Abstract

Background

Pediatric cancers are the leading cause of death by disease in children despite improved survival rates overall. The contribution of germline genetic susceptibility to pediatric cancer survivors has not been extensively characterized. We assessed the frequency of pathogenic or likely pathogenic (P/LP) variants in 5451 long-term pediatric cancer survivors from the Childhood Cancer Survivor Study.

Methods

Exome sequencing was conducted on germline DNA from 5451 pediatric cancer survivors (cases who survived ≥5 years from diagnosis; n = 5105 European) and 597 European cancer-free adults (controls). Analyses focused on comparing the frequency of rare P/LP variants in 237 cancer-susceptibility genes and a subset of 60 autosomal dominant high-to-moderate penetrance genes, for both case-case and case-control comparisons.

Results

Of European cases, 4.1% harbored a P/LP variant in high-to-moderate penetrance autosomal dominant genes compared with 1.3% in controls (2-sided P = 3 × 10-4). The highest frequency of P/LP variants was in genes typically associated with adult onset rather than pediatric cancers, including BRCA1/2, FH, PALB2, PMS2, and CDKN2A. A statistically significant excess of P/LP variants, after correction for multiple tests, was detected in patients with central nervous system cancers (NF1, SUFU, TSC1, PTCH2), Wilms tumor (WT1, REST), non-Hodgkin lymphoma (PMS2), and soft tissue sarcomas (SDHB, DICER1, TP53, ERCC4, FGFR3) compared with other pediatric cancers.

Conclusion

In long-term pediatric cancer survivors, we identified P/LP variants in cancer-susceptibility genes not previously associated with pediatric cancer as well as confirmed known associations. Further characterization of variants in these genes in pediatric cancer will be important to provide optimal genetic counseling for patients and their families.

Survival rates have statistically significantly improved in recent decades, yet cancer still remains the leading cause of death by disease in children (1). As a result of advances in cancer treatment and supportive care, a growing population of childhood cancer survivors are at risk for long-term therapy-related sequelae (2-4). Most childhood cancers are not associated with identified environmental risk factors, and accumulating evidence suggests germline genetic susceptibility is an important contributor to pediatric cancer etiology (5-8).

To date, 4 studies (n = 150-3006) have identified pathogenic or likely pathogenic (P/LP) germline variants in well-described cancer-susceptibility genes (CSG) in 5.8%-10.0% of pediatric cancer patients (6-9). Germline variants in TP53 were most commonly observed in 3 studies with up to 4.5% of cases carrying a P/LP variant (7-9). In the only pediatric cancer survivor cohort analysis, 6% of children harbored a P/LP germline variant in a CSG, most commonly in RB1 and NF1 (6,10). Acute lymphoblastic leukemia cases (11) and osteosarcoma cases (12) with germline TP53 P/LP variants have also been observed to die earlier or present with higher risk disease compared with the cases without germline TP53 P/LP variants, but this has not been evaluated for CSGs in multiple cancer types. Most pediatric cancer studies have evaluated only a subset of CSGs, limiting our understanding of the broader spectrum of defined and plausible CSGs, including those important in adult-onset cancers.

Despite mounting evidence of the importance of germline susceptibility in pediatric cancers, a comprehensive understanding across pediatric cancer types and outcomes is needed. To better understand the genetic etiology of primary pediatric cancers, we conducted a comprehensive analysis of 5451 long-term cancer survivors from the Childhood Cancer Survivor Study (CCSS) using exome sequencing (ES) and characterized the frequency of germline pathogenic variants in CSGs in individuals of European ancestry.

Methods

Study Population

CCSS is a multi-institutional research effort that established a large and longitudinally characterized cohort of 14 361 participants who survived childhood and adolescent cancer for 5 or more years in its original cohort diagnosed at 1 of 26 North American hospitals between 1970 and 1986 (13-15). Participants were younger than 21 years of age at diagnosis, and cancer types included bone, central nervous system, Hodgkin lymphoma, leukemia, neuroblastoma, non-Hodgkin lymphoma (NHL), soft tissue sarcoma (STS), and Wilms tumor. Specific cancer types are outlined in Table 1. Among survivors actively participating in CCSS, those who provided a germline sample (ie, blood, buccal cells, or saliva) survived a longer period (mean follow-up = 32.5 years) compared with patients who did not provide a sample (mean follow-up = 24.2 years). Additional details are provided in the Supplementary Methods (available online).

Table 1.

Demographics and clinical characteristics of the Childhood Cancer Survivor Study cases

