As the understanding of the genetic etiology of childhood cancers increases, the need for the involvement of experts familiar with the provision of genetic counseling for this population is paramount. In October 2016, the American Association for Cancer Research organized the AACR Childhood Cancer Predisposition Workshop in which international experts in pediatric cancer predisposition met to establish surveillance guidelines for children with cancer predisposition. Identifying for whom, when, why, and how these cancer predisposition surveillance guidelines should be implemented is essential. Genetic counselors invited to this workshop provide a genetic counseling framework for oncology professionals in this article. Points of entry and recommendations regarding the provision and timing of the initial and subsequent genetic counseling sessions are addressed. The genetic counseling and testing processes are reviewed, and the psychologic impact related to surveillance is explored. Pediatric cancer genetics will continue to grow and evolve as a field, and genetic counseling services will be vital to ensure appropriate identification and management of at-risk children moving forward. Clin Cancer Res; 23(13); e91–e97. ©2017 AACR.

See all articles in the online-only CCR Pediatric Oncology Series.

The field of pediatric cancer predisposition is rapidly growing as new genes and syndromes are discovered, the phenotypes of known syndromes are expanded, and new cancer screening and genetic testing technologies are developed. With the increasing use of large gene sequencing panels, genome-wide chromosomal microarrays, and whole exome/genome sequencing, more children with cancer predisposition syndromes are being identified (1). The use of somatic tumor testing in childhood cancers is also increasing and carries the potential to identify underlying germline pathogenic variants (2, 3). This highlights the growing need for both pediatric cancer predisposition programs and genetic counselors specialized in pediatric cancer to address the unique issues and challenges of this population. Genetic counselors are experts in risk assessment and the interpretation of genetic test results, and are uniquely qualified to address the complex genetic, ethical, legal, and psychosocial issues encountered in the pediatric cancer genetics clinic.

Special issues and challenges in pediatric cancer genetic counseling include obtaining informed consent and assent for minors undergoing cancer genetic testing, determining the optimal timing of genetics referral and testing for at-risk children, providing education and counseling over time as children mature, disclosing genetic test results after the death of the child from cancer, and assisting adolescents with successful transition to adult cancer predisposition care. Given the rarity of childhood cancer predisposition syndromes, there is a paucity of guidelines for the referral, diagnosis, and management of patients with suspected or confirmed cancer predisposition syndromes (2). In addition, incidental diagnosis of cancer predisposition syndromes in children is increasing with the use of genomic testing and involves its own distinct psychosocial issues and challenges (4–7).

In this position article, we present recommendations for addressing issues specific to pediatric cancer genetics, including referral to pediatric cancer genetics clinics, pretest counseling and informed consent and assent for cancer genetic testing of children, test selection and timing of testing, posttest counseling, and psychosocial aspects of cancer surveillance for children with hereditary cancer syndromes.

Individuals with indication for hereditary cancer evaluation should be referred for genetic counseling.

There are several points of entry prompting a pediatric cancer genetics referral, including family history, physical features, high-risk tumor types, and incidental results from germline or somatic testing. The importance of family history in the identification of children affected by cancer predisposition has been well reported (8–10), yet genetics evaluation can be indicated in the absence of family history, especially in the pediatric setting. Studies assessing the utility of next-generation sequencing (NGS) technologies in the detection of pediatric cancer susceptibility have revealed that family history alone does not adequately identify children with predisposition syndromes (11–14). Family history will not be revealing in the setting of de novo variants or parental germline mosaicism. Furthermore, low penetrance, recessive inheritance, and small and/or young families can mask an inherited syndrome that is being passed through the family. Referral recommendations and tools for providers to recognize appropriate referrals are available. Positive family histories, high-genetic risk solid tumor types (see Table 1), multiple primary tumors, additional physical or clinical features, and treatment toxicity (15–18) all should be factored into the referral process. In the future, however, genetic testing may be considered for all children with cancer given the limitations of current referral and genetic testing criteria.

Table 1.

Recommendations for pediatric solid tumors (diagnosed <18 years of age) warranting referral for genetic evaluation regardless of family historya

