Article

Cancer and Heart Failure: Understanding the Intersection

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Average (ratings)
No ratings
Your rating

Abstract

Cancer and cardiovascular disease account for nearly half of all deaths in the US. The majority of cancer therapies are known to cause potential cardiac toxicity in some form. Patients with underlying cardiac disease are at a particularly increased risk for worse outcomes following cancer therapy. Most alarming is the potential for heart failure as a result of cancer treatment, which may lead to early disruption or withdrawal of life-saving cancer therapies and can potentially increase cardiovascular mortality. A multi-disciplinary cardio-oncology approach can improve outcomes through early surveillance, prevention and treatment strategies.

Disclosure:MWB reports grant support from Gilead. CEH has no conflicts of interest to declare.

Received:

Accepted:

Correspondence Details:Michelle W Bloom, Outpatient Services, Heart Failure and Cardiomyopathy Center and Co-Director, Cardiology Division, Stony Brook University, Health Sciences Center, T-16, Room 080, SUNY at Stony Brook, NY 11794, US. E: michelle.bloom@stonybrookmedicine.edu

Copyright Statement:

The copyright in this work belongs to Radcliffe Medical Media. Only articles clearly marked with the CC BY-NC logo are published with the Creative Commons by Attribution Licence. The CC BY-NC option was not available for Radcliffe journals before 1 January 2019. Articles marked ‘Open Access’ but not marked ‘CC BY-NC’ are made freely accessible at the time of publication but are subject to standard copyright law regarding reproduction and distribution. Permission is required for reuse of this content.

Heart disease and cancer are the two leading causes of death, together accounting for almost 50 % of deaths in the US.1 Unfortunately, the tremendous success in improving cancer survival is often attenuated by downstream cardiovascular complications from cancer therapy. Acute cardiac toxicity may limit the ability to use life-saving cancer therapy and chronic toxicity limits overall survival. As novel agents with more specific targets become available, unanticipated cardiac complications arise. The spectrum of cardiac toxicities is broad, ranging from ischaemia and arrhythmias to hypertension, left ventricular dysfunction (LVD) and the development of clinical heart failure (HF). Notably, there is increasing evidence that suggests an underlying relationship between cancer and the heart, independent of cancer therapies’ effect on left ventricular ejection fraction (LVEF) and heart rate.2,3 HF caused by chemotherapy portends a worse prognosis compared with idiopathic and ischaemic cardiomyopathy,4 highlighting the need for enhanced understanding and management.

Definitions of Cancer Therapy-related Cardiac Toxicity

There is currently no universally accepted definition of cancer therapyrelated cardiac toxicity; however, several definitions pertaining to specific agents have been proposed. A recent consensus defines cancer therapy-related cardiac dysfunction (CTRCD) as a decrease in LVEF of >10 % to a value of <53 %.5 The Cardiac Review and Evaluation Committee involved in trials with trastuzumab defines cardiotoxicity as a decrease in LVEF that is either global or more severe in the septum and decline in LVEF of at least 5 % to <55 % with signs or symptoms of HF, or a decline of at least 10 % to <55 % without HF signs or symptoms.6 According to the Food and Drug Administration, anthracycline cardiotoxicity is defined as a >20 % decrease in LVEF when the baseline LVEF is normal, or >10 % decrease when baseline LVEF is abnormal.7 The American Society of Echocardiography and the European Association of Cardiovascular Imaging define cardiac toxicity as a decrease in LVEF of >10 %, to a value <53 % confirmed by repeat imaging.5

