Pediatric Cardio-Oncology: Focusing on the Heart of Childhood Cancer Survivorship

Advancements in treatment and supportive care for childhood cancer have led to a dramatic increase in the overall survival such that >80% of newly diagnosed children will become long-term survivors.1 In response to this emerging population, a number of cohorts have been established to facilitate investigation of the long-term health impacts of cancer therapy.2-5 These and other similar initiatives have contributed to the recognition of the many potential chronic and late-occurring health conditions that survivors may experience, among which are myriad cardiovascular (CV) complications.6-12

We now recognize that childhood cancer survivors are seven times more likely than their siblings to die from a CV etiology, establishing CV conditions as the leading cause of noncancer-related mortality in this population.13 The risks for and spectrum of CV conditions in survivors are notable. In the largest investigation to date, investigators from The Childhood Cancer Survivor Study reported a fivefold-or-greater risk for congestive heart failure (HF), myocardial infarction, pericardial disease, and valvular abnormalities among nearly 15,000 childhood cancer survivors compared with siblings.10 Cardiomyopathy, the most widely investigated of these outcomes, is largely attributable to age- and dose-dependent exposures to the anthracycline (e.g., doxorubicin, daunorubicin, idarubicin, and epirubicin) and anthraquinone (e.g., mitoxantrone) classes of chemotherapy and cardiac-directed radiation. Myocardial dysfunction can be seen following nearly any cumulative anthracycline/anthraquinone exposure,14,15 but those patients who receive a doxorubicin-equivalent dose of ≥250 mg/m2 seem to be at highest lifetime risk.16,17 In addition to the risks for cardiomyopathy, chest radiation has also been associated with increased risk for valvular disease,10,17,18 pericarditis,10 coronary artery disease,10,17 and conduction disorders.17

Not all CV conditions experienced by survivors, however, can be solely attributed to direct cardiotoxic exposures. For example, the constellation of CV risk factors known as the metabolic syndrome occurs more frequently in survivors of acute lymphoblastic leukemia previously exposed to cranial radiation compared with those who were not.19-21 The occurrence of these risk factors is particularly concerning given reports that the presence of any one of hypertension, dyslipidemia, obesity, or diabetes conveys multiplicative, rather than additive, increases in risk for coronary artery disease, HF, valvular disease, and arrhythmia in long-term childhood cancer survivors.22 Similar findings are reported in survivors previously treated with hematopoietic stem cell transplant, where those who develop CV risk factors (hypertension, diabetes, and/or dyslipidemia) are at greater risk for CV disease than those who do not.23,24 Although the underlying mechanisms remain unestablished, the strength of association with these potentially modifiable risk factors supports future investigations seeking to establish pathophysiologic targets for subsequent intervention.

As knowledge of late toxicities of cancer therapies has grown, frontline treatment regimens for many childhood cancers have reduced, and in some cases eliminated, cardiotoxic exposures.25,26 For example, many Hodgkin lymphoma consortia have successfully incorporated alternative treatment agents in order to permit reduced exposures to anthracyclines and cardiac radiation.27 Reassuringly, a similar evolution in therapy across the broader spectrum of pediatric oncology has contributed to a fivefold reduction in the incidence of cardiac-related causes of death since the 1970s.28 Although this progress is encouraging, there are a number of childhood cancers for which cardiotoxic therapy reductions are not yet feasible without consequence to overall survival rates; therefore, many newly diagnosed individuals continue to incur risk for subsequent CV disease.

