Skip to main content

Advertisement

Log in

Unique Genetic Counseling Considerations in the Pediatric Oncology Setting

  • Genetic Counseling and Clinical Testing (BS LeRoy & N Callanan, Section Editors)
  • Published:
Current Genetic Medicine Reports Aims and scope Submit manuscript

Abstract

Genetic counseling in the pediatric oncology setting involves unique challenges. Various ethical and psychosocial considerations must be taken into account when evaluating minors for hereditary cancer syndromes. Factors that often must be addressed in this setting include issues surrounding if and when to consider genetic testing in affected individuals, limited data and guidelines regarding surveillance or management, predictive testing in at risk minor relatives, access to care, and family dynamics. This paper reviews some of the important considerations for genetic counseling in the pediatric cancer genetics setting using examples from Li–Fraumeni syndrome, constitutional mismatch repair deficiency syndrome, SDH-related hereditary paraganglioma and pheochromocytoma, hereditary retinoblastoma, and familial adenomatous polyposis. Common genetic counseling challenges include identifying appropriate patients for evaluation, helping families with genetic testing decisions and options, determining appropriate clinical management, and incorporating the future directions of hereditary cancer syndromes in pediatric care.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. NSGC. 2014 Professional Status Survey: Work Environment; 2014:6.

  2. Knapke BS, Zelley K, Nichols KE, Kohlmann W, Schiffman JD. Identification, management, and evaluation of children with cancer-predisposition syndromes. Am Soc Clin Oncol. 2012;32:576–84.

    Google Scholar 

  3. Plon SE, Nathanson K. Inherited susceptibility for pediatric cancer. Cancer J. 2005;11(4):255–67. doi:10.1097/00130404-200507000-00002.

    Article  CAS  PubMed  Google Scholar 

  4. • Banks KC, Moline JJ, Marvin ML, Newlin AC, Vogel KJ. 10 Rare Tumors That Warrant a Genetics Referral. Fam Cancer. 2013;12(1):1–18. doi:10.1007/s10689-012-9584-9. This is a recent article that highlights a few of the rare tumor types that warrant a referral for genetic consultation regardless of other clinical or family history features. Tumor types reviewed include adrenocortical carcinoma, carcinoid tumors, diffuse gastric cancer, fallopian tube/primary peritoneal cancer, leiomyosarcoma, medullary thyroid cancer, paraganglioma/pheochromocytoma, renal cell carcinoma of chromophobe, hybrid oncocytotic, or oncocytoma histology, sebaceous carcinoma, and sex cord tumors with annual tubules.

  5. • Hampel H. A practice guideline from the American College of Medical Genetics and Genomics and the National Society of Genetic Counselors: referral indications for cancer predisposition assessment. Genet Med. 2015;17(1):70. This is a practice guideline that presents a single set of comprehensive personal and family history criteria to facilitate identification and maximize appropriate referral of at-risk individuals for cancer genetic consultation. Referral for a cancer genetic consultation should be considered if a patient or any of their first-degree relatives meet any of the referral criteria outlined in this guideline.

  6. Zhukova N, Ramaswamy V, Remke M, et al. Subgroup-specific prognostic implications of TP53 mutation in medulloblastoma. J Clin Oncol. 2013;31(23):2927–35. doi:10.1200/JCO.2012.48.5052.

    Article  PubMed  Google Scholar 

  7. Holmfeldt L, Wei L, Diaz-Flores E, et al. The genomic landscape of hypodiploid acute lymphoblastic leukemia. Nat Genet. 2013;45(3):242–52. doi:10.1038/ng.2532.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  8. Hettmer S, Archer NM, Somers GR, et al. Anaplastic rhabdomyosarcoma in TP53 germline mutation carriers. Cancer. 2014;120(7):1068–75. doi:10.1002/cncr.28507.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  9. Daly M, Pilarski R, Axilbund J, et al. Genetic/familial high-risk assessment: breast and ovarian, version 1.2014. J Natl Comp Cancer Netw. 2014;12(9):1326–38.

    CAS  Google Scholar 

  10. • Ross LF, Saal HM, Al E. Technical Report: Ethical and policy issues in genetic testing and screening of children. Genet Med. 2013;15(3):234–45. This recent report provides ethical justification and empirical data regarding making decisions about whether to offer genetic testing and screening to minors. Decisions should be driven by the best interest of the child.

  11. Burt R, Cannon J, David D, et al. Colorectal cancer screening. J Natl Comp Cancer Netw. 2013;11(12):1538–75.

    CAS  Google Scholar 

  12. Riley BD, Culver JO, Skrzynia C, et al. Essential elements of genetic cancer risk assessment, counseling, and testing: updated recommendations of the National Society of Genetic Counselors. J Genet Couns. 2012;21(2):151–61. doi:10.1007/s10897-011-9462-x.