CharacteristicsNo. of participants (%)Median age of diagnosis (range), y
Total5451 (100)
Sex
 Male2629 (48.2)
 Female2822 (51.8)
Ancestry
 European (CEU > 0.8)5105 (93.7)
 Non-European (CEU < 0.8)346 (6.3)
Diagnosis
 Bone449 (8.2)14.3 (2.0-21.0)
  Ewing sarcoma144 (2.6)12.5 (2.9-20.3)
  Osteosarcomaa284 (5.2)14.7 (3.0-20.9)
  Other/NOS21 (0.4)16.8 (2.0-21.0)
 Central nervous system718 (13.2)7.8 (0.0-20.9)
  Astrocytoma365 (6.7)7.7 (0.0-20.9)
  Medulloblastoma142 (2.6)7.9 (0.5-18.3)
  Other/NOS211 (3.9)8.2 (0.0-20.9)
 Hodgkin lymphoma741 (13.6)15.1 (3.5-21.0)
  Mixed cellularity94 (1.7)13.8 (3.5-20.6)
  Nodular sclerosis468 (8.6)15.7 (3.9-21.0)
  Other/NOS179 (3.3)14.0 (3.5-20.9)
 Leukemia1719 (31.5)4.8 (0-20.8)
  Acute lymphoblastic leukemia1586 (29.1)5.0 (0.0-20.8)
  Acute myeloid leukemia98 (1.8)5.8 (0.1-20.4)
  Other/NOS35 (0.6)4.1 (0.7-15.4)
 Neuroblastoma394 (7.2)0.8 (0.0-20.7)
 Non-Hodgkin lymphoma372 (6.8)10.8 (0.8-20.7)
  Burkitt lymphoma65 (1.2)8.7 (2.7-19.8)
  Diffuse large B-cell lymphoma71 (1.3)13.5 (5.2-20.7)
  Other/NOS236 (4.3)10.8 (0.8-20.7)
 Soft tissue sarcoma515 (9.4)9.6 (0.0-20.9)
  Rhabdomyosarcoma296 (5.4)6.1 (0.1-20.9)
  Other/NOS219 (4.0)13.6 (0.0-20.8)
 Wilms543 (10.0)3.2 (0.0-18.5)
Vital status
 Alive5077 (93.1)
 Dead374 (6.9)
CharacteristicsNo. of participants (%)Median age of diagnosis (range), y
Total5451 (100)
Sex
 Male2629 (48.2)
 Female2822 (51.8)
Ancestry
 European (CEU > 0.8)5105 (93.7)
 Non-European (CEU < 0.8)346 (6.3)
Diagnosis
 Bone449 (8.2)14.3 (2.0-21.0)
  Ewing sarcoma144 (2.6)12.5 (2.9-20.3)
  Osteosarcomaa284 (5.2)14.7 (3.0-20.9)
  Other/NOS21 (0.4)16.8 (2.0-21.0)
 Central nervous system718 (13.2)7.8 (0.0-20.9)
  Astrocytoma365 (6.7)7.7 (0.0-20.9)
  Medulloblastoma142 (2.6)7.9 (0.5-18.3)
  Other/NOS211 (3.9)8.2 (0.0-20.9)
 Hodgkin lymphoma741 (13.6)15.1 (3.5-21.0)
  Mixed cellularity94 (1.7)13.8 (3.5-20.6)
  Nodular sclerosis468 (8.6)15.7 (3.9-21.0)
  Other/NOS179 (3.3)14.0 (3.5-20.9)
 Leukemia1719 (31.5)4.8 (0-20.8)
  Acute lymphoblastic leukemia1586 (29.1)5.0 (0.0-20.8)
  Acute myeloid leukemia98 (1.8)5.8 (0.1-20.4)
  Other/NOS35 (0.6)4.1 (0.7-15.4)
 Neuroblastoma394 (7.2)0.8 (0.0-20.7)
 Non-Hodgkin lymphoma372 (6.8)10.8 (0.8-20.7)
  Burkitt lymphoma65 (1.2)8.7 (2.7-19.8)
  Diffuse large B-cell lymphoma71 (1.3)13.5 (5.2-20.7)
  Other/NOS236 (4.3)10.8 (0.8-20.7)
 Soft tissue sarcoma515 (9.4)9.6 (0.0-20.9)
  Rhabdomyosarcoma296 (5.4)6.1 (0.1-20.9)
  Other/NOS219 (4.0)13.6 (0.0-20.8)
 Wilms543 (10.0)3.2 (0.0-18.5)
Vital status
 Alive5077 (93.1)
 Dead374 (6.9)
a

Osteosarcoma data were previously published (12). Cancer types are indented below each groups of cancers. CEU = Northern and Western European ancestry; NOS = not otherwise specified.

Table 1.

Demographics and clinical characteristics of the Childhood Cancer Survivor Study cases

CharacteristicsNo. of participants (%)Median age of diagnosis (range), y
Total5451 (100)
Sex
 Male2629 (48.2)
 Female2822 (51.8)
Ancestry
 European (CEU > 0.8)5105 (93.7)
 Non-European (CEU < 0.8)346 (6.3)
Diagnosis
 Bone449 (8.2)14.3 (2.0-21.0)
  Ewing sarcoma144 (2.6)12.5 (2.9-20.3)
  Osteosarcomaa284 (5.2)14.7 (3.0-20.9)
  Other/NOS21 (0.4)16.8 (2.0-21.0)
 Central nervous system718 (13.2)7.8 (0.0-20.9)
  Astrocytoma365 (6.7)7.7 (0.0-20.9)
  Medulloblastoma142 (2.6)7.9 (0.5-18.3)
  Other/NOS211 (3.9)8.2 (0.0-20.9)
 Hodgkin lymphoma741 (13.6)15.1 (3.5-21.0)
  Mixed cellularity94 (1.7)13.8 (3.5-20.6)
  Nodular sclerosis468 (8.6)15.7 (3.9-21.0)
  Other/NOS179 (3.3)14.0 (3.5-20.9)
 Leukemia1719 (31.5)4.8 (0-20.8)
  Acute lymphoblastic leukemia1586 (29.1)5.0 (0.0-20.8)
  Acute myeloid leukemia98 (1.8)5.8 (0.1-20.4)
  Other/NOS35 (0.6)4.1 (0.7-15.4)
 Neuroblastoma394 (7.2)0.8 (0.0-20.7)
 Non-Hodgkin lymphoma372 (6.8)10.8 (0.8-20.7)
  Burkitt lymphoma65 (1.2)8.7 (2.7-19.8)
  Diffuse large B-cell lymphoma71 (1.3)13.5 (5.2-20.7)
  Other/NOS236 (4.3)10.8 (0.8-20.7)
 Soft tissue sarcoma515 (9.4)9.6 (0.0-20.9)
  Rhabdomyosarcoma296 (5.4)6.1 (0.1-20.9)
  Other/NOS219 (4.0)13.6 (0.0-20.8)
 Wilms543 (10.0)3.2 (0.0-18.5)
Vital status
 Alive5077 (93.1)
 Dead374 (6.9)
CharacteristicsNo. of participants (%)Median age of diagnosis (range), y
Total5451 (100)
Sex
 Male2629 (48.2)
 Female2822 (51.8)
Ancestry
 European (CEU > 0.8)5105 (93.7)
 Non-European (CEU < 0.8)346 (6.3)
Diagnosis
 Bone449 (8.2)14.3 (2.0-21.0)
  Ewing sarcoma144 (2.6)12.5 (2.9-20.3)
  Osteosarcomaa284 (5.2)14.7 (3.0-20.9)
  Other/NOS21 (0.4)16.8 (2.0-21.0)
 Central nervous system718 (13.2)7.8 (0.0-20.9)
  Astrocytoma365 (6.7)7.7 (0.0-20.9)
  Medulloblastoma142 (2.6)7.9 (0.5-18.3)
  Other/NOS211 (3.9)8.2 (0.0-20.9)
 Hodgkin lymphoma741 (13.6)15.1 (3.5-21.0)
  Mixed cellularity94 (1.7)13.8 (3.5-20.6)
  Nodular sclerosis468 (8.6)15.7 (3.9-21.0)
  Other/NOS179 (3.3)14.0 (3.5-20.9)
 Leukemia1719 (31.5)4.8 (0-20.8)
  Acute lymphoblastic leukemia1586 (29.1)5.0 (0.0-20.8)
  Acute myeloid leukemia98 (1.8)5.8 (0.1-20.4)
  Other/NOS35 (0.6)4.1 (0.7-15.4)
 Neuroblastoma394 (7.2)0.8 (0.0-20.7)
 Non-Hodgkin lymphoma372 (6.8)10.8 (0.8-20.7)
  Burkitt lymphoma65 (1.2)8.7 (2.7-19.8)
  Diffuse large B-cell lymphoma71 (1.3)13.5 (5.2-20.7)
  Other/NOS236 (4.3)10.8 (0.8-20.7)
 Soft tissue sarcoma515 (9.4)9.6 (0.0-20.9)
  Rhabdomyosarcoma296 (5.4)6.1 (0.1-20.9)
  Other/NOS219 (4.0)13.6 (0.0-20.8)
 Wilms543 (10.0)3.2 (0.0-18.5)
Vital status
 Alive5077 (93.1)
 Dead374 (6.9)
a

Osteosarcoma data were previously published (12). Cancer types are indented below each groups of cancers. CEU = Northern and Western European ancestry; NOS = not otherwise specified.

