1932

Abstract

Survivors of childhood cancer are at increased risk for therapy-related morbidities and mortality. Although the demographic and clinical factors predicting the risk for long-term effects of cancer therapy are well known, the impact of genetic risk for specific late effects is less clearly defined. Here, we review the extant literature and recent research describing genetic modifiers to risk for the more common late effects of childhood cancer therapy. Results of this research support the need for clinical trials that attempt to further refine risk prediction by incorporating genetic testing into existing algorithms that are primarily based on clinical and demographic factors. Confirmation of genetic predisposition, as defined by reproducibility and prospective validation, would permit therapeutic modification and discussion of individualized survivor care plans even at initial cancer diagnosis.

Loading

Article metrics loading...

/content/journals/10.1146/annurev-med-041916-124328
2018-01-29
2024-03-28
Loading full text...

Full text loading...

/deliver/fulltext/med/69/1/annurev-med-041916-124328.html?itemId=/content/journals/10.1146/annurev-med-041916-124328&mimeType=html&fmt=ahah

Literature Cited

  1. Howlader N, Noone AM, Krapcho N. 1.  et al. 2012. SEER Cancer Statistics Review, 1975–2009 (Vintage 2009 Populations) Bethesda, MD: Natl. Cancer Inst.
  2. Robison LL, Hudson MM. 2.  2014. Survivors of childhood and adolescent cancer: life-long risks and responsibilities. Nat. Rev. Cancer 14:61–70 [Google Scholar]
  3. Hudson MM, Ness KK, Gurney JG. 3.  et al. 2013. Clinical ascertainment of health outcomes among adults treated for childhood cancer. JAMA 309:2371–81 [Google Scholar]
  4. Yeh JM, Hanmer J, Ward ZJ. 4.  et al. 2016. Chronic conditions and utility-based health-related quality of life in adult childhood cancer survivors. J. Natl. Cancer Inst.108
  5. Armenian SH, Robison LL. 5.  2013. Childhood cancer survivorship: an update on evolving paradigms for understanding pathogenesis and screening for therapy-related late effects. Curr. Opin. Pediatr. 25:16–22 [Google Scholar]
  6. Gibson TM, Robison LL. 6.  2015. Impact of cancer therapy-related exposures on late mortality in childhood cancer survivors. Chem. Res. Toxicol. 28:31–37 [Google Scholar]
  7. Armstrong GT, Liu Q, Yasui Y. 7.  et al. 2009. Late mortality among 5-year survivors of childhood cancer: a summary from the Childhood Cancer Survivor Study. J. Clin. Oncol. 27:2328–38 [Google Scholar]
  8. Landier W, Armenian S, Bhatia S. 8.  2015. Late effects of childhood cancer and its treatment. Pediatr. Clin. North Am. 62:275–300 [Google Scholar]
  9. Nunez SB, Mulrooney DA, Laverdiere C, Hudson MM. 9.  2007. Risk-based health monitoring of childhood cancer survivors: a report from the Children's Oncology Group. Curr. Oncol. Rep. 9:440–52 [Google Scholar]
  10. Landier W, Bhatia S, Eshelman DA. 10.  et al. 2004. Development of risk-based guidelines for pediatric cancer survivors: the Children's Oncology Group Long-Term Follow-Up Guidelines from the Children's Oncology Group Late Effects Committee and Nursing Discipline. J. Clin. Oncol. 22:4979–90 [Google Scholar]
  11. Armenian SH, Bhatia S. 11.  2009. Chronic health conditions in childhood cancer survivors: Is it all treatment-related—or do genetics play a role?. J. Gen. Intern. Med. 24:Suppl. 2S395–400 [Google Scholar]