Cancer Therapy-induced Heart Failure

Anthracyclines

Anthracylines are a class of chemotherapies that include doxorubicin and daunorubicin, widely used in the treatment of various solid and liquid tumours. Their mechanism of action involves the generation of reactive oxygen species as well as DNA intercalation to impair protein synthesis and inhibition of topoisomerase II, inhibiting DNA repair.8 In contrast to topoisomerase II-alpha, which is located on rapidly dividing cells, topoisomerase II-beta, located on cardiac myocytes is the proposed mechanism behind the cardiotoxicity.9 Anthracycline use has been shown to result in LVD with an incidence of 1–5 %, ranging from acute to late onset.10 Anthracycline-induced HF is thought to be cumulative and dose-dependent, with an incidence reaching 5% at a total doxorubicin dose of 450 mg/m2.11 However, examination of endomyocardial biopsies reveals histopathological changes with doses as low as 240 mg/m2, suggesting that subclinical cardiotoxity may be present as early as the first dose.12

HER2-targeted Agents

Trastuzumab, a monoclonal antibody targeted against humanised epidermal growth factor receptor 2 (HER2) has played a pivotal role in breast cancer patients whose tumours overexpress HER2. Under normal circumstances, in cardiac myocytes, the HER2 signalling pathway is responsible for adaptation to stress.8 The incidence of LVD with trastuzumab ranges from 2 % to 28 %,10 with a 1.7–4.1 % incidence of HF.13 Other HER2-targeted agents, such as lapatinib and pertuzumab, used in combination with trastuzumab have not shown additive cardiotoxicity.14

Mitotic Inhibitors

Taxanes such as paclitaxel and docetaxel impair cell division through inhibition of microtubule disassembly during mitosis.15 The incidence of HF from docetaxel has been shown to range from 2.3 % to 8.0 %.16 Combination of doxorubicin and paclitaxel resulted in an unanticipated increase in HF, thought to occur from an increase in plasma levels of doxorubicin by paclitaxel. This in turn results in increased anthracycline uptake into the heart.17 The increased risk can be attenuated by increasing the duration between the administration of agents or using docetaxel instead of paclitaxel as an interaction; this was not observed with this combination.17

Table 1: Cancer Therapies Associated with Left Ventricular Dysfunction and Heart Failure

Article image

Alkylating Agents

Alkylating agents such as cyclophosphamide and ifosfamide impair protein synthesis via inhibition of DNA transcription.18 LVD from cyclophosphamide has been shown to occur in 7–28 % of patients, with a risk that is dose-dependent.16

Proteasome Inhibitors

Proteasome inhibitors such as bortezomib and carfilzomib interfere with the degradation of cell cycle proteins, ultimately resulting in cell death. Compared with other tissues, proteasome activity is increased in the heart and thus disruption of protein homeostasis in cardiac myocytes results in cardiotoxic effects, including HF.19 The incidence of HF from bortezomib ranges from 2 % to 5 %.20 In a recent study of patients with relapsed multiple myeloma, the addition of carfilzomib to lenalidomide and dexamethasone resulted in a 6.4 % incidence of HF compared with 4.1 % with lenalidomide and dexamethasone alone.21

Small Tyrosine Kinase Inhibitors

Sunitinib is a non-selective inhibitor of vascular endothelial growth factor (VEGF) with multiple downstream target effects such as hypertension, arrhythmia and HF with a reported incidence of 2–11 %.10 Imatinib, an inhibitor of the fusion protein breakpoint cluster region (BCR)/Abelson (ABL) has been instrumental in the treatment of chronic myelogenous leukaemia; however, there has been a reported 0.5–1.7 % incidence of LVD, thought to be caused by mitochondrial abnormalities within cardiac myocytes following treatment with imatinib.10,22

Immune modulators

Immunotherapy has played an increasing role in cancer treatment, as targeting inhibitory T cells helps overcome tumour-induced immune evasion. The anti-cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) antibody, ipilimumab, and the anti-programmed death-1 (PD-1) antibody, nivolumab, have been shown to improve survival in patients with melanoma.23 However, the combination of these agents has been associated with an immune-mediated myocarditis, thought to occur from immune activation of cardiac as well as tumour tissue.24

Radiation

Radiation therapy results in the fragmentation of DNA, interfering with cell reproduction and viability, primarily impacting rapidly dividing cancer cells.20 Radiation to the chest at doses >30 Gy can affect the cardiovascular system, albeit toxicity usually manifests years after exposure. Risk factors for cardiovascular injury include higher doses, longer intervals from the time of radiation, younger age at time of irradiation, and coexisting cardiovascular risk factors (see Table 1).25