Central and peripheral nervous system tumorsNon-CNS solid tumorsRenal and genitourinary tumors (non-rhabdoid)
Acoustic/vestibular schwannoma Adrenocortical carcinoma Botryoid-type embryonal rhabdomyosarcoma 
Atypical teratoid/rhabdoid tumor Anaplastic rhabdomyosarcoma Cystic nephroma 
Choroid plexus carcinoma Basal cell carcinoma Gonadoblastoma 
CNS hemangioblastoma Carcinoid tumor Gynandroblastoma 
Malignant nerve sheath tumors Cardiac rhabdomyoma Juvenile granulosa cell tumor 
Medulloblastoma (sonic hedgehog, desmoplastic, nodular) Ciliary body medulloepithelioma Large cell calcifying Sertoli-Leydig cell tumor (testicular) 
Neurofibroma (two or more or one plexiform neurofibroma) Gastrointestinal cancer Ovarian Sertoli-Leydig cell tumor 
Optic pathway glioma Cribriform-morular variant of papillary thyroid cancer Renal angiomyolipoma 
Pineoblastoma Desmoid tumor Renal cell carcinoma 
Pituitary blastoma Endolymphatic sac tumors (ELST) Renal sarcoma 
Subependymal giant cell astrocytoma Gastrointestinal stromal tumor (GIST) Urothelial cell carcinoma 
 Hepatoblastoma Wilms tumor (bilateral/multifocal) 
 Malignant rhabdoid tumor  
 Medullary thyroid cancer  
 Melanoma  
 Multinodular goiter  
 Myxoma  
 Nasal chondromesenchymal hamartoma  
 Osteosarcoma (dx <10 y)  
 Parathyroid carcinoma  
 Pheochromocytoma/paraganglioma  
 Pleuropulmonary blastoma  
 Retinal hemangioblastoma  
 Retinoblastoma  
Central and peripheral nervous system tumorsNon-CNS solid tumorsRenal and genitourinary tumors (non-rhabdoid)
Acoustic/vestibular schwannoma Adrenocortical carcinoma Botryoid-type embryonal rhabdomyosarcoma 
Atypical teratoid/rhabdoid tumor Anaplastic rhabdomyosarcoma Cystic nephroma 
Choroid plexus carcinoma Basal cell carcinoma Gonadoblastoma 
CNS hemangioblastoma Carcinoid tumor Gynandroblastoma 
Malignant nerve sheath tumors Cardiac rhabdomyoma Juvenile granulosa cell tumor 
Medulloblastoma (sonic hedgehog, desmoplastic, nodular) Ciliary body medulloepithelioma Large cell calcifying Sertoli-Leydig cell tumor (testicular) 
Neurofibroma (two or more or one plexiform neurofibroma) Gastrointestinal cancer Ovarian Sertoli-Leydig cell tumor 
Optic pathway glioma Cribriform-morular variant of papillary thyroid cancer Renal angiomyolipoma 
Pineoblastoma Desmoid tumor Renal cell carcinoma 
Pituitary blastoma Endolymphatic sac tumors (ELST) Renal sarcoma 
Subependymal giant cell astrocytoma Gastrointestinal stromal tumor (GIST) Urothelial cell carcinoma 
 Hepatoblastoma Wilms tumor (bilateral/multifocal) 
 Malignant rhabdoid tumor  
 Medullary thyroid cancer  
 Melanoma  
 Multinodular goiter  
 Myxoma  
 Nasal chondromesenchymal hamartoma  
 Osteosarcoma (dx <10 y)  
 Parathyroid carcinoma  
 Pheochromocytoma/paraganglioma  
 Pleuropulmonary blastoma  
 Retinal hemangioblastoma  
 Retinoblastoma  

Abbreviations: CNS, central nervous system; dx, diagnosis; y, years.

aOf note, these lists are not comprehensive of indications that may warrant automatic consideration for referrals. Referral practices may vary, and indications may change with time

Many non-oncologic physical findings should prompt referral for cancer predisposition evaluation. Examples include the classic lip pigmentation associated with Peutz–Jeghers syndrome; >3 café au lait macules associated with neurofibromatosis type 1 and biallelic mismatch repair deficiency; and multiple, bilateral congenital hypertrophy of the retinal pigment epithelium associated with familial adenomatous polyposis (16). In addition, incidental findings identified on germline genetic testing performed for other clinical reasons may initiate referral. This includes the disruption of cancer predisposition genes detected by chromosomal microarray technology (5–7) and secondary findings detected by exome sequencing (4). Referrals generated by non-oncologic physical findings or incidental findings on a genetic test can come with unique challenges in the discussion with both parents and clinicians given the unanticipated nature of the findings and their association with cancer predisposition.

Finally, the uptake of precision medicine and molecular tumor analysis by NGS increases the possibility of identifying somatic variants within cancer predisposition genes (19). Oncologists are increasingly challenged with interpreting whether a reported variant is an isolated somatic change or if it indeed represents germline susceptibility. A referral to the cancer genetics clinic for additional interpretation may be indicated when a variant identified on tumor testing is suspected to represent an underlying germline mutation (3).

Referral for genetic counseling should be made at the time of tumor diagnosis so that genetic testing, if indicated, can be completed in a timely manner.

DNA banking should be considered if the genetic counseling appointment may occur after the initiation of treatment or if the child has a poor prognosis.

Genetic counselors or other genetics professionals are integral members to the patient's care team, and we recommend that referrals are made at the time of tumor diagnosis. The genetic counselor or genetic professional in consultation with the oncologist can decide when to see the family based on the child's prognosis, treatment plan, and psychologic well-being of the family. As the genetic counseling appointment may not arise immediately, DNA banking should be considered if available and if testing may be more difficult once treatment has started (i.e., due to lowered lymphocyte counts following chemotherapy or difficulty of obtaining a germline sample after a patient has undergone donor bone marrow transplant) or if the patient has a poor prognosis. In addition, testing a child postmortem may be important for the care of relatives but is unfeasible if no DNA is banked. Every effort to organize genetic counseling before the child passes away should be made, as the cost of postmortem genetic testing often falls to the family and, therefore, may not be feasible even if a sample is available.

Children and their parents/guardians should meet with a genetic counselor or other qualified professional prior to undergoing germline cancer genetic testing.