  12. Bhatia S. 12.  2015. Genetic variation as a modifier of association between therapeutic exposure and subsequent malignant neoplasms in cancer survivors. Cancer 121:648–63 [Google Scholar]
  13. Kremer LC, van der Pal HJ, Offringa M. 13.  et al. 2002. Frequency and risk factors of subclinical cardiotoxicity after anthracycline therapy in children: a systematic review. Ann. Oncol. 13:819–29 [Google Scholar]
  14. Risch N, Merikangas K. 14.  1996. The future of genetic studies of complex human diseases. Science 273:1516–17 [Google Scholar]
  15. Tabor HK, Risch NJ, Myers RM. 15.  2002. Candidate-gene approaches for studying complex genetic traits: practical considerations. Nat. Rev. Genet. 3:391–97 [Google Scholar]
  16. Hirschhorn JN, Daly MJ. 16.  2005. Genome-wide association studies for common diseases and complex traits. Nat. Rev. Genet. 6:95–108 [Google Scholar]
  17. Green DM, Zhu L, Wang M. 17.  et al. 2017. Effect of cranial irradiation on sperm concentration of adult survivors of childhood acute lymphoblastic leukemia: a report from the St. Jude Lifetime Cohort Study. Hum. Reprod. 32:1192–201 [Google Scholar]
  18. Wasilewski-Masker K, Seidel KD, Leisenring W. 18.  et al. 2014. Male infertility in long-term survivors of pediatric cancer: a report from the childhood cancer survivor study. J. Cancer Surviv. 8:437–47 [Google Scholar]
  19. Green DM, Liu W, Kutteh WH. 19.  et al. 2014. Cumulative alkylating agent exposure and semen parameters in adult survivors of childhood cancer: a report from the St Jude Lifetime Cohort Study. Lancet Oncol 15:1215–23 [Google Scholar]
  20. Romerius P, Stahl O, Moell C. 20.  et al. 2011. High risk of azoospermia in men treated for childhood cancer. Int. J. Androl. 34:69–76 [Google Scholar]
  21. Romerius P, Giwercman A, Moell C. 21.  et al. 2011. Estrogen receptor alpha single nucleotide polymorphism modifies the risk of azoospermia in childhood cancer survivors. Pharmacogenet. Genom. 21:263–69 [Google Scholar]
  22. Green DM, Kawashima T, Stovall M. 22.  et al. 2009. Fertility of female survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. J. Clin. Oncol. 27:2677–85 [Google Scholar]
  23. Lie Fong S, Laven JS, Hakvoort-Cammel FG. 23.  et al. 2009. Assessment of ovarian reserve in adult childhood cancer survivors using anti-Mullerian hormone. Hum. Reprod. 24:982–90 [Google Scholar]
  24. van Dorp W, van den Heuvel-Eibrink MM, Stolk L. 24.  et al. 2013. Genetic variation may modify ovarian reserve in female childhood cancer survivors. Hum. Reprod. 28:1069–76 [Google Scholar]
  25. Brooke RJ, Chemaitilly W, Wilson CL. 25.  et al. 2017. A high risk genetic profile for premature menopause in childhood cancer survivors exposed to gonadotoxic therapy: a report from the St. Jude Lifetime Cohort and Childhood Cancer Survivor Study. Presented at Int. Conf. Long-Term Complicat. Treat. Child. Adolesc. for Cancer, 15th, June 15–17, Atlanta, GA
  26. Robinson KE, Kuttesch JF, Champion JE. 26.  et al. 2010. A quantitative meta-analysis of neurocognitive sequelae in survivors of pediatric brain tumors. Pediatr. Blood Cancer 55:525–31 [Google Scholar]
  27. Ris MD, Walsh K, Wallace D. 27.  et al. 2013. Intellectual and academic outcome following two chemotherapy regimens and radiotherapy for average-risk medulloblastoma: COG A9961. Pediatr. Blood Cancer 60:1350–57 [Google Scholar]