Prevention and Surveillance

Biomarkers

Various biomarkers have been studied with regard to detection of cancer therapy-related cardiotoxicity.26 While their role is not firmly established, they may provide an additive benefit in the diagnosis of cardiotoxicity, cardiac risk stratification, prevention of cardiac events and assessment of cardioprotective effectiveness. Among cancer patients receiving high-dose chemotherapy, elevations in troponin-I were predictive of a higher incidence of cardiac events and LVEF reduction.27 In children with high-risk acute lymphoblastic leukaemia, increases in N-terminal pro-brain natriuretic peptide (NT-proBNP) were associated with abnormal LV thickness-to-dimension ratio, suggestive of LV remodelling.28 Other studies with natriuretic peptide have not shown similar promise.29–31 Abnormal high-sensitivity C-reactive protein (hs-CRP) was predictive of LVEF decline in breast cancer patients treated with trastuzumab.32

Imaging

Multigated acquisition (MUGA) scan has been the imaging modality of choice for periodic assessment of LVEF; however, it poses a risk of radiation exposure and provides limited information on systolic and diastolic function.28 Echocardiography with strain imaging enables regional and global assessment of cardiac function, and is more sensitive for subtle damage to myocardial ultrastructure, which is otherwise only confirmed histologically. LVEF is based on assumptions of cardiac geometry, depends on 2D image quality and cannot detect subtle regional alterations in myocardial mechanics.29

Figure 1: Algorithm for Surveillance and Treatment of Cardiac Toxicity Prior (A), During (B) and Following (C) Therapy

Article image

Reduction of longitudinal strain (>10 % from baseline) after three months can predict future reduction in LVEF (after six months) with a sensitivity of 78–79 % and specificity of 79–82 %.30 In patients treated with trastuzumab, a decrease in global longitudinal and radial strain but not LVEF was observed as early as three months in patients who later developed cardiotoxicity.31 In a study of breast cancer patients treated with anthracyclines, systolic dysfunction could be detected with strain imaging within seven days of completing anthracyclines without similar change in LVEF. Furthermore, findings showed that low-dose anthracycline-induced myocardial injury was transient; however, persistently reduced global strain occurred in approximately 16 % of HER2/neu negative patients when systolic strain was reduced ≤-17.2 % at six months following therapy.32 Cardiac MRI is the gold standard of ventricular volume quantification and function, and may play a role in the detection of early as well as late cardiac toxicity.5

Therapies for Prevention

Several therapeutic agents including dexrazoxane, statins, angiotensin converting-enzyme (ACE)-Inhibitors and beta-blockers have been shown to be beneficial in reducing cardiotoxicity.33 Patients previously treated with anthracyclines who were given dexrazoxane with anthracycline treatment showed decreased cardiac events, decreased incidence of HF and increased cardiac event-free survival. Additionally, there was a threefold decline in the risk of developing cardiac events, and the risk of developing HF was reduced by 90 %. The proposed mechanisms behind the cardiac protection include reduction of free radical formation as well as prevention of topoisomerase II-DNA complex formation.34,35

The protective effect of antioxidants has been reinforced in animal studies of carvedilol, which has the ability to chelate iron, enabling the prevention of the cardiac histopathology caused by doxorubicin.

Interesting, this same benefit was not seen in other beta-blockers such as atenolol.36 The use of a beta-blocker, initiated before chemotherapy and maintained after treatment, has been shown to preserve LV systolic function.37,38 However, one of these studies was a small prospective trial37 and the other was a retrospective analysis of a larger cohort of patients in which the use of beta-blockers was associated with fewer clinical HF events.38

When used in combination, enalapril and carvedilol, given 1–2 weeks post chemotherapy, increased LVEF recovery. After twelve months, the chance of getting partial LVEF improvement was lost, reinforcing the importance of early treatment.39 In patients receiving chemotherapy with anthracyclines plus or minus trastuzumab, candesartan, given in parallel with cancer therapy reduced LVEF decline.40 Interestingly, in this small clinical trial, the use of the beta-blocker metoprolol succinate was not associated with a reduction in LVEF decline.40 Although the benefit of certain agents has been suggested for prevention of a decrease in LVEF or HF, the data are primarily based on small clinical or observational studies, and as such larger clinical trials are needed to definitively demonstrate the utility of these agents.