Controls consisted of 597 cancer-free adults [median age at the last follow-up = 69 years, see Table 2; selected for European ancestry from the National Cancer Institute Prostate Lung, Colon and Ovarian Cancer Prevention Trial (16) and the American Cancer Society Cancer Prevention Study II (17); hereafter, “controls”]. All participants provided written informed consent and were recruited through institutional review board–approved protocols.

Table 2.

Demographics and clinical characteristics of the cancer-free controls

CharacteristicsNo. of participants (%)Average age at blood collection (range), y
Total597 (100)
Sex
 Male325 (54.4)68.8 (55-88)
 Female272 (45.6)68.2 (55-87)
CharacteristicsNo. of participants (%)Average age at blood collection (range), y
Total597 (100)
Sex
 Male325 (54.4)68.8 (55-88)
 Female272 (45.6)68.2 (55-87)
Table 2.

Demographics and clinical characteristics of the cancer-free controls

CharacteristicsNo. of participants (%)Average age at blood collection (range), y
Total597 (100)
Sex
 Male325 (54.4)68.8 (55-88)
 Female272 (45.6)68.2 (55-87)
CharacteristicsNo. of participants (%)Average age at blood collection (range), y
Total597 (100)
Sex
 Male325 (54.4)68.8 (55-88)
 Female272 (45.6)68.2 (55-87)

Exome Sequencing and Bioinformatic Analysis

ES was performed as previously described (18). Briefly, after alignment to human genome assembly hg19 with NovoAlign (http://www.novocraft.com), variants were jointly called using GATK2 HaplotypeCaller (v3.3-0-g37228af), GATK UnifiedGenotyper (v3.1-1-g07a4bf8), and FreeBayes (v0.9.14-24-gc292036). Variants were retained if called by HaplotypeCaller and UnifiedGenotyper and/or FreeBayes within 5 base pairs of the target capture region, and the frequency was less than 10% within the dataset. Individual genotypes were retained if the HaplotypeCaller genotype quality score was 20 or more and an alternative allele read depth of less than 1. The average depth was 50x (Supplementary Figure 1, available online); 808 samples were whole-genome amplified (19). SNPWEIGHTS version 2.1 (20) was used to infer genetic ancestry for all CCSS cases and cancer-free controls; individuals with more than 80% European ancestry were considered European. Additional details are provided in the Supplementary Methods (available online).

Candidate Cancer-Susceptibility Gene List

We investigated 237 CSGs (hereafter, CSG-237) from 4 published studies (9,21-23) (Supplementary Table 1, available online). We focused our analyses on 172 autosomal dominant genes, including both autosomal dominant and autosomal recessive genes and those of unknown inheritance (hereafter, CSG-172), and a subset of 60 autosomal dominant genes with high-to-moderate penetrance (hereafter, CSG-60) previously reported (6,7).

Pathogenicity Classification

The analysis was restricted to variants with a minor allele frequency of less than 1% in cases or controls drawn from all ethnic subgroups reported in Exome Aggregation Consortium (excluding The Cancer Genome Atlas data) (24), 1000 Genomes Project (25), and the Exome Sequencing Project (26). Variants were categorized as pathogenic (P), likely pathogenic (LP), variant of uncertain significance, likely benign, or benign using a hierarchical classification system based on ClinVar, the Human Gene Mutation Database, and InterVar (Supplementary Figure 2, Supplementary Table 2, available online). Details are described in the Supplementary Methods (available online).

Statistical Methods

Rare-variant association tests were conducted for both case-case and case-control comparisons, based on the burden test statistics included in the optimal unified test R package. The case-case analyses were performed using all cases in 2 different ways: individual cancer type and groups of cancers. Details are described in the Supplementary Methods (available online). The case-control analyses were performed using European ancestry individuals only and used the individual cancer type and groups of cancers approaches but compared the P/LP frequencies to controls.

In a time-to-event analysis of overall survival, we compared cases harboring P/LP variants in the CSG-172 vs those without any P/LP variants. At-risk time for death started at the age at sample collection and ended either with age at death (all-cause mortality as the event of interest) or age at last follow-up (censoring). Cox proportional hazards regression models were used to estimate the hazard ratio (HR) of death and 95% confidence intervals (CIs) adjusting for individual cancer type, age at diagnosis, and treatments (anthracycline, cyclophosphamide, and any radiation within 5 years). The R package “survival” was used to fit the Cox models and generate Kaplan-Meier survival curves.

Statistical tests were performed with R version 3.3.2. To correct for multiple testing, false discovery rates were calculated using the P adjust function, and a Q value of less than 0.1 was considered statistically significant for the burden test and denoted as Qburden.

Results

Summary Findings

We evaluated germline exonic variants in 5451 long-term pediatric cancer survivors within the CCSS cohort. The median age at diagnosis for primary cancers was 7.2 years (range = 0-21.0 years), and the median follow-up time since age at diagnosis was 32.6 years (range = 8.6-46.3 years) with a median age at follow-up of 41.0 years (range = 10.9-64.3 years). Cases were 48.2% male, 93.7% were of European ancestry, and 93.1% were alive at last follow-up (Table 1).

P/LP Variants in Autosomal Dominant CSGs

In 5105 pediatric cancer survivors with European ancestry, 211 (4.1%, 95% CI = 3.6% to 4.7%) harbored at least 1 P/LP variant in a high-to-moderate penetrance autosomal dominant CSG (CSG-60), which was statistically significantly more than the 1.3% (95% CI = 0.4% to 2.3%) observed in controls (PFisher = 3 x 10-4) (Table 3). Out of 123 cases with P/LP variants, 7 cases carried a P/LP variant in more than 1 CSG (FH/NF1, FH/BRCA2, MSH2/BRCA2, VHL/PALB2, PMS2/BRCA1, BRAF/NF1, NF1/BRCA1). In analyses by cancer type, the highest frequency of P/LP variants was observed for patients with astrocytoma (7.4%, n = 27), followed by medulloblastoma (6.3%, n = 9) and osteosarcoma (6.3%, n = 18); the latter differed substantially from a recent survey of more than 1000 cases of osteosarcoma (720 unselected cases, 284 CCSS cases), where 11.1% of unselected cases had a P/LP variant in the CSG-60, suggesting that P/LP variants could be of poor prognosis in osteosarcoma (12). In the more expanded set of autosomal dominant CSGs (CSG-172), there were also statistically significantly more P/LP variants in cases (11.9%) compared with controls (7.7%) (PFisher = 2.2 × 10-3), and the highest frequency of P/LP variants was found in leukemia other or not otherwise specified (NOS) (17.1%) followed by rhabdomyosarcoma (14.9%) and astrocytoma (14.5%) (Supplementary Figure 3, Supplementary Table 3, available online).