  28. Moxon-Emre I, Bouffet E, Taylor MD. 28.  et al. 2014. Impact of craniospinal dose, boost volume, and neurologic complications on intellectual outcome in patients with medulloblastoma. J. Clin. Oncol. 32:1760–68 [Google Scholar]
  29. Moxon-Emre I, Taylor MD, Bouffet E. 29.  et al. 2016. Intellectual outcome in molecular subgroups of medulloblastoma. J. Clin. Oncol. 34:4161–70 [Google Scholar]
  30. Schreiber JE, Gurney JG, Palmer SL. 30.  et al. 2014. Examination of risk factors for intellectual and academic outcomes following treatment for pediatric medulloblastoma. Neuro-oncology 16:1129–36 [Google Scholar]
  31. Palmer SL, Armstrong C, Onar-Thomas A. 31.  et al. 2013. Processing speed, attention, and working memory after treatment for medulloblastoma: an international, prospective, and longitudinal study. J. Clin. Oncol. 31:3494–500 [Google Scholar]
  32. Brackett J, Krull KR, Scheurer ME. 32.  et al. 2012. Antioxidant enzyme polymorphisms and neuropsychological outcomes in medulloblastoma survivors: a report from the Childhood Cancer Survivor Study. Neuro-oncology 14:1018–25 [Google Scholar]
  33. Peterson CC, Johnson CE, Ramirez LY. 33.  et al. 2008. A meta-analysis of the neuropsychological sequelae of chemotherapy-only treatment for pediatric acute lymphoblastic leukemia. Pediatr. Blood Cancer 51:99–104 [Google Scholar]
  34. Krull KR, Brouwers P, Jain N. 34.  et al. 2008. Folate pathway genetic polymorphisms are related to attention disorders in childhood leukemia survivors. J. Pediatr. 152:101–5 [Google Scholar]
  35. Kamdar KY, Krull KR, El-Zein RA. 35.  et al. 2011. Folate pathway polymorphisms predict deficits in attention and processing speed after childhood leukemia therapy. Pediatr. Blood Cancer 57:454–60 [Google Scholar]
  36. Krull KR, Bhojwani D, Conklin HM. 36.  et al. 2013. Genetic mediators of neurocognitive outcomes in survivors of childhood acute lymphoblastic leukemia. J. Clin. Oncol. 31:2182–88 [Google Scholar]
  37. Krajinovic M, Robaey P, Chiasson S. 37.  et al. 2005. Polymorphisms of genes controlling homocysteine levels and IQ score following the treatment for childhood ALL. Pharmacogenomics 6:293–302 [Google Scholar]
  38. Marcoux S, Robaey P, Gahier A. 38.  et al. 2013. Role of NOS3 DNA variants in externalizing behavioral problems observed in childhood leukemia survivors. J. Pediatr. Hematol. Oncol. 35:e157–62 [Google Scholar]
  39. Green DM, Cox CL, Zhu L. 39.  et al. 2012. Risk factors for obesity in adult survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. J. Clin. Oncol. 30:246–55 [Google Scholar]
  40. Garmey EG, Liu Q, Sklar CA. 40.  et al. 2008. Longitudinal changes in obesity and body mass index among adult survivors of childhood acute lymphoblastic leukemia: a report from the Childhood Cancer Survivor Study. J. Clin. Oncol. 26:4639–45 [Google Scholar]
  41. Elmquist JK, Elias CF, Saper CB. 41.  1999. From lesions to leptin: hypothalamic control of food intake and body weight. Neuron 22:221–32 [Google Scholar]
  42. Karaman S, Ercan O, Yildiz I. 42.  et al. 2010. Late effects of childhood ALL treatment on body mass index and serum leptin levels. J. Pediatr. Endocrinol. Metab. 23:669–74 [Google Scholar]
  43. Follin C, Gabery S, Petersen A. 43.  et al. 2016. Associations between metabolic risk factors and the hypothalamic volume in childhood leukemia survivors treated with cranial radiotherapy. PLOS ONE 11:e0147575 [Google Scholar]