To address these needs, investigators have studied the use of alternative preventative strategies. Liposomal doxorubicin preparations, for example, have shown promise in adults with cancer largely due to reduced capillary penetration of cardiac (compared with tumor) tissue, thereby aiming to reduce cardiotoxicity without negative consequence to overall treatment efficacy.29,30 Pediatric investigators have utilized liposomal doxorubicin in frontline pediatric treatment with encouraging early success; however, long-term follow-up studies are needed to determine potential CV benefits.30,31 Another strategy has been the use of dexrazoxane, shown to attenuate anthracycline-mediated cardiotoxicity through what is believed to be the chelation of anthracycline-iron complexes, resulting in a reduction of iron-dependent free radical damage to cardiac myocytes.32 Its efficacy has gained approval by the United States Food and Drug Administration for use in women with metastatic breast cancer exposed to higher doses of doxorubicin, and its off-label use has shown promise in a number of pediatric oncology studies.33 As a historical note, initial results from a large Children's Oncology Group (COG) investigation in children diagnosed with Hodgkin lymphoma in the late 1990s suggested a non-statistically significant but concerning increase in the 4-year cumulative incidence of subsequent neoplasms in those who received dexrazoxane versus those who did not (3.43 vs. 0.6%, respectively; p = 0.060).34 The results from a second COG clinical trial performed during the same era were recently reported by Asselin and colleagues. In this study, children with T-cell acute lymphoblastic leukemia and lymphoblastic lymphoma were randomized whether to receive dexrazoxane. In contrast to the report from the Hodgkin studies, there was no statistically significant or suggested difference in the rates of subsequent neoplasms between groups (0.8 vs. 0.7%, respectively; p = 0.17).35 To address the concerns raised by the Hodgkin study, Chow and colleagues extended follow-up for the participants in both of these COG investigations.36 At a median of 12.6 years (range of 0-15.5 years) follow-up, dexrazoxane was not significantly associated with increased mortality (hazard ratio 1.03, 95% confidence interval, 0.73-1.45) or occurrence of second cancers (hazard ratio 1.24, 95% confidence interval, 0.49-3.15). These findings are reassuring as to the safety of anthracycline use in children with cancer. Providers should therefore consider the use of this agent for individuals likely to receive significant exposures to anthracycline chemotherapies.

The aforementioned risks for CV disease have prompted leading survivorship organizations to put forth recommendations for surveillance in individuals exposed to established cardiotoxic cancer treatments.37-41 A major focus within these guidelines has been that of cardiomyopathy screening, inherently due to the ready availability of effective surveillance mechanisms.42,43 Although a number of mechanisms exist by which to detect subclinical cardiac dysfunction, 2-dimensional echocardiography remains most frequently utilized due to its widespread availability.43,44 Current screening strategies comprise risk-stratified surveillance with 2-dimensional echocardiography, based upon age at exposure to cumulative doses of anthracyclines and radiation. However, the prolonged latency of cardiomyopathy onset has limited the ability to determine treatment efficacy in this unique population, thereby limiting optimal refinement of surveillance strategies. To overcome these limitations, recent simulation models have informed the development of more cost-effective screening approaches.45,46

Paramount to the young but growing field of pediatric cardio-oncology is the need to develop effective intervention strategies for individuals with cancer treatment-related cardiac dysfunction. Promising results from studies of adults treated for cancer indicate reduced rates of early anthracycline-induced cardiac dysfunction through the use of the angiotensin-converting enzyme inhibitor enalapril, with or without the addition of the beta-blocker carvedilol.47,48 However, when enalapril was given to childhood cancer survivors who had already developed left ventricular dysfunction, improved function was transient, suggesting that conventional treatments are likely ineffective following the onset of measurable cardiac dysfunction.49 Therefore, interventions that prevent the progression to clinically detectable HF are needed. A randomized, placebo-controlled trial investigating the impact of carvedilol for primary prevention of cardiomyopathy in childhood cancer survivors exposed to high cumulative doses of anthracyclines but with no current cardiac dysfunction is ongoing (PREVENT-HF [Carvedilol in Preventing Heart Failure in Childhood Cancer Survivors]). Despite the limitations of existing data, the American College of Cardiology and the American Heart Association recommend initiation of angiotensin-converting enzyme inhibitors or beta-blockers for subclinical cardiomyopathy regardless of etiology.50,51 As such, consensus statements from leaders in the field of cardio-oncology endorse the same practices in childhood cancer survivors.7 The same experts endorse consideration of screening for other CV late effects on a case-by-case basis because highly sensitive and specific screening modalities have not yet reached routine practice.

Given the paucity of data to guide secondary and tertiary preventative strategies for cardiomyopathy and other CV late effects, one approach has been to address other, modifiable CV risk factors, such as those aforementioned germane to the metabolic syndrome. A number of studies support the safety and modest benefits of exercise in cancer survivors.52-55 Although these results do not address the uncertain impact of intervention on the disease trajectory of these CV comorbidities in survivors, early and aggressive management has been endorsed by leading organizations.7

Many cancer treatments undoubtedly increase the risk for CV events in long-term childhood cancer survivors. Yet modifications to treatment intensity and advances in supportive care have reassuringly coincided with a reduction in CV mortality in survivors. This emerging group of aging survivors is uniquely suited for a multidisciplinary, cardio-oncology approach to care centered on early detection, risk reduction, and intervention in order to limit these late-occurring sequelae. Future efforts are needed to better understand underlying disease mechanisms and to subsequently optimize interventions in those for whom risk-reduction strategies are ineffective.