Table 3.

Classification of rare variants in 60 autosomal dominant cancer-susceptibility genes (CSG) with high-to-moderate penetrance (CSG-60)a and 172 autosomal dominant CSG (CSG-172)b in European-ancestry cases compared with European-ancestry controls

ClassificationCases (CEU > 0.8)Controls (CEU > 0.8)P (Fisher exact test)
(n = 5105)
(n = 597)
Allele count (%)Unique countAllele count (%)Unique count
CSG-60a2.71 × 10-4
 P127 (2.5)1044 (0.7)4
 LP84 (1.6)534 (0.7)3
 Percent P/LP (95% CI)4.1 (3.6 to 4.7)1.3 (0.4 to 2.3)
CSG-172b.002
 P355 (7.0)21228 (4.7)22
 LP251 (4.9)18418 (3.0)17
 Percent P/LP (95% CI)11.9 (11.0 to 12.8)7.7 (5.6 to 9.8)
ClassificationCases (CEU > 0.8)Controls (CEU > 0.8)P (Fisher exact test)
(n = 5105)
(n = 597)
Allele count (%)Unique countAllele count (%)Unique count
CSG-60a2.71 × 10-4
 P127 (2.5)1044 (0.7)4
 LP84 (1.6)534 (0.7)3
 Percent P/LP (95% CI)4.1 (3.6 to 4.7)1.3 (0.4 to 2.3)
CSG-172b.002
 P355 (7.0)21228 (4.7)22
 LP251 (4.9)18418 (3.0)17
 Percent P/LP (95% CI)11.9 (11.0 to 12.8)7.7 (5.6 to 9.8)
a

60 well-described autosomal dominant inheritance genes with high-to-moderate penetrance (6,7). CEU = Northern and Western European ancestry; CI = confidence intervals; LP = likely pathogenic; P = pathogenic.

b

172 genes from Rahman (21), Forbes et al. (22), Ripperger et al. (23), and Grobner et al. (9). GJB2 was excluded from our analysis because its major associated disease is deafness (27).

Table 3.

Classification of rare variants in 60 autosomal dominant cancer-susceptibility genes (CSG) with high-to-moderate penetrance (CSG-60)a and 172 autosomal dominant CSG (CSG-172)b in European-ancestry cases compared with European-ancestry controls

ClassificationCases (CEU > 0.8)Controls (CEU > 0.8)P (Fisher exact test)
(n = 5105)
(n = 597)
Allele count (%)Unique countAllele count (%)Unique count
CSG-60a2.71 × 10-4
 P127 (2.5)1044 (0.7)4
 LP84 (1.6)534 (0.7)3
 Percent P/LP (95% CI)4.1 (3.6 to 4.7)1.3 (0.4 to 2.3)
CSG-172b.002
 P355 (7.0)21228 (4.7)22
 LP251 (4.9)18418 (3.0)17
 Percent P/LP (95% CI)11.9 (11.0 to 12.8)7.7 (5.6 to 9.8)
ClassificationCases (CEU > 0.8)Controls (CEU > 0.8)P (Fisher exact test)
(n = 5105)
(n = 597)
Allele count (%)Unique countAllele count (%)Unique count
CSG-60a2.71 × 10-4
 P127 (2.5)1044 (0.7)4
 LP84 (1.6)534 (0.7)3
 Percent P/LP (95% CI)4.1 (3.6 to 4.7)1.3 (0.4 to 2.3)
CSG-172b.002
 P355 (7.0)21228 (4.7)22
 LP251 (4.9)18418 (3.0)17
 Percent P/LP (95% CI)11.9 (11.0 to 12.8)7.7 (5.6 to 9.8)
a

60 well-described autosomal dominant inheritance genes with high-to-moderate penetrance (6,7). CEU = Northern and Western European ancestry; CI = confidence intervals; LP = likely pathogenic; P = pathogenic.

b

172 genes from Rahman (21), Forbes et al. (22), Ripperger et al. (23), and Grobner et al. (9). GJB2 was excluded from our analysis because its major associated disease is deafness (27).

In pediatric cancer survivors carrying a germline P/LP variant in the CSG-172, the risk of all-cause mortality was elevated compared with those without a germline P/LP variant after adjusting for individual cancer type, age at diagnosis, and treatment (anthracycline, cyclophosphamide equivalent, and radiation within 5 years) (HR = 1.70, 95% CI = 1.27 to 2.28, PCox regression = 3 × 10-4) (Supplementary Figure 4, available online).

Rare-Variant Association Tests

We used a case-control rare-variant association test approach to identify genes with P/LP variants associated with childhood cancer susceptibility for both groups of cancers and specific cancer types. There was a statistically significant excess (Pburden < .05) of P/LP variants in cases compared with controls in NF1 (CNS, STS) (28), WT1 (Wilms tumor) (29), TP53 (bone and STS) (30), EGFR (Hodgkin lymphoma), and PMS2 (NHL) (31) (Table 4). However, after correcting for multiple testing, only NF1 (CNS) and WT1 (Wilms tumor) remained statistically significant (Qburden < 0.1). All of these genes are known to be associated with susceptibility to these specific cancer types and/or groups, except for EGFR to Hodgkin lymphoma.

Table 4.