  44. Heo M, Leibel RL, Boyer BB. 44.  et al. 2001. Pooling analysis of genetic data: the association of leptin receptor (LEPR) polymorphisms with variables related to human adiposity. Genetics 159:1163–78 [Google Scholar]
  45. Ross JA, Oeffinger KC, Davies SM. 45.  et al. 2004. Genetic variation in the leptin receptor gene and obesity in survivors of childhood acute lymphoblastic leukemia: a report from the Childhood Cancer Survivor Study. J. Clin. Oncol. 22:3558–62 [Google Scholar]
  46. Skoczen S, Tomasik PJ, Bik-Multanowski M. 46.  et al. 2011. Plasma levels of leptin and soluble leptin receptor and polymorphisms of leptin gene –18G >A and leptin receptor genes K109R and Q223R, in survivors of childhood acute lymphoblastic leukemia. J. Exp. Clin. Cancer Res. 30:64 [Google Scholar]
  47. Sawicka-Zukowska M, Krawczuk-Rybak M, Muszynska-Roslan K. 47.  et al. 2013. Does Q223R polymorphism of leptin receptor influence on anthropometric parameters and bone density in childhood cancer survivors?. Int. J. Endocrinol. 2013:805312 [Google Scholar]
  48. Wilson CL, Liu W, Yang JJ. 48.  et al. 2015. Genetic and clinical factors associated with obesity among adult survivors of childhood cancer: a report from the St. Jude Lifetime Cohort. Cancer 121:2262–70 [Google Scholar]
  49. Wasilewski-Masker K, Kaste SC, Hudson MM. 49.  et al. 2008. Bone mineral density deficits in survivors of childhood cancer: long-term follow-up guidelines and review of the literature. Pediatrics 121:e705–13 [Google Scholar]
  50. French SA, Fulkerson JA, Story M. 50.  2000. Increasing weight-bearing physical activity and calcium intake for bone mass growth in children and adolescents: a review of intervention trials. Prev. Med. 31:722–31 [Google Scholar]
  51. Klibanski A, Adams-Campbell L, Bassford T. 51.  et al. 2001. Osteoporosis prevention, diagnosis, and therapy: NIH Consensus Development Panel. JAMA 285:6785–95 [Google Scholar]
  52. den Hoed MA, Pluijm SM, Stolk L. 52.  et al. 2016. Genetic variation and bone mineral density in long-term adult survivors of childhood cancer. Pediatr. Blood Cancer 63:2212–20 [Google Scholar]
  53. Muszynska-Roslan K, Galicka A, Sawicka M. 53.  et al. 2004. Association of collagen type I alpha1 gene polymorphism with bone density in survivors of childhood cancer—preliminary report. Rocz. Akad. Med. Bialymst. 49:Suppl. 146–48 [Google Scholar]
  54. Friedman DL, Whitton J, Leisenring W. 54.  et al. 2010. Subsequent neoplasms in 5-year survivors of childhood cancer: the Childhood Cancer Survivor Study. J. Natl. Cancer Inst. 102:1083–95 [Google Scholar]
  55. Turcotte LM, Whitton JA, Friedman DL. 55.  et al. 2015. Risk of subsequent neoplasms during the fifth and sixth decades of life in the Childhood Cancer Survivor Study cohort. J. Clin. Oncol. 33:3568–75 [Google Scholar]
  56. Zhang J, Walsh MF, Wu G. 56.  et al. 2015. Germline mutations in predisposition genes in pediatric cancer. N. Engl. J. Med. 373:2336–46 [Google Scholar]
  57. Knapke S, Nagarajan R, Correll J. 57.  et al. 2012. Hereditary cancer risk assessment in a pediatric oncology follow-up clinic. Pediatr. Blood Cancer 58:85–89 [Google Scholar]
  58. Sherborne AL, Lavergne V, Yu K. 58.  et al. 2017. Somatic and germline TP53 alterations in second malignant neoplasms from pediatric cancer survivors. Clin. Cancer Res. 23:1852–61 [Google Scholar]