Surveillance Strategies

Primary prevention and early detection of subclinical toxicity may provide the key to mortality benefit within the field of cardio oncology. Early and diligent surveillance allows for the detection of cardiac dysfunction prior to the development of symptoms and downstream toxicity. Although many algorithms for surveillance have been proposed,5,41–43 there are currently no universally accepted guidelines regarding how to approach these patients. Ideally, a careful screening of oncological and cardiovascular risk factors as well as baseline imaging, inclusive of echocardiography with strain, should occur prior to initiation of potentially cardiotoxic chemotherapy.

Cardiac risk factors include but are not limited to age, female gender, history of myocardial infarction or LVD and tachycardia, as this may be an early sign of cardiac damage.44 Cardiac biomarkers may provide an additive role in this setting. For patients considered to be at higher risk for the development of cardiotoxicity, a cardio-oncology consultation should be offered. In a context of a multidisciplinary approach to cancer care, the cardiac component of the evaluation should not translate to a delay in initiation of cancer therapy. Imaging and biomarker surveillance during and after cancer therapy should be tailored based on individual risk factors.45 According to the Food and Drug Administration, trastuzumab should be withheld for four weeks if the LVEF decreases ≥16 % from pre-treatment values or decreases below normal and ≥10 % absolute decrease in LVEF from pre-treatment values with resumption of chemotherapy once LVEF normalises for the absolute decrease from baseline is ≤15 % within 4–8 weeks.46 Doxorubicin should be discontinued upon development of HF.47 However, guidelines for withholding or stopping cancer therapy in the setting of asymptomatic LVD are not clearly delineated (see Figure 1).

Conclusion

Whether patients with pre-existing cardiovascular disease require cancer therapy with potentially cardiotoxic agents or previously healthy patients develop cardiac complications from cancer therapy, a collaborative patient-centred approach between the cardiologist and oncologist is essential to successful patient care. The rapid and vast advancement in cancer therapies suggests that we are perhaps only witnessing the beginning of what can be expected in the realm of downstream cardiovascular complications. The overarching goal of a cardio-oncology partnership should encompass a multifaceted approach to best facilitate early detection and appreciation of subclinical toxicity to enable successful uninterrupted completion of cancer therapy and prevent cardiovascular mortality.