Rare variant association analyses for gene-specific P/LP variant enrichment in pediatric cancer groups compared with cancer-free controlsa

Cancer groupbGenePQP/LP variants Case No. (%)No P/LP variants Case No. (%)P/LP variants Control No. (%)No P/LP variants Control No. (%)
BoneTP53.0413 (0.7)446 (99.3)0 (0)597 (100)
Central nervous systemNF13.43 × 10-55.01 × 10-319 (2.8)699 (97.4)0 (0)597 (100)
Hodgkin lymphomacEGFR.0415 (0.7)736 (99.3)0 (0)597 (100)
Wilms tumorWT15.67 × 10-48.28 × 10-210 (2.0)533 (98.0)0 (0)597 (100)
Non-Hodgkin lymphomaPMS2.0213 (0.8)369 (99.2)0 (0)597 (100)
Soft tissue sarcomaNF1.0068.03 x 10-16 (1.2)509 (98.8)0 (0)597 (100)
TP53.038.03 x 10-14 (0.8)511 (99.2)0 (0)597 (100)
Cancer groupbGenePQP/LP variants Case No. (%)No P/LP variants Case No. (%)P/LP variants Control No. (%)No P/LP variants Control No. (%)
BoneTP53.0413 (0.7)446 (99.3)0 (0)597 (100)
Central nervous systemNF13.43 × 10-55.01 × 10-319 (2.8)699 (97.4)0 (0)597 (100)
Hodgkin lymphomacEGFR.0415 (0.7)736 (99.3)0 (0)597 (100)
Wilms tumorWT15.67 × 10-48.28 × 10-210 (2.0)533 (98.0)0 (0)597 (100)
Non-Hodgkin lymphomaPMS2.0213 (0.8)369 (99.2)0 (0)597 (100)
Soft tissue sarcomaNF1.0068.03 x 10-16 (1.2)509 (98.8)0 (0)597 (100)
TP53.038.03 x 10-14 (0.8)511 (99.2)0 (0)597 (100)
a

All genes with a P value of less than .05 and at least 3 subjects with a P/LP variant were included. P/LP = pathogenic/likely pathogenic.

b

Cancer grouping was based on similarities in histology or location and are listed in italics in Table 1.

c

Novel findings.

Table 4.

Rare variant association analyses for gene-specific P/LP variant enrichment in pediatric cancer groups compared with cancer-free controlsa

Cancer groupbGenePQP/LP variants Case No. (%)No P/LP variants Case No. (%)P/LP variants Control No. (%)No P/LP variants Control No. (%)
BoneTP53.0413 (0.7)446 (99.3)0 (0)597 (100)
Central nervous systemNF13.43 × 10-55.01 × 10-319 (2.8)699 (97.4)0 (0)597 (100)
Hodgkin lymphomacEGFR.0415 (0.7)736 (99.3)0 (0)597 (100)
Wilms tumorWT15.67 × 10-48.28 × 10-210 (2.0)533 (98.0)0 (0)597 (100)
Non-Hodgkin lymphomaPMS2.0213 (0.8)369 (99.2)0 (0)597 (100)
Soft tissue sarcomaNF1.0068.03 x 10-16 (1.2)509 (98.8)0 (0)597 (100)
TP53.038.03 x 10-14 (0.8)511 (99.2)0 (0)597 (100)
Cancer groupbGenePQP/LP variants Case No. (%)No P/LP variants Case No. (%)P/LP variants Control No. (%)No P/LP variants Control No. (%)
BoneTP53.0413 (0.7)446 (99.3)0 (0)597 (100)
Central nervous systemNF13.43 × 10-55.01 × 10-319 (2.8)699 (97.4)0 (0)597 (100)
Hodgkin lymphomacEGFR.0415 (0.7)736 (99.3)0 (0)597 (100)
Wilms tumorWT15.67 × 10-48.28 × 10-210 (2.0)533 (98.0)0 (0)597 (100)
Non-Hodgkin lymphomaPMS2.0213 (0.8)369 (99.2)0 (0)597 (100)
Soft tissue sarcomaNF1.0068.03 x 10-16 (1.2)509 (98.8)0 (0)597 (100)
TP53.038.03 x 10-14 (0.8)511 (99.2)0 (0)597 (100)
a

All genes with a P value of less than .05 and at least 3 subjects with a P/LP variant were included. P/LP = pathogenic/likely pathogenic.

b

Cancer grouping was based on similarities in histology or location and are listed in italics in Table 1.

c

Novel findings.

We performed case-case rare-variant association tests for individual cancer types and confirmed known associations (28-31) between specific CSG-237 P/LP variants and susceptibility to individual cancer types and observed a novel association. There was a statistically significant excess (Qburden < 0.1) of DHCR7 P/LP variants in rhabdomyosarcoma cases that has not been previously described (Table 5). In our case-case analyses by groups of cancer, we observed a statistically significant excess (Qburden < 0.1) of P/LP variants in the following genes: PMS2 in NHL and SDHB, ERCC4 and FGFR3 in STS (Table 5). All findings remained statistically significant after adjusting for population stratification (using the top 15 principal components) except for TP53 association with bone (Supplementary Tables 4 and 5, available online).

Table 5.

Rare variant association analyses for gene-specific P/LP variant enrichment in individual cancer types using case-case comparisons and case-case comparisons for cancer groupsa