References

  1. Howlader N, Noon AM, Krapcho M, et al. SEER Cancer Statistics Review, 1975-2012 (National Cancer Institute website). 2015. Available at: http://seer.cancer.gov/csr/1975_2012/. Accessed 05/04/2016.
  2. Hudson MM, Ness KK, Nolan VG, et al. Prospective medical assessment of adults surviving childhood cancer: study design, cohort characteristics, and feasibility of the St. Jude Lifetime Cohort study. Pediatr Blood Cancer 2011;56:825-36.
  3. Hawkins MM, Lancashire ER, Winter DL, et al. The British Childhood Cancer Survivor Study: objectives, methods, population structure, response rates and initial descriptive information. Pediatr Blood Cancer 2008;50:1018-25.
  4. Kuehni CE, Rueegg CS, Michel G, et al. Cohort profile: the Swiss childhood cancer survivor study. Int J Epidemiol 2012;41:1553-64.
  5. Robison LL, Armstrong GT, Boice JD, et al. The Childhood Cancer Survivor Study: a National Cancer Institute-supported resource for outcome and intervention research. J Clin Oncol. 2009;27:2308-18.
  6. Oeffinger KC, Mertens AC, Sklar CA, et al. Chronic health conditions in adult survivors of childhood cancer. N Engl J Med 2006;355:1572-82.
  7. Lipshultz SE, Adams MJ, Colan SD, et al. Long-term cardiovascular toxicity in children, adolescents, and young adults who receive cancer therapy: pathophysiology, course, monitoring, management, prevention, and research directions: a scientific statement from the American Heart Association. Circulation 2013;128:1927-95.
  8. Robison LL, Hudson MM. Survivors of childhood and adolescent cancer: life-long risks and responsibilities. Nat Rev Cancer 2014;14:61-70.
  9. Haddy N, Diallo S, El-Fayech C, et al. Cardiac diseases following childhood cancer treatment: cohort study. Circulation 2016;133:31-8.
  10. Mulrooney DA, Yeazel MW, Kawashima T, et al. Cardiac outcomes in a cohort of adult survivors of childhood and adolescent cancer: retrospective analysis of the Childhood Cancer Survivor Study cohort. Br Med J. 2009;339:b4606.
  11. Tukenova M, Guibout C, Oberlin O, et al. Role of cancer treatment in long-term overall and cardiovascular mortality after childhood cancer. J Clin Oncol 2010;28:1308-15.
  12. van der Pal HJ, van Dalen EC, van Delden E, et al. High risk of symptomatic cardiac events in childhood cancer survivors. J Clin Oncol 2012;30:1429-37.
  13. Armstrong GT, Liu Q, Yasui Y, et al. Late mortality among 5-year survivors of childhood cancer: a summary from the Childhood Cancer Survivor Study. J Clin Oncol 2009;27:2328-38.
  14. Leger K, Slone T, Lemler M, et al. Subclinical cardiotoxicity in childhood cancer survivors exposed to very low dose anthracycline therapy. Pediatr Blood Cancer 2015;62:123-7.
  15. Vandecruys E, Mondelaers V, De Wolf D, Benoit Y, Suys B. Late cardiotoxicity after low dose of anthracycline therapy for acute lymphoblastic leukemia in childhood. J Cancer Surviv 2012;6:95-101.
  16. Chow EJ, Chen Y, Kremer LC, et al. Individual prediction of heart failure among childhood cancer survivors. J Clin Oncol 2015;33:394-402.
  17. Mulrooney DA, Armstrong GT, Huang S, et al. Cardiac outcomes in adult survivors of childhood cancer exposed to cardiotoxic therapy: a cross-sectional study. Ann Intern Med 2016;164:93-101.
  18. van der Pal HJ, van Dijk IW, Geskus RB, et al. Valvular abnormalities detected by echocardiography in 5-year survivors of childhood cancer: a long-term follow-up study. Int J Radiat Oncol Biol Phys 2015;91:213-22.
  19. Gibson TM, Ehrhardt MJ, Ness KK. Obesity and metabolic syndrome among adult survivors of childhood leukemia. Curr Treat Options Oncol 2016;17:17.
  20. Nottage KA, Ness KK, Li C, Srivastava D, Robison LL, Hudson MM. Metabolic syndrome and cardiovascular risk among long-term survivors of acute lymphoblastic leukaemia - From the St. Jude Lifetime Cohort. Br J Haematol 2014;165:364-74.