Informed consent should be obtained from the parent(s)/guardian(s) of children undergoing germline cancer genetic testing. Verbal or written informed assent should be obtained from older children and adolescents undergoing germline cancer genetic testing and should be documented in the medical record. Younger children undergoing testing should also be included in the pretest discussions in a manner that is appropriate for their age and understanding.

Cancer genetic counseling is meant to increase family understanding of testing options, ensure that the most appropriate test(s) is ordered, allow for informed decision making, and ensure that families are prepared for the outcomes of testing. Genetic testing for adult-onset conditions in children should not be undertaken without medical and/or psychosocial justification and a discussion with family members, although pathogenic variants in adult-onset cancer genes may be revealed by large panels, tumor-based testing, or whole exome/genome analysis as an incidental finding (20). Genetic testing without genetic counseling has been linked with a variety of negative testing outcomes including a lack of informed decision making around testing; ordering of costly, unnecessary genetic testing; misinterpretation of genetic test results; inappropriate or inadequate medical management; violations of ethical standards; and adverse psychosocial outcomes (21–23). The informed consent process should include a discussion about the implications of the results to the patient (including discussion of the surveillance protocol, or lack thereof, for the child if found to have a hereditary cancer predisposition syndrome), psychosocial and ethical considerations, confidentiality and privacy concerns (which varies with the laws of different countries), the implications of the results for other relatives, logistics including a plan for disclosing results, and the option of deferring or declining testing (15, 24–26).

The trend toward openly discussing treatment plans and prognosis with children who have cancer has mirrored the trend toward involving children in a developmentally appropriate manner in pretest counseling discussions. Obtaining verbal or written assent from older children and adolescents further ensures that they have a voice in the decision-making process, allows them to have their fears or misconceptions addressed, and helps prepare them to learn their test results (27, 28).

Despite concerns about potentially exacerbating parental and patient anxiety, genetic counseling has been found to allay fears and to empower families by providing fact-based risk information and potential options for proactive risk reduction and/or early detection strategies if a gene mutation is identified (29, 30).

Genetic testing is an important tool for identifying individuals with hereditary cancer predisposition syndromes. Test selection should be undertaken with input from a genetic counselor or other qualified professional.

The transition to NGS platforms is the most significant recent development in genetic testing. This technology allows for the analysis of multiple genes or even whole genomes at moderately low cost and increases the ability to efficiently screen children with cancer for genetic predisposition.

When selecting a laboratory and a specific genetic test, it is important to consider inclusion of the gene(s) of interest, testing methodology and validation, variant interpretation and reinterpretation practices, cost, turnaround time, and the laboratory's policies regarding data sharing. The American College of Medical Genetics and Genomics (ACMG) proposed a five-tier variant classification structure that recommends reporting variants discovered during clinical testing as benign, likely benign, uncertain significance, likely pathogenic, and pathogenic (31). Although this system provides the clinician with insight into the classification decision, further tailoring is often needed to apply the findings from genetic testing to the clinical context of the patient. Testing with multigene panels and exome/genome sequencing results in a higher yield of both pathogenic variants and variants of uncertain significance (VUS; ref. 32) than with single-gene testing (Table 2).

Table 2.