Cancer typeGenePQP/LP variantNo P/LP variantP/LP variantNo P/LP variant
CaseCaseOther casesOther cases
No. (%)No. (%)No. (%)No. (%)
Individual cancer type case-case comparisons
 AstrocytomaNF12.67 × 10-223.89 × 10-2013 (3.6)352 (96.4)6 (0.1)5080 (99.9)
PTPN11.0024.43 × 10-23 (0.8)362 (99.2)5 (0.1)5081 (99.9)
 MedulloblastomaSUFU1.28 × 10-161.88 × 10-143 (2.1)139 (97.9)1 (0.0)5308 (100)
 RhabdomyosarcomaDICER12.74 × 10-84.01 × 10-63 (1.0)293 (99.0)1 (0.0)5154 (100)
TP531.57 × 10-71.15 × 10-55 (1.7)291 (98.3)6 (0.1)5149 (99.9)
DHCR7  b3.26 × 10-35.94 × 10-211 (3.7)285 (96.3)66 (1.3)5089 (98.7)
 Wilms tumorWT17.99 × 10-181.17 × 10-1510 (1.8)533 (98.2)0 (0.0)4908 (100)
REST2.52 × 10-61.84 × 10-43 (0.6)540 (99.4)0 (0.0)4908 (100)
Cancer group case-case comparisons
 Central nervous systemNF12.06 × 10-193.01 × 10-1721 (2.9)697 (97.1)10 (0.2)4723 (99.8)
SUFU2.53 × 10-41.24 × 10-23 (0.4)715 (99.6)1 (0.0)4732 (100)
TSC12.54 × 10-41.24 × 10-23 (0.4)715 (99.6)1 (0.0)4732 (100)
PTCH2.0027.18 × 10-23 (0.4)715 (99.6)2 (0.0)4731 (100)
 Wilms tumorcWT11.80 × 10-212.63 × 10-1910 (1.8)533 (98.2)0 (0.0)4908 (100)
REST1.90 × 10-71.39 × 10-53 (0.6)540 (99.4)0 (0.0)4908 (100)
 Non-Hodgkin lymphomaPMS2  b5.78 × 10-42.11 × 10-23 (0.8)369 (99.2)5 (0.1)5074 (99.9)
 Soft tissue sarcomaSDHB  b7.33 × 10-65.35 × 10-43 (0.6)512 (99.4)1 (0.0)4935 (100)
DICER17.33 × 10-65.35 × 10-43 (0.6)512 (99.4)1 (0.0)4935 (100)
TP534.38 × 10-52.13 × 10-35 (1.0)510 (99.0)6 (0.1)4930 (99.9)
ERCC4  b1.11 × 10-44.05 × 10-33 (0.6)512 (99.4)2 (0.0)4934 (100)
FGFR3  b6.79 × 10-41.98 × 10-23 (0.6)512 (99.4)3 (0.1)4933 (99.9)
Cancer typeGenePQP/LP variantNo P/LP variantP/LP variantNo P/LP variant
CaseCaseOther casesOther cases
No. (%)No. (%)No. (%)No. (%)
Individual cancer type case-case comparisons
 AstrocytomaNF12.67 × 10-223.89 × 10-2013 (3.6)352 (96.4)6 (0.1)5080 (99.9)
PTPN11.0024.43 × 10-23 (0.8)362 (99.2)5 (0.1)5081 (99.9)
 MedulloblastomaSUFU1.28 × 10-161.88 × 10-143 (2.1)139 (97.9)1 (0.0)5308 (100)
 RhabdomyosarcomaDICER12.74 × 10-84.01 × 10-63 (1.0)293 (99.0)1 (0.0)5154 (100)
TP531.57 × 10-71.15 × 10-55 (1.7)291 (98.3)6 (0.1)5149 (99.9)
DHCR7  b3.26 × 10-35.94 × 10-211 (3.7)285 (96.3)66 (1.3)5089 (98.7)
 Wilms tumorWT17.99 × 10-181.17 × 10-1510 (1.8)533 (98.2)0 (0.0)4908 (100)
REST2.52 × 10-61.84 × 10-43 (0.6)540 (99.4)0 (0.0)4908 (100)
Cancer group case-case comparisons
 Central nervous systemNF12.06 × 10-193.01 × 10-1721 (2.9)697 (97.1)10 (0.2)4723 (99.8)
SUFU2.53 × 10-41.24 × 10-23 (0.4)715 (99.6)1 (0.0)4732 (100)
TSC12.54 × 10-41.24 × 10-23 (0.4)715 (99.6)1 (0.0)4732 (100)
PTCH2.0027.18 × 10-23 (0.4)715 (99.6)2 (0.0)4731 (100)
 Wilms tumorcWT11.80 × 10-212.63 × 10-1910 (1.8)533 (98.2)0 (0.0)4908 (100)
REST1.90 × 10-71.39 × 10-53 (0.6)540 (99.4)0 (0.0)4908 (100)
 Non-Hodgkin lymphomaPMS2  b5.78 × 10-42.11 × 10-23 (0.8)369 (99.2)5 (0.1)5074 (99.9)
 Soft tissue sarcomaSDHB  b7.33 × 10-65.35 × 10-43 (0.6)512 (99.4)1 (0.0)4935 (100)
DICER17.33 × 10-65.35 × 10-43 (0.6)512 (99.4)1 (0.0)4935 (100)
TP534.38 × 10-52.13 × 10-35 (1.0)510 (99.0)6 (0.1)4930 (99.9)
ERCC4  b1.11 × 10-44.05 × 10-33 (0.6)512 (99.4)2 (0.0)4934 (100)
FGFR3  b6.79 × 10-41.98 × 10-23 (0.6)512 (99.4)3 (0.1)4933 (99.9)
a

All genes with a Q value of less than 0.1 and at least 3 subjects with a P/LP variant were included. P/LP = pathogenic/likely pathogenic.

b

Novel findings. Counts (%) are shown for the specified tested cancer type as “case” compared with all “other cases” for each comparison.

c

Wilms did not fit into any histologic or location grouping (Supplementary Table 2, available online) and was therefore analyzed as its own group.

Table 5.

Rare variant association analyses for gene-specific P/LP variant enrichment in individual cancer types using case-case comparisons and case-case comparisons for cancer groupsa