  21. van Waas M, Neggers SJ, Pieters R, van den Heuvel-Eibrink MM. Components of the metabolic syndrome in 500 adult long-term survivors of childhood cancer. Ann Oncol 2010;21:1121-6.
  22. Armstrong GT, Oeffinger KC, Chen Y, et al. Modifiable risk factors and major cardiac events among adult survivors of childhood cancer. J Clin Oncol 2013;31:3673-80.
  23. Armenian SH, Sun CL, Vase T, et al. Cardiovascular risk factors in hematopoietic cell transplantation survivors: role in development of subsequent cardiovascular disease. Blood 2012;120:45-4512.
  24. Chow EJ, Baker KS, Lee SJ, et al. Influence of conventional cardiovascular risk factors and lifestyle characteristics on cardiovascular disease after hematopoietic cell transplantation. J Clin Oncol 2014;32:191-8.
  25. Hudson MM, Neglia JP, Woods WG, et al. Lessons from the past: opportunities to improve childhood cancer survivor care through outcomes investigations of historical therapeutic approaches for pediatric hematological malignancies. Pediatr Blood Cancer 2012;58:334-43.
  26. Bhatia S, Armenian SH, Armstrong GT, et al. Collaborative research in childhood cancer survivorship: the current landscape. J Clin Oncol 2015;33:3055-64.
  27. Mauz-Korholz C, Metzger ML, Kelly KM, et al. Pediatric Hodgkin lymphoma. J Clin Oncol 2015;33:2975-85.
  28. Armstrong GT, Chen Y, Yasui Y, et al. Reduction in late mortality among 5-year survivors of childhood cancer. N Engl J Med 2016;374:833-42.
  29. O'Brien ME, Wigler N, Inbar M, et al. Reduced cardiotoxicity and comparable efficacy in a phase III trial of pegylated liposomal doxorubicin HCl (CAELYX/Doxil) versus /conventional doxorubicin for first-line treatment of metastatic breast cancer. Ann Oncol 2004;15:440-9.
  30. van Dalen EC, Michiels EM, Caron HN, Kremer LC. Different anthracycline derivates for reducing cardiotoxicity in cancer patients. Cochrane Database Syst Rev 2010:CD005006.
  31. Sieswerda E, Kremer LC, Caron HN, van Dalen EC. The use of liposomal anthracycline analogues for childhood malignancies: a systematic review. Eur J Cancer 2011;47:2000-8.
  32. Wiseman LR, Spencer CM. Dexrazoxane. A review of its use as a cardioprotective agent in patients receiving anthracycline-based chemotherapy. Drugs 1998;56:385-403.
  33. van Dalen EC, Caron HN, Dickinson HO, Kremer LC. Cardioprotective interventions for cancer patients receiving anthracyclines. Cochrane Database Syst Rev 2011:CD003917.
  34. 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.
  35. Asselin BL, Devidas M, Chen L, et al. Cardioprotection and safety of dexrazoxane in patients treated for newly diagnosed T-cell acute lymphoblastic leukemia or advanced-stage lymphoblastic non-Hodgkin lymphoma: a report of the Children's Oncology Group Randomized Trial Pediatric Oncology Group 9404. J Clin Oncol 2016;34:854-62.
  36. Chow EJ, Asselin BL, Schwartz CL, et al. Late mortality after dexrazoxane treatment: a report from the Children's Oncology Group. J Clin Oncol 2015;33:2639-45.
  37. Long-Term Follow-Up Guidelines for Survivors of Childhood, Adolescent, and Young Adult Cancers (Children's Oncology Group website). 2014. Available at: www.survivorshipguidelines.org. Accessed 04/25/2016.
  38. United Kingdom Children's Cancer Study Group Late Effects Group. Therapy based long term follow up practice statement (Children's Cacner and Leukaemia Group). 2005. Available at http://www.cclg.org.uk. Accessed 04/25/2016.
  39. Sieswerda E, Postma A, van Dalen EC, et al. The Dutch Childhood Oncology Group guideline for follow-up of asymptomatic cardiac dysfunction in childhood cancer survivors. Ann Oncol 2012;23:2191-8.
  40. Wallace WH, Thompson L, Anderson RA, Guideline Development. Long term follow-up of survivors of childhood cancer: summary of updated SIGN guidance. BMJ 2013;346:f1190.