  59. Ma YP, van Leeuwen FE, Cooke R. 59.  et al. 2012. FGFR2 genotype and risk of radiation-associated breast cancer in Hodgkin lymphoma. Blood 119:1029–31 [Google Scholar]
  60. Best T, Li D, Skol AD. 60.  et al. 2011. Variants at 6q21 implicate PRDM1 in the etiology of therapy-induced second malignancies after Hodgkin's lymphoma. Nat. Med. 17:941–43 [Google Scholar]
  61. Knight JA, Skol AD, Shinde A. 61.  et al. 2009. Genome-wide association study to identify novel loci associated with therapy-related myeloid leukemia susceptibility. Blood 113:5575–82 [Google Scholar]
  62. Blanco JG, Edick MJ, Hancock ML. 62.  et al. 2002. Genetic polymorphisms in CYP3A5, CYP3A4 and NQO1 in children who developed therapy-related myeloid malignancies. Pharmacogenetics 12:605–11 [Google Scholar]
  63. Bolufer P, Collado M, Barragan E. 63.  et al. 2007. Profile of polymorphisms of drug-metabolising enzymes and the risk of therapy-related leukaemia. Br. J. Haematol. 136:590–96 [Google Scholar]
  64. Woo MH, Shuster JJ, Chen C. 64.  et al. 2000. Glutathione S-transferase genotypes in children who develop treatment-related acute myeloid malignancies. Leukemia 14:232–37 [Google Scholar]
  65. Hosking FJ, Feldman D, Bruchim R. 65.  et al. 2011. Search for inherited susceptibility to radiation-associated meningioma by genomewide SNP linkage disequilibrium mapping. Br. J. Cancer 104:1049–54 [Google Scholar]
  66. Mertens AC, Mitby PA, Radloff G. 66.  et al. 2004. XRCC1 and glutathione-S-transferase gene polymorphisms and susceptibility to radiotherapy-related malignancies in survivors of Hodgkin disease. Cancer 101:1463–72 [Google Scholar]
  67. Bhatia S, Sun C-L, Hageman L. 67.  et al. 2017. Combined clinical and genetic risk prediction of subsequent central nervous system tumors in childhood cancer survivors—a report from COG ALTE03N1 Presented at Int. Conf. Long-Term Complicat. Treat. Child. Adolesc. for Cancer, 15th, June 15–17 Atlanta, GA:
  68. Xie S, Shan XF, Shang K. 68.  et al. 2014. Relevance of LIG4 gene polymorphisms with cancer susceptibility: evidence from a meta-analysis. Sci. Rep. 4:6630 [Google Scholar]
  69. Morton LM, Sampson JN, Armstrong GT. 69.  et al. 2017. Genome-wide association study to identify susceptibility loci that modify radiation-related risk for breast cancer after childhood cancer. J. Natl. Cancer Inst. 109: In press [Google Scholar]
  70. Mulrooney DA, Yeazel MW, Kawashima T. 70.  et al. 2009. Cardiac outcomes in a cohort of adult survivors of childhood and adolescent cancer: retrospective analysis of the Childhood Cancer Survivor Study cohort. BMJ 339:b4606 [Google Scholar]
  71. Lipshultz SE, Lipsitz SR, Sallan SE. 71.  et al. 2005. Chronic progressive cardiac dysfunction years after doxorubicin therapy for childhood acute lymphoblastic leukemia. J. Clin. Oncol. 23:2629–36 [Google Scholar]
  72. Lipshultz SE, Alvarez JA, Scully RE. 72.  2008. Anthracycline associated cardiotoxicity in survivors of childhood cancer. Heart 94:525–33 [Google Scholar]
  73. Aminkeng F, Bhavsar AP, Visscher H. 73.  et al. 2015. A coding variant in RARG confers susceptibility to anthracycline-induced cardiotoxicity in childhood cancer. Nat. Genet. 47:1079–84 [Google Scholar]