References

  1. CDC. Leading Causes of Death, 2014. Available at: www.cdc.gov/nchs/fastats/leading-causes-of-death.htm (Accessed 20 November 2016).
  2. Cramer L, Hildebrandt B, Kung T, et al. Cardiovascular function and predictors of exercise capacity in patients with colorectal cancer. J Am Coll Cardiol 2014;64:1310–9.
    Crossref | PubMed
  3. Anker MS, Ebner N, Hildebrandt B, et al. Resting heart rate is an independent predictor of death in patients with colorectal, pancreatic, and non-small cell lung cancer: results of a prospective cardiovascular long-term study. Eur J Heart Fail 2016;18:1524–34.
    Crossref | PubMed
  4. 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.
    Crossref | PubMed
  5. 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. PMCID:PMC4402366
    Crossref | PubMed
  6. Seidman A, Hudis C, Pierri MK, et al. Cardiac dysfunction in the trastuzumab clinical trials experience. J Clin Oncol 2002;20:1215–21.
    Crossref | PubMed
  7. FDA Drug Label for DOXIL- doxorubicin hydrochloride injection, suspension, liposomal, 2016. Available at: https://dailymed.nlm.nih.gov/dailymed/drugInfo. cfm?setid=21d9c619-7e94-49e2-ac41-31e9ea96554a (Accessed 20 November 2016).
  8. Monsuez JJ, Charniot JC, Vignat N, Artigou JY. Cardiac side effects of cancer chemotherapy. Int J Cardiol 2010;144:3–15.
    Crossref | PubMed
  9. Vejpongsa P, Yeh ET. Topoisomerase 2beta: a promising molecular target for primary prevention of anthracycline induced cardiotoxicity. Clin Pharmacol Ther 2014;95:45–52.
    Crossref | PubMed
  10. Wells QS, Lenihan DJ. Reversibility of left ventricular dysfunction resulting from chemotherapy: can this be reversed? Prog Cardiovasc Dis 2010;53:140–8.
    Crossref | PubMed
  11. Von Hoff DD, Layard MW, Basa P, et al. Risk factors for doxorubicin-induced congestive heart failure. Ann Intern Med 1979;91 :710–7.
    Crossref | PubMed
  12. Billingham ME, Mason JW, Bristow MR, Daniels JR. Anthracycline cardiomyopathy monitored by morphologic changes. Cancer Treat Rep 1978;62:865–72.
    PubMed
  13. 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. PMCID:PMC3433392
    Crossref | PubMed
  14. Valachis A, Nearchou A, Lind P, Mauri D. Lapatinib, trastuzumab or the combination added to preoperative chemotherapy for breast cancer: a meta-analysis of randomized evidence. Breast Cancer Res Treat 2012;135: 655–62.
    Crossref | PubMed
  15. Field JJ, Kanakkanthara A, Miller JH. Microtubule-targeting agents are clinically successful due to both mitotic and interphase impairment of microtubule function. Bioorg Med Chem 2014;22:5050–9.
    Crossref | PubMed
  16. Curigliano G, Mayer EL, Burstein HJ, et al. Cardiac toxicity from systemic cancer therapy: a comprehensive review. Prog Cardiovasc Dis 2010;53:94–104.
    Crossref | PubMed
  17. Minotti G, Menna P, Salvatorelli E, et al. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev 2004;56:185–229.
    Crossref | PubMed
  18. Gershwin ME, Goetzl EJ, Steinberg AD. Cyclophosphamide: use in practice. Ann Intern Med 1974;80:531–40.
    Crossref | PubMed
  19. Grandin EW, Ky B, Cornell RK, et al. Patterns of cardiac toxicity associated with irreversible protesome inhibition in the treatment of multiple myeloma. J Card Fail 2015;21 :138–44.
    Crossref | PubMed
  20. Herrmann J, Lerman A, Sandhu NP, et al. Evaluation and management of patients with heart disease and cance: cardio-oncology. Mayo Clin Proc 2014;89:1287–306.; PMCID:PMC4258909
    Crossref | PubMed
  21. Stewart AK, Rajkumar SV, Dimopoulos MA, et al. Carfilzomib, lenalidomide, and dexamethasone for relapsed multiple myeloma. N Engl J Med 2015;372:142–52.
    Crossref | PubMed
  22. Kerkelä R, Grazette L, Yacobi R, et al. Cardiotoxicity of the cancer therapeutic agent imatinib mesylate. Nat Med 2006;12:908–16.
    Crossref | PubMed
  23. Postow MA, Chesney J, Pavlick AC, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med 2015;372:2006–17.
    Crossref | PubMed
  24. Johnson DB, Balko JM, Compton ML, et al. Fulminant Myocarditis with Combination Immune Checkpoint Blockade. N Engl J Med 2016;375:1749–55.