Cancer typeGenePQP/LP variantNo P/LP variantP/LP variantNo P/LP variant
CaseCaseOther casesOther cases
No. (%)No. (%)No. (%)No. (%)
Individual cancer type case-case comparisons
 AstrocytomaNF12.67 × 10-223.89 × 10-2013 (3.6)352 (96.4)6 (0.1)5080 (99.9)
PTPN11.0024.43 × 10-23 (0.8)362 (99.2)5 (0.1)5081 (99.9)
 MedulloblastomaSUFU1.28 × 10-161.88 × 10-143 (2.1)139 (97.9)1 (0.0)5308 (100)
 RhabdomyosarcomaDICER12.74 × 10-84.01 × 10-63 (1.0)293 (99.0)1 (0.0)5154 (100)
TP531.57 × 10-71.15 × 10-55 (1.7)291 (98.3)6 (0.1)5149 (99.9)
DHCR7  b3.26 × 10-35.94 × 10-211 (3.7)285 (96.3)66 (1.3)5089 (98.7)
 Wilms tumorWT17.99 × 10-181.17 × 10-1510 (1.8)533 (98.2)0 (0.0)4908 (100)
REST2.52 × 10-61.84 × 10-43 (0.6)540 (99.4)0 (0.0)4908 (100)
Cancer group case-case comparisons
 Central nervous systemNF12.06 × 10-193.01 × 10-1721 (2.9)697 (97.1)10 (0.2)4723 (99.8)
SUFU2.53 × 10-41.24 × 10-23 (0.4)715 (99.6)1 (0.0)4732 (100)
TSC12.54 × 10-41.24 × 10-23 (0.4)715 (99.6)1 (0.0)4732 (100)
PTCH2.0027.18 × 10-23 (0.4)715 (99.6)2 (0.0)4731 (100)
 Wilms tumorcWT11.80 × 10-212.63 × 10-1910 (1.8)533 (98.2)0 (0.0)4908 (100)
REST1.90 × 10-71.39 × 10-53 (0.6)540 (99.4)0 (0.0)4908 (100)
 Non-Hodgkin lymphomaPMS2  b5.78 × 10-42.11 × 10-23 (0.8)369 (99.2)5 (0.1)5074 (99.9)
 Soft tissue sarcomaSDHB  b7.33 × 10-65.35 × 10-43 (0.6)512 (99.4)1 (0.0)4935 (100)
DICER17.33 × 10-65.35 × 10-43 (0.6)512 (99.4)1 (0.0)4935 (100)
TP534.38 × 10-52.13 × 10-35 (1.0)510 (99.0)6 (0.1)4930 (99.9)
ERCC4  b1.11 × 10-44.05 × 10-33 (0.6)512 (99.4)2 (0.0)4934 (100)
FGFR3  b6.79 × 10-41.98 × 10-23 (0.6)512 (99.4)3 (0.1)4933 (99.9)
Cancer typeGenePQP/LP variantNo P/LP variantP/LP variantNo P/LP variant
CaseCaseOther casesOther cases
No. (%)No. (%)No. (%)No. (%)
Individual cancer type case-case comparisons
 AstrocytomaNF12.67 × 10-223.89 × 10-2013 (3.6)352 (96.4)6 (0.1)5080 (99.9)
PTPN11.0024.43 × 10-23 (0.8)362 (99.2)5 (0.1)5081 (99.9)
 MedulloblastomaSUFU1.28 × 10-161.88 × 10-143 (2.1)139 (97.9)1 (0.0)5308 (100)
 RhabdomyosarcomaDICER12.74 × 10-84.01 × 10-63 (1.0)293 (99.0)1 (0.0)5154 (100)
TP531.57 × 10-71.15 × 10-55 (1.7)291 (98.3)6 (0.1)5149 (99.9)
DHCR7  b3.26 × 10-35.94 × 10-211 (3.7)285 (96.3)66 (1.3)5089 (98.7)
 Wilms tumorWT17.99 × 10-181.17 × 10-1510 (1.8)533 (98.2)0 (0.0)4908 (100)
REST2.52 × 10-61.84 × 10-43 (0.6)540 (99.4)0 (0.0)4908 (100)
Cancer group case-case comparisons
 Central nervous systemNF12.06 × 10-193.01 × 10-1721 (2.9)697 (97.1)10 (0.2)4723 (99.8)
SUFU2.53 × 10-41.24 × 10-23 (0.4)715 (99.6)1 (0.0)4732 (100)
TSC12.54 × 10-41.24 × 10-23 (0.4)715 (99.6)1 (0.0)4732 (100)
PTCH2.0027.18 × 10-23 (0.4)715 (99.6)2 (0.0)4731 (100)
 Wilms tumorcWT11.80 × 10-212.63 × 10-1910 (1.8)533 (98.2)0 (0.0)4908 (100)
REST1.90 × 10-71.39 × 10-53 (0.6)540 (99.4)0 (0.0)4908 (100)
 Non-Hodgkin lymphomaPMS2  b5.78 × 10-42.11 × 10-23 (0.8)369 (99.2)5 (0.1)5074 (99.9)
 Soft tissue sarcomaSDHB  b7.33 × 10-65.35 × 10-43 (0.6)512 (99.4)1 (0.0)4935 (100)
DICER17.33 × 10-65.35 × 10-43 (0.6)512 (99.4)1 (0.0)4935 (100)
TP534.38 × 10-52.13 × 10-35 (1.0)510 (99.0)6 (0.1)4930 (99.9)
ERCC4  b1.11 × 10-44.05 × 10-33 (0.6)512 (99.4)2 (0.0)4934 (100)
FGFR3  b6.79 × 10-41.98 × 10-23 (0.6)512 (99.4)3 (0.1)4933 (99.9)
a

All genes with a Q value of less than 0.1 and at least 3 subjects with a P/LP variant were included. P/LP = pathogenic/likely pathogenic.

b

Novel findings. Counts (%) are shown for the specified tested cancer type as “case” compared with all “other cases” for each comparison.

c

Wilms did not fit into any histologic or location grouping (Supplementary Table 2, available online) and was therefore analyzed as its own group.

Multiple Cancer Types Associated With Well-Known CSGs

Figure 1 depicts the landscape of P/LP variants in genes from the CSG-60. We observed P/LP variants in 40 of the 60 genes (67.7%). The 10 genes with the most P/LP variants were genes typically associated with germline variants in adult-onset cancers rather than pediatric cancers, including BRCA2, FH, BRCA1, PALB2, PMS2, and CDKN2A, even after correction for gene size (data not shown). P/LP variants in both PMS2 and MSH6 were observed in NHL, leukemia, and Hodgkin lymphoma, suggesting, if replicated, a previously unsuspected link of leukemia and lymphoma with Lynch syndrome. In addition, we observed cases with Wilms tumor and bone malignancies to harbor P/LP variants in PMS2 and a neuroblastoma with a P/LP variant in MSH6. Numerous patients with leukemia carried P/LP variants in genes associated with pheochromocytoma or paraganglioma (SDHA, SDHB, SDHD, MAX, TMEM127) and a P/LP variant in SDHC with a neuroblastoma. Both leukemia and neuroblastoma are phenotypes not known to be associated with these genes.

Summary of the pathogenic/likely pathogenic (P/LP) variants in the CSG-60
                            by cancer type and patient demographics. Each column represents 1 case.
                            The right y-axis represents the total number of P/LP
                            variants found in each gene and the method for variant classification.
                            The top (-axis summarizes the patient demographics by cancer type. CNS
                            = central nervous system; CSG = cancer susceptibility
                            gene; HL = Hodgkin lymphoma; NBL = neuroblastoma; NHL
                            = non-Hodgkin lymphoma; STS = soft tissue sarcoma; Wilms
                            = Wilms tumor.
Figure 1.

Summary of the pathogenic/likely pathogenic (P/LP) variants in the CSG-60 by cancer type and patient demographics. Each column represents 1 case. The right y-axis represents the total number of P/LP variants found in each gene and the method for variant classification. The top (-axis summarizes the patient demographics by cancer type. CNS = central nervous system; CSG = cancer susceptibility gene; HL = Hodgkin lymphoma; NBL = neuroblastoma; NHL = non-Hodgkin lymphoma; STS = soft tissue sarcoma; Wilms = Wilms tumor.

Homozygous P/LP Variants in Autosomal Recessive CSGs

We identified 2 individuals who were homozygous carriers of P/LP autosomal recessive gene variants: 1 each in MUTYH and SBDS. The MUTYH homozygote carrier developed a Wilms tumor and harbored no other P/LP variant in a CSG (including WT1 and REST). The SBDS homozygote carrier developed a STS other or NOS and did not have P/LP variants in known STS-associated genes (including NF1, TP53, or DICER1).

Discussion

Among 5451 long-term survivors of European ancestry with a prior diagnosis of pediatric cancer, we identified an excess of P/LP variants in established CSGs compared with adult cancer-free controls. P/LP variants in specific CSGs and cancer types not previously recognized have been identified. Of the cases, 4% had a P/LP variant in a well-described subset of autosomal dominant CSGs (CSG-60) with high-to-moderate penetrance, and 11.9% had a P/LP variant in a more expansive list of autosomal dominant CSGs (CSG-172). This large pediatric cancer survivor population and comprehensive CSG evaluation enabled us to confirm and, importantly, to extend understanding of the phenotypic consequences of rare germline variants in CSGs and, at the same time, highlight pleiotropy in well-characterized CSGs.

