiotoxicity, Breast Neoplasms, Anthracyclines, Antineoplastic Agents, Aromatase Inhibitors, Aromatase, Atrial Fibrillation, Cardiomyopathies, Cardiotoxins, Coronary Artery Disease, Diabetes Mellitus, Doxorubicin, Dyslipidemias, Echocardiography, Hypertension, Pericarditis, Peripheral Vascular Diseases, Postmenopause, Risk Factors, Selective Estrogen Receptor Modulators, Stroke, Tamoxifen, Adrenergic beta-Antagonists, Angiotensin-Converting Enzyme Inhibitors, Hypercholesterolemia, Smoking Cessation, Smoking Cessation, Carbazoles, Leukemia, Lymphoma, Myocardial Infarction, Heart Failure


< Back to Listings