    Article  PubMed  Google Scholar 

  13. Robson ME, Storm CD, Weitzel J, Wollins DS, Offit K. American Society of Clinical Oncology policy statement update: genetic and genomic testing for cancer susceptibility. J Clin Oncol. 2010;28(5):893–901. doi:10.1200/JCO.2009.27.0660.

    Article  PubMed  Google Scholar 

  14. •• American Academy of Pediatrics. Ethical and policy issues in genetic testing and screening of children. Pediatrics. 2013;131(3):620–2. doi:10.1542/peds.2012-3680. This is the most recent policy statement from the American Academy of Pediatrics regarding the genetic testing and genetic screening of children. This policy statement represents recommendations developed collaboratively by the American Academy of Pediatrics and the American College of Medical Genetics and Genomics with respect to many of the scenarios in which genetic testing and screening can occur.

  15. Canadian Paediatric Society. Guidelines for genetic testing of healthy children-addendum: a joint statement with the Canadian College of Medical Geneticists. Paediatr Child Health. 2008;13(4):311–2.

    Google Scholar 

  16. Duncan AF, Caughy MO. Parenting style and the vulnerable child syndrome. J Child Adolesc Psychiatr Nurs. 2009;22(4):228–34. doi:10.1111/j.1744-6171.2009.00203.x.

    Article  PubMed  Google Scholar 

  17. • Kattentidt-Mouravieva AA, den Heijer M, van Kessel I, Wagner A. How harmful is genetic testing for familial adenomatous polyposis (FAP) in young children; the parents’ experience. Fam Cancer. 2014;13(3):391–9. doi:10.1007/s10689-014-9724-5. This article discusses parents’ perspectives regarding genetic testing for FAP at ages younger than 10 years. The authors contacted parents of 13 children who were tested for FAP before 10 years of age: 7 mutation-carriers and 6 non-carriers. None of the contacted parents regretted the timing of genetic testing. Genetic testing for FAP at a young age was experienced as causing no harm by parents.

  18. Levine FR, Coxworth JE, Stevenson DA, Tuohy T, Burt RW, Kinney AY. Parental attitudes, beliefs, and perceptions about genetic testing for FAP and colorectal cancer surveillance in minors. J Genet Couns. 2010;19(3):269–79. doi:10.1007/s10897-010-9285-1.

    Article  PubMed Central  PubMed  Google Scholar 

  19. Gjone H, Diseth TH, Fausa O, Nøvik TS, Heiberg A. Familial adenomatous polyposis: mental health, psychosocial functioning and reactions to genetic risk in adolescents. Clin Genet. 2011;79(1):35–43. doi:10.1111/j.1399-0004.2010.01534.x.

    Article  CAS  PubMed  Google Scholar 

  20. • Alderfer MA, Zelley K, Lindell RB, et al. Parent decision-making around the genetic testing of children for germline TP53 mutations. Cancer. 2014:1–8. doi:10.1002/cncr.29027. This is a recent study that examined how parents make decisions regarding TP53 testing for their children. Qualitative interviews with 46 parents (39 families) were analyzed to describe decision-making styles and perceived advantages and disadvantages of testing. Although empirical evidence regarding the benefits and risks of TP53 testing during childhood are lacking, the majority of parents in this study decided in favor of testing and perceived a range of advantages.

  21. •• Rowland E, Metcalfe A. Communicating inherited genetic risk between parent and child: a meta-thematic synthesis. Int J Nurs Stud. 2013;50(6):870–80. This systematic review identifies and explores the challenges faced by parents and their (non)affected or at risk children caused by the (non)disclosure of genetic risk information. Nine papers which focused on the disclosure of genetic information between parent and child (<18 years) were reviewed. This meta-analysis found that early, age appropriate disclosure can better prepare children for future considerations, contributes to effective coping strategies, and reduces parental anxieties concerning disclosure from an unwitting source.

  22. Lammens CRM, Aaronson NK, Wagner A, et al. Genetic testing in Li-Fraumeni syndrome: uptake and psychosocial consequences. J Clin Oncol. 2010;28(18):3008–14. doi:10.1200/JCO.2009.27.2112.