  41. Armenian SH, Hudson MM, Mulder RL, et al. Recommendations for cardiomyopathy surveillance for survivors of childhood cancer: a report from the International Late Effects of Childhood Cancer Guideline Harmonization Group. Lancet Oncol 2015;16:e123-36.
  42. Armstrong GT, Joshi VM, Ness KK, et al. Comprehensive echocardiographic detection of treatment-related cardiac dysfunction in adult survivors of childhood cancer: results from the St. Jude Lifetime Cohort Study. J Am Coll Cardiol 2015;65:2511-22.
  43. Armstrong GT, Plana JC, Zhang N, et al. Screening adult survivors of childhood cancer for cardiomyopathy: comparison of echocardiography and cardiac magnetic resonance imaging. J Clin Oncol 2012;30:2876-84.
  44. 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. J Am Soc Echocardiogr 2014;27:911-39.
  45. Wong FL, Bhatia S, Landier W, et al. Cost-effectiveness of the Children's Oncology Group long-term follow-up screening guidelines for childhood cancer survivors at risk for treatment-related heart failure. Ann Intern Med 2014;160:672-83.
  46. Yeh JM, Nohria A, Diller L. Routine echocardiography screening for asymptomatic left ventricular dysfunction in childhood cancer survivors: a model-based estimation of the clinical and economic effects. Ann Intern Med 2014;160:661-71.
  47. Bosch X, Rovira M, Sitges M, 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;61:2355-62.
  48. Cardinale D, Colombo A, Sandri MT, et al. Prevention of high-dose chemotherapy-induced cardiotoxicity in high-risk patients by angiotensin-converting enzyme inhibition. Circulation 2006;114:2474-81.
  49. Lipshultz SE, Lipsitz SR, Sallan SE, et al. Long-term enalapril therapy for left ventricular dysfunction in doxorubicin-treated survivors of childhood cancer. J Clin Oncol 2002;20:4517-22.
  50. Hunt SA, Abraham WT, Chin MH, et al. 2009 Focused update incorporated into the ACC/AHA 2005 guidelines for the diagnosis and management of heart failure in adults: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines developed in collaboration with the International Society for Heart and Lung Transplantation. J Am Coll Cardiol 2009;53:e1-90.
  51. Hunt SA, American College of Cardiology, American Heart Association Task Force on Practice Guidelines (Writing Committee to Update the 2001 Guidelines for the Evaluation and Managemetn of Heart Failure). ACC/AHA 2005 guideline update for the diagnosis and management of chronic heart failure in the adult: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines (writing committee to update the 2001 guidelines for the evaluation and management of heart failure). J Am Coll Cardiol 2005;46:e1-82.
  52. Hoffman MC, Mulrooney DA, Steinberger J, Lee J, Baker KS, Ness KK. Deficits in physical function among young childhood cancer survivors. J Clin Oncol 2013;31:2799-2805.
  53. Huang TT, Ness KK. Exercise interventions in children with cancer: a review. Int J Pediatr 2011;2011:461512.
  54. Jones LW, Liang Y, Pituskin EN, et al. Effect of exercise training on peak oxygen consumption in patients with cancer: a meta-analysis. Oncologist 2011;16:112-20.
  55. Smith WA, Ness KK, Joshi V, Hudson MM, Robison LL, Green DM. Exercise training in childhood cancer survivors with subclinical cardiomyopathy who were treated with anthracyclines. Pediatr Blood Cancer 2013.

Keywords: Angiotensin-Converting Enzyme Inhibitors, Anthracyclines, Arrhythmias, Cardiac, Breast Neoplasms, Cardiomyopathies, Cardiotoxicity, Comorbidity, Coronary Artery Disease, Diabetes Mellitus, Dyslipidemias, Echocardiography, Heart Conduction System, Heart Failure, Hematopoietic Stem Cell Transplantation, Hypertension, Metabolic Syndrome, Myocardial Infarction, Myocytes, Cardiac, Obesity, Pericarditis, Precursor Cell Lymphoblastic Leukemia-Lymphoma, Primary Prevention, Risk Factors, Risk Reduction Behavior, Ventricular Dysfunction, Left, Pediatrics


< Back to Listings