Genetic testing approaches

Type of genetic testingLevel of analysisWhen usedAdvantagesLimitations/disadvantages
Familial variant testing of a single gene Analysis of a single variant Known familial variant Least expensive May miss a cancer-predisposing mutation in a different gene not previously identified in the individual or family (a rare circumstance) 
   Very accurate Testing should be performed in the same lab that identified the mutation in a family member, or a positive control sample should be sent 
   Results are definitive  
Single genea Sanger sequencing or NGS coding sequence and intron/exon borders of the selected gene Phenotype fits a known cancer syndrome caused by one or a few genes Highly specific May not identify certain types of variants (i.e., deep intronic variants) 
 +/− copy number analysis for intragenic deletions/duplications  Lower risk for VUS May miss variants in more rarely associated genes not tested with this approach 
    Additional testing may be required if initial testing is negative or inconclusive 
Multigene panela NGS of coding sequence and intron/exon borders of the selected genes Phenotype that does not clearly fit with a specific syndrome Increases the chance of identifying a causative variant May identify incidental findings/VUS 
 May include targeted panel of genes associated with specific cancer type or broad panel of genes associated with cancer predisposition Patients suspected to have a condition that can be associated with variants in multiple genes (e.g., pheochromocytoma/paraganglioma) Cost-effective Depending on nature of the panel, may identify moderate-risk genes for which limited surveillance recommendations may be available 
 +/− copy number analysis for intragenic deletions/duplications    
WES/WGSa NGS technology to evaluate coding areas (exons and intron/exon borders only) or entire genome (exons and introns) Patients with unclear cancer phenotypes or multisystem phenotypes Cost of WES/WGS is approaching the cost of multigene panels and may be the most cost-effective approach in the future Not all platforms will have adequate coverage of the genes of greatest interest for a patient 
  Patients with negative/uninformative results on prior single-gene or multigene testing Data can be reanalyzed as new genetic associations are made Sequencing technology may miss intragenic copy number variants 
  Research settings identify novel genetic associations  Greatest chance for incidental findings/VUS 
    Ethical considerations regarding possible identification of variants associated with adulthood disease risk 
    Challenges in storing and reinterpreting data and communicating implications over time 
    Increased time to obtain informed consent and determine preference for possible result types 
    NGS technologies not consistently available as a clinically certified test 
SNP Microarray Genome-wide microarray to detect large chromosomal deletions/duplications Patients with developmental delays/intellectual disability/autism spectrum disorder and/or congenital abnormalities that may be indicative of a chromosomal deletion/duplication Identifies large deletions or duplications that may be missed by single-gene, multigene, or WES/WGS testing Chance of incidental findings (including consanguinity)/VUS 
  Patients identified by single-gene or multigene testing to have a whole gene deletion Defines size of deletion or duplication and genes included Will not identify balanced chromosomal rearrangements 
    Will not identify sequence alterations or deletions/duplications below a certain size 
Type of genetic testingLevel of analysisWhen usedAdvantagesLimitations/disadvantages
Familial variant testing of a single gene Analysis of a single variant Known familial variant Least expensive May miss a cancer-predisposing mutation in a different gene not previously identified in the individual or family (a rare circumstance) 
   Very accurate Testing should be performed in the same lab that identified the mutation in a family member, or a positive control sample should be sent 
   Results are definitive  
Single genea Sanger sequencing or NGS coding sequence and intron/exon borders of the selected gene Phenotype fits a known cancer syndrome caused by one or a few genes Highly specific May not identify certain types of variants (i.e., deep intronic variants) 
 +/− copy number analysis for intragenic deletions/duplications  Lower risk for VUS May miss variants in more rarely associated genes not tested with this approach 
    Additional testing may be required if initial testing is negative or inconclusive 
Multigene panela NGS of coding sequence and intron/exon borders of the selected genes Phenotype that does not clearly fit with a specific syndrome Increases the chance of identifying a causative variant May identify incidental findings/VUS 
 May include targeted panel of genes associated with specific cancer type or broad panel of genes associated with cancer predisposition Patients suspected to have a condition that can be associated with variants in multiple genes (e.g., pheochromocytoma/paraganglioma) Cost-effective Depending on nature of the panel, may identify moderate-risk genes for which limited surveillance recommendations may be available 
 +/− copy number analysis for intragenic deletions/duplications    
WES/WGSa NGS technology to evaluate coding areas (exons and intron/exon borders only) or entire genome (exons and introns) Patients with unclear cancer phenotypes or multisystem phenotypes Cost of WES/WGS is approaching the cost of multigene panels and may be the most cost-effective approach in the future Not all platforms will have adequate coverage of the genes of greatest interest for a patient 
  Patients with negative/uninformative results on prior single-gene or multigene testing Data can be reanalyzed as new genetic associations are made Sequencing technology may miss intragenic copy number variants 
  Research settings identify novel genetic associations  Greatest chance for incidental findings/VUS 
    Ethical considerations regarding possible identification of variants associated with adulthood disease risk 
    Challenges in storing and reinterpreting data and communicating implications over time 
    Increased time to obtain informed consent and determine preference for possible result types 
    NGS technologies not consistently available as a clinically certified test 
SNP Microarray Genome-wide microarray to detect large chromosomal deletions/duplications Patients with developmental delays/intellectual disability/autism spectrum disorder and/or congenital abnormalities that may be indicative of a chromosomal deletion/duplication Identifies large deletions or duplications that may be missed by single-gene, multigene, or WES/WGS testing Chance of incidental findings (including consanguinity)/VUS 
  Patients identified by single-gene or multigene testing to have a whole gene deletion Defines size of deletion or duplication and genes included Will not identify balanced chromosomal rearrangements 
    Will not identify sequence alterations or deletions/duplications below a certain size 

Abbreviation: WES/WGS, whole exome sequencing/whole genome sequencing.

aNGS-based testing has the ability to detect mosaicism due to a de novo mutation.

When pathogenic or likely pathogenic variants are identified, that information can be used to tailor management and perform targeted testing for at-risk family members. Typically, the identification of a VUS should not influence medical management decisions, and relatives should not be tested for these for the purposes of risk assessment and clinical management. However, variant tracking may be performed for informative relatives to assist with variant interpretation. The family and treating oncologist should be aware that interpretation of this variant may change over time. Periodic rereferrals for genetic counseling are recommended for individuals with a VUS or negative/uninformative genetic test results but continued suspicion for a cancer predisposition syndrome. Periodic reevaluation for these patients allows families to be updated on new information and changes in family history to be incorporated into the assessment.

Although NGS testing of multiple genes increases identification of pathogenic variants, many individuals with features suggestive of familial risk will not have an identifiable pathogenic variant. The sensitivity of genetic testing for identifying the underlying mutation varies quite significantly with >95% for some conditions (e.g., von Hippel-Lindau syndrome) and less than 50% for juvenile polyposis. For these families, risk assessment and management will be based on empiric data and clinical judgment.

Genetic counseling for children diagnosed with a hereditary cancer syndrome should be an ongoing process to reinforce information, address age-specific risks and management, ensure information is disseminated to at-risk relatives and, evaluate needs for additional psychologic support. We recommend that in addition to genetic counseling at the time of initial assessment, children diagnosed with hereditary cancer syndromes should revisit genetic counseling in the mid- to late teenage years and again at times of family planning.