    Article  PubMed  Google Scholar 

  23. Wilson JM, Junger YG. Principles and practice of mass screening for disease. Of Sanit Panam. 1968;65(4):281–393.

    CAS  Google Scholar 

  24. •• Schiffman JD, Geller JI, Mundt E, Means A, Means L. Update on pediatric cancer predisposition syndromes. Pediatr Blood Cancer. 2013;1247–52. doi:10.1002/pbc. A recent workshop held at the American Society of Pediatric Hematology/Oncology (ASPHO) 2012 Annual Meeting included several interactive sessions related to specific familial cancer syndromes, genetic testing and screening, and ethical issues in caring for families with inherited cancer risk. This review highlights the workshop presentations, including a brief background about pediatric cancer predisposition syndromes and is followed by a brief summary of newly described cancer predisposition syndromes including Rhabdoid Tumor Predisposition Syndrome, Hereditary Paragangliomas and Pheochromocytoma Syndrome, and Familial Pleuropulmonary blastoma Tumor Predisposition (DICER1) Syndrome. Genetic testing and screening for pediatric cancer predisposition syndromes and ethical issues are also discussed.

  25. Hwang S-J, Lozano G, Amos CI, Strong LC. Germline p53 mutations in a cohort with childhood sarcoma: sex differences in cancer risk. Am J Hum Genet. 2003;72(4):975–83. doi:10.1086/374567.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  26. Villani A, Tabori U, Schiffman J, et al. Biochemical and imaging surveillance in germline TP53 mutation carriers with Li-Fraumeni syndrome: a prospective observational study. Lancet Oncol. 2011;12(6):559–67. doi:10.1016/S1470-2045(11)70119-X.

    Article  CAS  PubMed  Google Scholar 

  27. Lammens CRM, Bleiker EMA, Aaronson NK, et al. Regular surveillance for Li-Fraumeni Syndrome: advice, adherence and perceived benefits. Fam Cancer. 2010;9(4):647–54. doi:10.1007/s10689-010-9368-z.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  28. Raygada M. Counseling patients with succinate dehydrogenase subunit defects: genetics, preventive guidelines, and dealing with uncertainty. J Pediatr Endocrinol. 2014;27(9–10):837.

    CAS  Google Scholar 

  29. Mediouni A, Ammari S, Wassef M, et al. Malignant head/neck paragangliomas. Comparative study. Eur Ann Otorhinolaryngol Head Neck Dis. 2014;131(3):159–66. doi:10.1016/j.anorl.2013.05.003.

    Article  CAS  PubMed  Google Scholar 

  30. • Jasperson KW, Kohlmann W, Gammon A, et al. Role of rapid sequence whole-body MRI screening in SDH-associated hereditary paraganglioma families. Fam Cancer. 2014;13(2):257–65. doi:10.1007/s10689-013-9639-6. This was a single-center observational study which evaluated the results of screening in 37 SDH carriers who underwent 45 whole-body MRIs and 47 biochemical tests.

  31. • Friedman DN, Lis E, Sklar CA, et al. Whole-body magnetic resonance imaging (WB-MRI) as surveillance for subsequent malignancies in survivors of hereditary retinoblastoma: a pilot study. Pediatr Blood Cancer. 2014;61(8):1440–4. doi:10.1002/pbc.24835.Whole-body. This is a review of the results of a WB-MRI screening program in hereditary RB survivors from February 2008 – August 2012. The primary outcome was to determine the sensitivity and specificity of WB-MRI in detecting SMNs. Preliminary results suggest that annual WB-MRI surveillance detects SMN in survivors of hereditary RB, but with modest sensitivity.

  32. Wolfe Schneider K, Anguiano A, Axell L, et al. Collaboration of colorado cancer genetic counselors to integrate next generation sequencing panels into clinical practice. J Genet Couns. 2014;23(4):640–6. doi:10.1007/s10897-014-9718-3.

    Article  CAS  PubMed  Google Scholar 

  33. Scollon S, Bergstrom K, Kerstein RA, et al. Obtaining informed consent for clinical tumor and germline exome sequencing of newly diagnosed childhood cancer patients. Genome Med. 2014;6(9):69. doi:10.1186/s13073-014-0069-3.

    Article  PubMed Central  PubMed  Google Scholar 

  34. Keppler-Noreuil KM, Sapp JC, Lindhurst MJ, et al. Clinical delineation and natural history of the PIK3CA-related overgrowth spectrum. Am J Med Genet A. 2014;164A(7):1713–33. doi:10.1002/ajmg.a.36552.

    Article  PubMed  Google Scholar 

  35. Brohl AS, Solomon DA, Chang W, et al. The genomic landscape of the Ewing Sarcoma family of tumors reveals recurrent STAG2 mutation. PLoS Genet. 2014;10(7):e1004475. doi:10.1371/journal.pgen.1004475.