    Crossref | PubMed
  25. Groarke JD, Nguyen PL, Nohria A, et al. Cardiovascular complications of radiation therapy for thoracic malignancies: the role for non-invasive imaging for detection of cardiovascular disease. Eur Heart J 2014;35:612–23. PMCID:PMC3945797
    Crossref | PubMed
  26. Bloom MW, Hamo CE, Cardinale D, et al. Cancer Therapy Related Cardiac Dysfunction and Heart Failure: Part 1: Definitions, Pathophysiology, Risk Factors, and Imaging. Circ Heart Fail 2016;9:e002661. PMCID:PMC4709035
    Crossref | PubMed
  27. 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.
    Crossref | PubMed
  28. 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. PMCID:PMC3341148
    Crossref | PubMed
  29. 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. PMCID:PMC4286181
    Crossref | PubMed
  30. 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. PMCID:PMC3703313
    Crossref | PubMed
  31. 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.
    Crossref | PubMed
  32. Onitilo AA, Engel JM, Stankowski RV, et al. High-sensitivity C-reactive protein (hs-CRP) as a biomarker for trastuzumabinduced cardiotoxicity in HER2-positive early-stage breast cancer: a pilot study. Breast Cancer Res Treat 2012;134:291–8.
    Crossref | PubMed
  33. Kalam K, Marwick TH. Role of cardioprotective therapy for prevention of cardiotoxicity with chemotherapy: a systematic review and meta-analysis. Eur J Cancer 2013;49:2900–9.
    Crossref | PubMed
  34. Marty M, Espié 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.
    Crossref | PubMed
  35. Hahn VS, Lenihan DJ, Ky B. Cancer therapy-induced cardiotoxicity: basic mechanisms and potential cardioprotective therapies. J Am Heart Assoc 2014;3: e000665.
    Crossref | PubMed
  36. Oliveira PJ, Bjork JA, Santos MS, et al. Carvedilol-mediated antioxidant protection against doxorubicin-induced cardiac mitochondrial toxicity. Toxicol Appl Pharmacol 2004;200:159–68.
    Crossref | PubMed
  37. Kalay N, Basar E, Ozdogru I, et al. Protective effects of carvedilol against anthracycline-induced cardiomyopathy. J Am Coll Cardiol 2006;48:2258–62.
    Crossref | PubMed
  38. Seicean S, Seicean A, Alan N, et al. Cardioprotective effect of beta-adrenoreceptor blockage in patients with breast cancer undergoing chemotherapy: a follow up study of heart failure. Circ Heart Fail 2013;6:420–6.
    Crossref | PubMed
  39. Cardinale D, Colombo A, Lamantia G, et al. Anthracyclineinduced cardiomyopathy: clinical relevance and response to pharmacologic therapy. J Am Coll Cardiol 2010;55:213–20.
    Crossref | PubMed
  40. Gulati G, Heck SL, Ree AH, et al. Prevention of cardiac dysfunction during adjuvant breast cancer therapy (PRADA): a 2 x 2 factorial, randomized, placebo-controlled, doubleblind clinical trial of candesartan and metoprolol. Eur Heart J 2016;37:1671–80. PMCID:PMC4887703
    Crossref | PubMed
  41. Eschenhagen T, Force T, Ewer MS, et al. Cardiovascular side effects of cancer therapies: a position statement from the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2011;13:1–10.
    Crossref | PubMed
  42. 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.
    Crossref | PubMed
  43. 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.
    Crossref | PubMed
  44. Senkus E, Jassem J. Cardiovascular effects of systemic cancer treatment. Cancer Treat Rev 2011;37:300–11.
    Crossref | PubMed
  45. Hamo CE, Bloom MW, Cardinale D, et al. Cancer Therapy-Related Cardiac Dysfunction and Heart Failure: Part 2: Prevention, Treatment, Guidelines, and Future Directions. Circ Heart Fail 2016;9:e002843. PMCID:PMC4743885
    Crossref | PubMed
  46. FDA Drug Label for HERCEPTIN- trastuzumab, 2016. Available at: https://dailymed.nlm.nih.gov/dailymed/ drugInfo.cfm?setid=492dbdb2-077e-4064-bff3-372d6af0a7a2 (Accessed 23 December 2016).
  47. McMurray JJ, Adamopoulos S, Anker SD, et al. ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: The Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC. Eur Heart J 2012;33:1787–847.
    Crossref | PubMed