Incorporating cancer predisposition test results into lifelong cancer risk management and understanding of reproductive implications is particularly challenging when this information is initially received in early childhood (2). Children's psychologic needs may also change with age as they become more aware of the implications of having a hereditary cancer risk and as they reach an age at which physical features or cancers may begin to manifest. Evaluation of psychologic and emotional support needs for children and their caregivers is imperative, and processes for referring to support services should be in place (28). Revisiting genetic counseling in the mid- to late teenage years and again at the time of the family planning is also vital to ensure that patients understand their diagnoses, medical management recommendations, and reproductive risks and options.

Discussion of results and their implications with at-risk relatives should be encouraged and facilitated. When a pathogenic variant is identified, the inheritance pattern of the condition will determine which relatives are at risk and in need of testing. For autosomal dominant cancer syndromes, first-degree relatives have an up to 50% risk of having the pathogenic variant and the associated increased cancer risks. Disseminating information to relatives can be a daunting process for families also dealing with a cancer diagnosis. Genetic counselors can provide guidance and assistance in this process.

Risks for future children in the family should be addressed, and discussion should include options for preimplantation genetic diagnosis; prenatal diagnosis; testing children after birth; adoption; and/or utilizing donor eggs, sperm, or embryos.

Finally, the results of genetic testing should be incorporated into the management planning of the patient and at-risk relatives. Even in the context of a clearly pathogenic variant, surveillance recommendations may need to be tailored on the basis of personal and family history and genotype–phenotype correlations. In the absence of an identified genetic cause for the patient's personal and/or family history of cancer, families and referring physicians should be encouraged to follow up periodically with their local cancer genetics clinic to be informed of new genetic testing options and/or surveillance recommendations (see Table 3).

Table 3.

Circumstances in which surveillance recommendations should be followed

Pathogenic variant detected in cancer-predisposing gene 
Clinical criteria met for a syndrome, but genetic testing not pursued 
Clinical criteria met for a syndrome, but no pathogenic variant detected 
50% risk (e.g., parent/sibling with syndrome), but genetic testing not (yet) pursued 
50% risk (e.g., parent/sibling with syndrome), but informative genetic testing not available 
Pathogenic variant detected in cancer-predisposing gene 
Clinical criteria met for a syndrome, but genetic testing not pursued 
Clinical criteria met for a syndrome, but no pathogenic variant detected 
50% risk (e.g., parent/sibling with syndrome), but genetic testing not (yet) pursued 
50% risk (e.g., parent/sibling with syndrome), but informative genetic testing not available 

The family, in concert with multidisciplinary team, should be involved in decision making and coordination of tumor surveillance plans and be monitored for psychosocial impact of such plans on their overall quality of life.

As surveillance protocols for an increasing number of cancer-predisposing conditions are established, the importance of balancing the scientific merit with the psychologic burden needs to be carefully studied on a large scale. The efficacy of many cancer surveillance protocols for rare cancer syndromes has not been determined for many syndromes, although some data are beginning to accumulate (33–36), and for many pediatric cancers, no established surveillance exists. Clinicians and families must weigh the desire for early detection with possible negative outcomes of surveillance, which include risks from invasive medical procedures, procedures requiring sedation or general anesthesia, false positive results, incidental findings, cost, and psychologic burden.

Cancer-related distress is associated with impaired quality of life, reduced satisfaction with care, and worse overall survival (4, 37). At this time, only a few small studies have attempted to address the psychosocial factors associated with tumor surveillance in pediatric-onset cancer syndromes (38–45). A multidisciplinary approach with clear surveillance recommendations and support from providers has been demonstrated to improve adherence to surveillance protocols (39, 43).

“Scanxiety” (46) has been described in cancer patients and refers to the often-debilitating anxiety patients experience in the period surrounding imaging studies to identify cancer. Scanxiety can be compounded by a fear of future tumor development amongst cancer survivors. Undergoing imaging and/or laboratory studies as part of a cancer surveillance protocol may also cause potentially unnecessary worry for individuals with cancer predisposition syndromes (43).

Some patients have fears and anxiety related to specific aspects of surveillance such as confined spaces in an MRI, anesthesia, or blood draws. Uncertainty can arise when tumor surveillance reveals ambiguous findings, necessitating additional imaging or biopsy. Certain types of screening, such as whole-body MRI, may have high rates of incidental or false positive findings (34–36). Ultimately families can become frustrated with the lack of information regarding optimal surveillance protocols, high frequency of exams, inconclusive outcomes, and the challenges and costs of coordinating complex specialty care. Adolescents and young adults, who are more likely to be uninsured, have less stable employment and less social support and may be at greatest risk for discontinuing screening, often as they are reaching an age where cancer risk may increase. Detailed discussions with parents and teens about these issues of access to health care should be started in the later years of high school to decrease the possibility of a lapse in needed cancer surveillance.

Conversely, tumor surveillance plans can provide psychologic benefits. Families may feel a sense of empowerment and control with a proactive surveillance approach (38). A negative test can provide relief, especially in an organ with high cancer risk or for a tumor type known to be in the family. Relationships formed over time with the surveillance team can generate a sense of trust and support, especially when a new tumor diagnosis is made. Understanding the impact of intensive, long-term cancer surveillance programs on families is necessary to ensure adherence and positive psychologic adjustment.