  74. Armenian SH, Ding Y, Mills G. 74.  et al. 2013. Genetic susceptibility to anthracycline-related congestive heart failure in survivors of haematopoietic cell transplantation. Br. J. Haematol. 163:205–13 [Google Scholar]
  75. Blanco JG, Sun CL, Landier W. 75.  et al. 2012. Anthracycline-related cardiomyopathy after childhood cancer: role of polymorphisms in carbonyl reductase genes—a report from the Children's Oncology Group. J. Clin. Oncol. 30:1415–21 [Google Scholar]
  76. Blanco JG, Leisenring WM, Gonzalez-Covarrubias VM. 76.  et al. 2008. Genetic polymorphisms in the carbonyl reductase 3 gene CBR3 and the NAD(P)H:quinone oxidoreductase 1 gene NQO1 in patients who developed anthracycline-related congestive heart failure after childhood cancer. Cancer 112:2789–95 [Google Scholar]
  77. Krajinovic M, Elbared J, Drouin S. 77.  et al. 2016. Polymorphisms of ABCC5 and NOS3 genes influence doxorubicin cardiotoxicity in survivors of childhood acute lymphoblastic leukemia. Pharmacogenomics J 16:530–35 [Google Scholar]
  78. Leger KJ, Cushing-Haugen K, Hansen JA. 78.  et al. 2016. Clinical and genetic determinants of cardiomyopathy risk among hematopoietic cell transplantation survivors. Biol. Blood Marrow Transplant 22:1094–101 [Google Scholar]
  79. Lipshultz SE, Lipsitz SR, Kutok JL. 79.  et al. 2013. Impact of hemochromatosis gene mutations on cardiac status in doxorubicin-treated survivors of childhood high-risk leukemia. Cancer 119:3555–62 [Google Scholar]
  80. Rajic V, Aplenc R, Debeljak M. 80.  et al. 2009. Influence of the polymorphism in candidate genes on late cardiac damage in patients treated due to acute leukemia in childhood. Leuk. Lymphoma 50:1693–98 [Google Scholar]
  81. Semsei AF, Erdelyi DJ, Ungvari I. 81.  et al. 2012. ABCC1 polymorphisms in anthracycline-induced cardiotoxicity in childhood acute lymphoblastic leukaemia. Cell Biol. Int. 36:79–86 [Google Scholar]
  82. Visscher H, Ross CJ, Rassekh SR. 82.  et al. 2012. Pharmacogenomic prediction of anthracycline-induced cardiotoxicity in children. J. Clin. Oncol. 30:1422–28 [Google Scholar]
  83. Visscher H, Ross CJ, Rassekh SR. 83.  et al. 2013. Validation of variants in SLC28A3 and UGT1A6 as genetic markers predictive of anthracycline-induced cardiotoxicity in children. Pediatr. Blood Cancer 60:1375–81 [Google Scholar]
  84. Visscher H, Rassekh SR, Sandor GS. 84.  et al. 2015. Genetic variants in SLC22A17 and SLC22A7 are associated with anthracycline-induced cardiotoxicity in children. Pharmacogenomics 16:1065–76 [Google Scholar]
  85. Wang X, Liu W, Sun CL. 85.  et al. 2014. Hyaluronan synthase 3 variant and anthracycline-related cardiomyopathy: a report from the children's oncology group. J. Clin. Oncol. 32:647–53 [Google Scholar]
  86. Wang X, Sun CL, Quinones-Lombrana A. 86.  et al. 2016. CELF4 Variant and anthracycline-related cardiomyopathy: a Children's Oncology Group genome-wide association study. J. Clin. Oncol. 34:863–70 [Google Scholar]
  87. Windsor RE, Strauss SJ, Kallis C. 87.  et al. 2012. Germline genetic polymorphisms may influence chemotherapy response and disease outcome in osteosarcoma: a pilot study. Cancer 118:1856–67 [Google Scholar]
  88. Aminkeng F, Ross CJ, Rassekh SR. 88.  et al. 2016. Recommendations for genetic testing to reduce the incidence of anthracycline-induced cardiotoxicity. Br. J. Clin. Pharmacol. 82:683–95 [Google Scholar]