    Article  PubMed Central  PubMed  Google Scholar 

  36. Mai PL, Malkin D, Garber JE, et al. Li-Fraumeni syndrome: report of a clinical research workshop and creation of a research consortium. Cancer Genet. 2012;205(10):479–87. doi:10.1016/j.cancergen.2012.06.008.

    Article  PubMed Central  PubMed  Google Scholar 

  37. Kamihara J, Rana HQ, Garber JE. Germline TP53 mutations and the changing landscape of Li-Fraumeni syndrome. Hum Mutat. 2014;35(6):654–62. doi:10.1002/humu.22559.

    Article  CAS  PubMed  Google Scholar 

  38. • McBride KA, Ballinger ML, Killick E, et al. Li-Fraumeni syndrome: cancer risk assessment and clinical management. Nat Rev Clin Oncol. 2014;11(5):260–71. doi:10.1038/nrclinonc.2014.41. This article reviews the clinical implications of germline mutations in TP53 and the evidence for cancer screening and prevention strategies in individuals carrying such mutations, as well as examining the potential psychosocial implications of lifelong management for a ubiquitous cancer risk. In addition, the authors propose an evidence-based framework for the clinical management of TP53 mutation carriers and provide a platform for addressing the management of other cancer predisposition syndromes that can affect multiple organs.

  39. Jasperson KW, Burt RW. APC-associated polyposis conditions. In: Pagon RA, Bird TD, Dolan CR, Stephens K, Adam MP, editors. GeneReviews. Seattle: University of Washington; 1993.

    Google Scholar 

  40. Marees T, van Leeuwen FE, Schaapveld M, et al. Risk of third malignancies and death after a second malignancy in retinoblastoma survivors. Eur J Cancer. 2010;46(11):2052–8. doi:10.1016/j.ejca.2010.03.029.

    Article  CAS  PubMed  Google Scholar 

  41. Marees T, Moll AC, Imhof SM, de Boer MR, Ringens PJ, van Leeuwen FE. Risk of second malignancies in survivors of retinoblastoma: more than 40 years of follow-up. J Natl Cancer Inst. 2008;100(24):1771–9. doi:10.1093/jnci/djn394.

    Article  PubMed  Google Scholar 

  42. Marees T, van Leeuwen FE, de Boer MR, Imhof SM, Ringens PJ, Moll AC. Cancer mortality in long-term survivors of retinoblastoma. Eur J Cancer. 2009;45(18):3245–53. doi:10.1016/j.ejca.2009.05.011.

    Article  CAS  PubMed  Google Scholar 

  43. • Bruwer Z, Algar U, Vorster A, et al. Predictive genetic testing in children: constitutional mismatch repair deficiency cancer predisposing syndrome. J Genet Couns. 2014;23(2):147–55. doi:10.1007/s10897-013-9659-2. This report addresses the challenges that are encountered when reporting on heterozygosity in a child younger than 18 years (disclosure of carrier status and risk for Lynch syndrome), when discovered during testing for homozygosity. In addition, this report further highlights the difficulty of developing surveillance and testing guidelines, as a result of the rarity of cases and diversity of presentations in CMMRD.

  44. Kantorovich V, King KS, Pacak K. SDH-related pheochromocytoma and paraganglioma. Best Pract Res Clin Endocrinol Metab. 2010;24(3):415–24. doi:10.1016/j.beem.2010.04.001.SDH-related.

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  45. • Vasen HFA, Ghorbanoghli Z, Bourdeaut F, et al. Guidelines for surveillance of individuals with constitutional mismatch repair-deficiency proposed by the European Consortium “Care for CMMR-D” (C4CMMR-D). J Med Genet. 2014;51(5):283–93. doi:10.1136/jmedgenet-2013-102238. Recently, a new European consortium was established with the aim of improving care for patients with CMMR-D. At a workshop of this group held in Paris in June 2013, one of the issues addressed was the development of surveillance guidelines. Based on the available knowledge and the discussions at the workshop, the European consortium proposed a surveillance protocol.

Download references

Disclosures

K Wolfe Schneider declares no conflicts of interest. K Jasperson received an honorarium for ad hoc consulting from Invitae.

Human and Animal Rights and Informed Consent Statement

This article does not contain any studies with human or animal subjects performed by any of the authors.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Kami Wolfe Schneider.

Additional information

This article is part of the Topical Collection on Genetic Counseling and Clinical Testing.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wolfe Schneider, K., Jasperson, K. Unique Genetic Counseling Considerations in the Pediatric Oncology Setting. Curr Genet Med Rep 3, 65–73 (2015). https://doi.org/10.1007/s40142-015-0064-z

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40142-015-0064-z

Keywords

Navigation