Several unique situations may arise which require careful consideration by the family and care team. Examples include the continuation of surveillance for other malignancies when a tumor is diagnosed or when a known tumor progresses; modification of a surveillance plan for children with intellectual disability, developmental delays, or other comorbid conditions; and the ideal timing and preparedness training for transferring adolescents to adult tumor surveillance providers.

The model for a referral to cancer genetics programs is an ever-changing landscape. This is particularly evident in pediatric settings. Traditionally, practitioners often relied on a significant family history of cancer to evaluate the risk for the presence of a hereditary cancer predisposition syndrome in the family. Family history is still an important tool in the initiation of some referrals to cancer genetics programs and should be taken at the initial tumor diagnosis and reviewed periodically (47). However, mounting evidence supports a review of this historical referral paradigm (11–14). Relying on family history alone could mean missing an early diagnosis and, therefore, the benefits of evaluation, diagnosis, and surveillance for the patient and family members.

If a referral for genetic counseling is indicated, we recommend this take place at diagnosis even if the genetic counseling appointment is deferred. The genetics professional and oncology team can decide on the best time for the evaluation to occur. At a minimum, the genetic counselor can help to facilitate DNA banking, when possible, before the initiation of treatment or if the child has a poor prognosis. Genetic counseling empowers children and families to be involved in the decision-making process surrounding genetic testing and tumor management.

When a hereditary cancer predisposition syndrome is identified, families are faced with the distress and anxiety of a new diagnosis of cancer in their child, along with information about increased risks for developing future cancers in the child and a risk for other family members. This distress can be mitigated in part by comprehensive pre- and posttest genetic counseling.

Genetic counselors and other genetic professionals will increasingly be integral members of the pediatric oncology team, providing expertise to help diagnose hereditary cancer syndromes, addressing the unique challenges associated with genetic evaluation in childhood, and helping families use information about cancer risk to plan screening and prevention strategies. Genetic counseling should be an ongoing process, and we recommend that patients receive repeat genetic counseling in their mid- to late teenage years and again when family planning. The field of hereditary cancer predisposition continues to evolve, and iterative genetic counseling will be vital to ensure that patients are kept abreast of new developments and options as they arise.

W.K. Kohlmann reports receiving commercial research grants from Myriad. No potential conflicts of interest were disclosed by the other authors.