  89. Leong SL, Chaiyakunapruk N, Lee SW. 89.  2017. Candidate gene association studies of anthracycline-induced cardiotoxicity: a systematic review and meta-analysis. Sci. Rep. 7:39 [Google Scholar]
  90. Januchowski R, Wojtowicz K, Andrzejewska M, Zabel M. 90.  2014. Expression of MDR1 and MDR3 gene products in paclitaxel-, doxorubicin- and vincristine-resistant cell lines. Biomed. Pharmacother. 68:111–17 [Google Scholar]
  91. Delacroix L, Moutier E, Altobelli G. 91.  et al. 2010. Cell-specific interaction of retinoic acid receptors with target genes in mouse embryonic fibroblasts and embryonic stem cells. Mol. Cell Biol. 30:231–44 [Google Scholar]
  92. Nagasawa K, Nagai K, Ohnishi N. 92.  et al. 2001. Contribution of specific transport systems to anthracycline transport in tumor and normal cells. Curr. Drug Metab. 2:355–66 [Google Scholar]
  93. Krishnaswamy S, Hao Q, Al-Rohaimi A. 93.  et al. 2005. UDP glucuronosyltransferase (UGT) 1A6 pharmacogenetics: II. Functional impact of the three most common nonsynonymous UGT1A6 polymorphisms (S7A, T181A, and R184S). J. Pharmacol. Exp. Ther. 313:1340–46 [Google Scholar]
  94. Biesiadecki BJ, Elder BD, Yu ZB, Jin JP. 94.  2002. Cardiac troponin T variants produced by aberrant splicing of multiple exons in animals with high instances of dilated cardiomyopathy. J. Biol. Chem. 277:50275–85 [Google Scholar]
  95. Armenian SH, Hudson MM, Chen MH. 95.  et al. 2016. Rationale and design of the Children's Oncology Group (COG) study ALTE1621: a randomized, placebo-controlled trial to determine if low-dose carvedilol can prevent anthracycline-related left ventricular remodeling in childhood cancer survivors at high risk for developing heart failure. BMC Cardiovasc. Disord. 16:187 [Google Scholar]
  96. Georgiou G, Wakefield CE, McGill BC. 96.  et al. 2016. Genetic testing for the risk of developing late effects among survivors of childhood cancer: consumer understanding, acceptance, and willingness to pay. Cancer 122:2876–85 [Google Scholar]
  97. Schuitema I, Deprez S, Van Hecke W. 97.  et al. 2013. Accelerated aging, decreased white matter integrity, and associated neuropsychological dysfunction 25 years after pediatric lymphoid malignancies. J. Clin. Oncol. 31:3378–88 [Google Scholar]
  98. Hudson MM, Oeffinger KC, Jones K. 98.  et al. 2015. Age-dependent changes in health status in the Childhood Cancer Survivor cohort. J. Clin. Oncol. 33:479–91 [Google Scholar]
  99. Ness KK, Krull KR, Jones KE. 99.  et al. 2013. Physiologic frailty as a sign of accelerated aging among adult survivors of childhood cancer: a report from the St Jude Lifetime cohort study. J. Clin. Oncol. 31:4496–503 [Google Scholar]
  100. Henderson TO, Ness KK, Cohen HJ. 100.  2014. Accelerated aging among cancer survivors: from pediatrics to geriatrics. Am. Soc. Clin. Oncol. Educ. Book 2014:e423–30 [Google Scholar]
  101. Song X, Mitnitski A, Rockwood K. 101.  2010. Prevalence and 10-year outcomes of frailty in older adults in relation to deficit accumulation. J. Am. Geriatr. Soc. 58:681–87 [Google Scholar]
  102. Ness KK, Armstrong GT, Kundu M. 102.  et al. 2015. Frailty in childhood cancer survivors. Cancer 121:1540–47 [Google Scholar]
/content/journals/10.1146/annurev-med-041916-124328
Loading
/content/journals/10.1146/annurev-med-041916-124328
Loading

Data & Media loading...

  • Article Type: Review Article
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error