1.
Fecteau
H
,
Vogel
KJ
,
Hanson
K
,
Morrill-Cornelius
S
. 
The evolution of cancer risk assessment in the era of next generation sequencing
.
J Genet Couns
2014
;
23
:
633
9
.
2.
Wolfe-Schneider
K
,
Jasperson
K
. 
Unique genetic counseling considerations in the pediatric oncology setting
.
Curr Genet Med Rep
2015
;
3
:
65
.
3.
Jain
R
,
Savage
MJ
,
Forman
AD
,
Mukherji
R
,
Hall
MJ
. 
The relevance of hereditary cancer risks to precision oncology: what should providers consider when conducting tumor genomic profiling?
J Natl Compr Canc Netw
2016
;
14
:
795
806
.
4.
Johnston
JJ
,
Rubinstein
WS
,
Facio
FM
,
Ng
D
,
Singh
LN
,
Teer
JK
, et al
Secondary variants in individuals undergoing exome sequencing: screening of 572 individuals identifies high-penetrance mutations in cancer-susceptibility genes
.
Am J Hum Genet
2012
;
91
:
97
108
.
5.
Boone
PM
,
Soens
ZT
,
Campbell
IM
,
Stankiewicz
P
,
Cheung
SW
,
Patel
A
, et al
Incidental copy-number variants identified by routine genome testing in a clinical population
.
Genet Med
2013
;
15
:
45
54
.
6.
Hamm
JA
,
Mikhail
FM
,
Hollenbeck
D
,
Farmer
M
,
Robin
NH
. 
Incidental detection of cancer predisposition gene copy number variations by array comparative genomic hybridization
.
J Pediatr
2014
;
165
:
1057
9
.
7.
Duong
BT
,
Savarirayan
R
,
Winship
I
. 
Incidental diagnosis of HLRCC following investigation for Asperger Syndrome: actionable and actioned
.
Fam Cancer
2016
;
15
:
25
9
.
8.
Knapke
S
,
Zelley
K
,
Nichols
KE
,
Kohlmann
W
,
Schiffman
JD
. 
Identification, management, and evaluation of children with cancer-predisposition syndromes
.
Am Soc Clin Oncol Educ Book
2012
:
576
84
.
9.
Knapke
S
,
Nagarajan
R
,
Correll
J
,
Kent
D
,
Burns
K
. 
Hereditary cancer risk assessment in a pediatric oncology follow-up clinic
.
Pediatr Blood Cancer
2012
;
58
:
85
9
.
10.
Curtin
K
,
Smith
KR
,
Fraser
A
,
Pimentel
R
,
Kohlmann
W
,
Schiffman
JD
. 
Familial risk of childhood cancer and tumors in the Li-Fraumeni spectrum in the Utah Population Database: implications for genetic evaluation in pediatric practice
.
Int J Cancer
2013
;
133
:
2444
53
.
11.
Parsons
DW
,
Roy
A
,
Yang
Y
,
Wang
T
,
Scollon
S
,
Bergstrom
K
, et al
Diagnostic yield of clinical tumor and germline whole-exome sequencing for children with solid tumors
.
JAMA Oncol
2016
;
2
:
616
24
.
12.
Zhang
J
,
Walsh
MF
,
Wu
G
,
Edmonson
MN
,
Gruber
TA
,
Easton
J
, et al
Germline mutations in predisposition genes in pediatric cancer
.
N Engl J Med
2015
;
373
:
2336
46
.
13.
Mody
RJ
,
Wu
YM
,
Lonigro
RJ
,
Cao
X
,
Roychowdhury
S
,
Vats
P
, et al
Integrative clinical sequencing in the management of refractory or relapsed cancer in youth
.
JAMA
2015
;
314
:
913
25
.
14.
Plon
SE
,
Wheeler
DA
,
Strong
LC
,
Tomlinson
GE
,
Pirics
M
,
Meng
Q
, et al
Identification of genetic susceptibility to childhood cancer through analysis of genes in parallel
.
Cancer Genet
2011
;
204
:
19
25
.
15.
Hampel
H
,
Bennett
RL
,
Buchanan
A
,
Pearlman
R
,
Wiesner
GL
,
Guideline Development Group, American College of Medical Genetics and Genomics Professional Practice and Guidelines Committee and National Society of Genetic Counselors Practice Guidelines Committee
. 
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
:
70
87
.
16.
Jongmans
MC
,
Loeffen
JL
,
Waanders
E
,
Hoogerbrugge
PM
,
Ligtenberg
MJ
,
Kuiper
RP
, et al
Recognition of genetic predisposition in pediatric cancer patients: an easy-to-use selection tool
.
Eur J Genet Med
2016
;
59
:
116
25
.
17.
Ripperger
T
,
Bielack
SS
,
Borkhardt
A
,
Brecht
IB
,
Burkhardt
B
,
Calaminus
G
et al 
Childhood cancer predisposition syndromes–a concise review and recommendations by the Cancer Predisposition Working Group of the Society for Pediatric Oncology and Hematology
.
Am J Med Genet A
2017
.
18.
Scollon
S
,
Knoth
A
,
Thomas
M
,
Turner
J
,
Wolfe-Schneider
K
. 
A comprehensive review of pediatric tumors and associated cancer predisposition syndromes
.
J Genet Counsel
2017
;
26
:
387
434
.
19.
Catenacci
DV
,
Amico
AL
,
Nielsen
SM
,
Geynisman
DM
,
Rambo
B
,
Carey
GB
, et al
Tumor genome analysis includes germline genome: are we ready for surprises?
Int J Cancer
2015
;
136
:
1559
67
.
20.
Green
RC
,
Berg
JS
,
Grody
WW
,
Kalia
SS
,
Korf
BR
,
Martin
CL
, et al
ACMG recommendations for reporting of incidental findings in clinical exome and genome sequencing
.
Genet Med
2013
;
15
:
565
74
.
21.
Bensend
TA
,
Veach
PM
,
Niendorf
KB
. 
What's the harm? Genetic counselor perceptions of adverse effects of genetics service provision by non-genetics professionals
.
J Genet Couns
2014
;
23
:
48
63
.
22.
Brierley
KL
,
Blouch
E
,
Cogswell
W
,
Homer
JP
,
Pencarinha
D
,
Stanislaw
CL
, et al
Adverse events in cancer genetic testing: medical, ethical, legal, and financial implications
.
Cancer J
2012
;
18
:
303
309
.
23.
Miller
CE
,
Krautscheid
P
,
Baldwin
EE
,
Tvrdik
T
,
Openshaw
AS
,
Hart
K
, et al
Genetic counselor review of genetic test orders in a reference laboratory reduces unnecessary testing
.
Am J Med Genet
2014
;
164A
:
1094
1101
.
24.
Robson
ME
,
Bradbury
AR
,
Arun
B
,
Domchek
SM
,
Ford
JM
,
Hampel
HL
, et al
ASCO policy statement update: genetic and genomic testing for cancer susceptibility
.
J Clin Oncol
2015
;
33
:
3660
7
.
25.
Botkin
JR
,
Belmont
JW
,
Berg
JS
,
Berkman
BE
,
Bombard
Y
,
Holm
IA
, et al
Points to consider: Ethical, legal and psychosocial implications of genetic testing in children and adolescents
.
Am J Hum Genet
2015
;
97
:
6
21
.
26.
Kesserwan
C
,
Ross
LF
,
Bradbury
AR
,
Nichols
KE
. 
The advantages and challenges of testing children for heritable predisposition to cancer
.
Am Soc Clin Oncol Educ Book
2016
;
35
:
251
69
.
27.
Patenaude
AF
,
Tung
N
,
Ryan
PD
,
Ellisen
LW
,
Hewitt
L
,
Schneider
KA
, et al
Young adult daughters of BRCA1/2-positive mothers: What do they know about hereditary cancer and how much do they worry?
Psychooncology
2013
;
22
:
2024
31
.
28.
Patenaude
AF
,
Schneider
KA
. 
Issues arising in psychological consultations to help parents talk to minor and young adult children about their cancer genetic test result: a guide to providers
.
J Genet Couns
2017
;
26
:
251
60
.
29.
Nelson
HD
,
Fu
R
,
Goddard
K
,
Mitchell
JP
,
Okinaka-Hu
L
,
Pappas
M
, et al
Risk assessment, genetic counseling, and genetic testing for BRCA-related cancer: systematic review to update the U.S. Preventive Services Task Force Recommendation [Internet]
.
Evidence syntheses no. 101. AHRQ publication no. 12-05164-EF-1
.
Rockville (MD)
:
Agency for Healthcare Research and Quality
; 
2013
.
30.
Wakefield
CE
,
Hanlon
LV
,
Tucker
KM
,
Patenaude
AF
,
Signorelli
C
,
McLoone
JK
, et al
The psychological impact of genetic information on children: a systematic review
.
Genet Med
2016
;
18
:
755
62
.
31.
Richards
S
,
Aziz
N
,
Bale
S
,
Bick
D
,
Das
S
,
Gastier-Foster
J
, et al
Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology
.
Genet Med
2015
;
17
:
405
24
.
32.
Plon
SE
,
Eccles
DM
,
Easton
D
,
Foulkes
WD
,
Genuardi
M
,
Greenblatt
MS
, et al
Sequence variant classification and reporting: recommendations for improving the interpretation of cancer susceptibility genetic test results
.
Hum Mutat
2008
;
29
:
1282
91
.
33.
Villani
A
,
Shore
A
,
Wasserman
JD
,
Stephens
D
,
Kim
RH
,
Druker
H
, et al
Biochemical and imaging surveillance in germline TP53 mutation carriers with Li-Fraumeni syndrome: 11 year follow-up of a prospective observational study
.
Lancet Oncol
2016
;
17
:
1295
305
.
34.
Friedman
DN
,
Lis
E
,
Sklar
CA
,
Oeffinger
KC
,
Reppucci
M
,
Fleischut
MH
, 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
:
1440
4
.
35.
Jasperson
KW
,
Kohlmann
W
,
Gammon
A
,
Slack
H
,
Buchmann
L
,
Hunt
J
, et al
Role of rapid sequence whole-body MRI screening in SDH-associated hereditary paraganglioma families
.
Fam Cancer
2014
;
13
:
257
65
.
36.
Anupindi
SA
,
Bedoya
MA
,
Lindell
RB
,
Rambhatla
SJ
,
Zelley
K
,
Nichols
KE
, et al
Diagnostic performance of whole-body MRI as a tool for cancer screening in children with genetic cancer-predisposing conditions
.
Am J Roentgenol
2015
;
205
:
400
8
.
37.
Bidstrup
PE
,
Johansen
C
,
Mitchell
AJ
. 
Screening for cancer-related distress: Summary of evidence from tools to programmes
.
Acta Oncol
2011
;
50
:
194
204
.
38.
Lammens
CR
,
Bleiker
EM
,
Aaronson
NK
,
Wagner
A
,
Sijmons
RH
,
Ausems
MG
, et al
Regular surveillance for Li-Fraumeni Syndrome: advice, adherence and perceived benefits
.
Fam Cancer
2010
;
9
:
647
54
.
39.
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
:
269
79
.
40.
Rasmussen
A
,
Alonso
E
,
Ochoa
A
,
De Biase
I
,
Familiar
I
,
Yescas
P
, et al
Uptake of genetic testing and long-term tumor surveillance in von Hippel-Lindau disease
.
BMC Med Genet
2010
;
11
:
1
9
.
41.
Gopie
JP
,
Vasen
HF
,
Tibben
A
. 
Surveillance for hereditary cancer: does the benefit outweigh the psychological burden?–A systematic review
.
Crit Rev Oncol Hematol
2012
;
83
:
329
40
.
42.
Barrow
P
,
Khan
M
,
Lalloo
F
,
Evans
DG
,
Hill
J
. 
Systematic review of the impact of registration and screening on colorectal cancer incidence and mortality in familial adenomatous polyposis and Lynch syndrome
.
Br J Surg
2013
;
100
:
1719
31
.
43.
Teplick
A
,
Kowalski
M
,
Biegel
JA
,
Nichols
KE
. 
Educational paper: screening in cancer predisposition syndromes: guidelines for the general pediatrician
.
Eur J Pediatr
2011
;
170
:
285
94
.
44.
Samuel
N
,
Villani
A
,
Fernandez
CV
,
Malkin
D
. 
Management of familial cancer: sequencing, surveillance and society
.
Nat Rev Clin Oncol
2014
;
11
:
723
31
.
45.
Jhaveri
AP
,
Bale
A
,
Lovick
N
,
Zuckerman
K
,
Deshpande
H
,
Rath
K
, et al
The benefit and burden of cancer screening in Li-Fraumeni syndrome: a case report
.
Yale J Biol Med
2015
;
88
:
181
5
.
46.
Feiler
B
. 
Scanxiety. Fear of a postcancer ritual
.
Time
2011
;
177
:
56
.
47.
Lu
KH
,
Wood
ME
,
Daniels
M
,
Burke
C
,
Ford
J
,
Kauff
ND
, et al
American society of clinical oncology expert statement: collection and use of a cancer family history for oncology providers
.
J Clin Oncol
2014
;
32
:
833
40
.