A comparison of the frequency of P/LP variants reported in large published pediatric pan-cancer studies is daunting because of differences in the distribution of cancer types, pathogenicity scoring criteria, and ascertainment biases in collecting cases (eg, from high-risk cases referred to tertiary centers). The frequency of CSG-60 P/LP variants in our cohort was similar to the previously published long-term pediatric cancer survivor study, St. Jude Lifetime Cohort Study (approximately 4%), after restricting cancer types to include only cancers in CCSS (excluding retinoblastoma and adrenal cortical carcinoma because of their known associations with RB1 and TP53 P/LP variants, respectively) (6). The 2 collected series (7,9) of available pediatric cancers reported a prevalence of approximately 8%, which differs from our current observation in the CCSS; the lower prevalence in CCSS could be due to specific biologic features of cases with underlying P/LP that could result in poorer prognosis. The overall lower P/LP carrier rate in long-term survivor cohorts could be partly due to early mortality in highest-risk cases (some harboring a P/LP variant) prior to eligibility in a long-term survival study.

Previous studies showing adverse prognosis for P/LP variant carriers in certain genes (eg, TP53) have primarily focused on survival within 10 years from diagnosis (11). Our data suggest that P/LP germline variants could adversely impact long-term survival in excess of 10 years from diagnosis; this is particularly notable for TP53 P/LP overall as well as in osteosarcoma overall (12,32). Our study population has an introduced bias because of the inclusion of only long-term survivors who provided a DNA sample. Additional research is needed to assess potential confounding factors and evaluate disease-specific mortality. Nevertheless, if validated, these results could highlight the importance of assessing germline variant status even among long-term survivors and, perhaps, be useful for management of future survivorship (33).

In our case-control and case-case analyses, we observed well-established associations between CSG and specific cancer types, which provides confidence in our data and variant classification: NF1 and astrocytoma (34), STS (35); WT1 and Wilms tumor (29); PTPN11 and astrocytoma (36); REST and Wilms tumor (37); SUFU and medulloblastoma (38); and TP53 and rhabdomyosarcoma (39), STS (39), bone (40). These results are consistent with prior reports, such as NF1 and astrocytoma (41). In contrast, TP53 P/LP variants have been shown to be associated with poorer overall survival in unselected patients with osteosarcoma, and the rate of P/LP carriers observed in our osteosarcoma survivors is lower than previously reported rates in unselected cases for TP53 (1% vs 5%-10% of cases) (12,32).

We have extended the range of cancer subtypes that are associated with P/LP variants in CSGs and, if replicated, could be incorporated into future counseling for those with these P/LP variants. For example, we observed an increase in SDHB P/LP variants in children with STS, which is notable because pathogenic variation in SDHB has been reported for susceptibility to pheochromocytoma and paraganglioma as well as the Carney triad but not yet in pediatric sarcomas (42). We also observed a Wilms tumor in a child with homozygous pathogenic MUTYH variants, a gene associated with increased risk for polyposis and colorectal cancer (43), and the child had no other P/LP variants in our CSGs (including WT1 and REST). Last, a patient with a SBDS homozygote pathogenic variant developed STS other or NOS and did not have P/LP variants in NF1, TP53, or DICER1, which are known to be associated with STS. Although pathogenic SBDS variants that cause the autosomal recessive disorder Shwachman-Diamond syndrome are known to be associated with risk of myelodysplastic syndrome and/or AML, solid malignancies are not a common outcome (44,45). If replicated, these findings could have implications for the clinical care of people with SDHB, MUTYH, and SBDS pathogenic variants.

We further extended our analysis of candidate genes (CSG-237) to include those somatically mutated in pediatric cancer but not yet associated with germline susceptibility. In particular, we observed a statistically significant excess of loss-of-function P/LP variants in FGFR3 in children with STS, which is intriguing because somatic mutations in FGFR3 are frequently observed in bladder cancer (46). In addition, Muenke syndrome, a disorder of craniosynostosis, arises from germline gain-of-function variation in FGFR3, but to date, it is not known to be associated with germline cancer susceptibility (47). Our observation, if replicated, demonstrates: 1) an unexpected, putative genotype-phenotype correlation with FGFR3, 2) a novel germline phenotype in an otherwise well-known gene, and 3) a rare example of a somatically mutated gene also associated with germline susceptibility.

NF1 and BRCA2 had the highest frequency of pathogenic variants among the CSG-60, and TP53 was the seventh most common. Interestingly, we identified that 1.7% (93 of 5451) of cases had a P/LP variant in 1 of 6 genes (BRCA1/2, FH, PALB2, PMS2, and CDKN2A) typically associated with adult-onset tumors. Variation in these genes has not been well studied in pediatric cancer populations and highlights the unexpected pleiotropy of even well-characterized CSGs. Understanding penetrance in these genes also remains a challenge. For example, BRCA2 P/LP variants were observed in 0.5% of cases, but the carrier frequency was the same as in our controls (0.7%). Although BRCA2 P/LP variants have been reported in pediatric cancers (48,49), individual cases will require careful interpretation and perhaps orthogonal data (eg, tumor sequencing) to confirm causality.

The identification of a germline P/LP variant could result in a clinical intervention such as risk-reduction surgery, surveillance imaging, choice of therapeutic agent (50-52), and/or clinical genetic testing of asymptomatic family members (49,53,54). Such testing has historically centered around adult disease processes (eg, breast and colon cancer), and because it was performed in younger family members, it has been termed cascade testing. (53,54) Here, we identified P/LP variants in CSGs (eg, BRCA1/2, MEN1) linked to adult cancers. Observing variants in these genes in our pediatric cohort suggests a reexamination of the phenotypes associated with these genes and potentially the consideration of whether the parents (and other relatives) of young children with P/LP variants could benefit from “reverse-cascade testing.”

The strengths of our study included the large size of the case cohort and comparison of germline ES data from pediatric cancer survivors with controls that were jointly called using the same pipeline and quality control metrics. Although our controls were modest in number, we did not use publicly available controls for gene discovery because of the detection of statistically significant hyperinflation likely due to bioinformatic pipeline differences (Supplementary Figure 5, available online). However, we do see confirmation of our findings in these public databases. As a limitation, we may be underestimating the prevalence of damaging variants within the CSGs given our conservative, clinically based American College of Medical Genetics and Genomics and Association of Molecular Pathology classification criteria (55). Our study includes long-term pediatric cancer survivors, which limits generalizability.

In summary, we identified new associations that could be important to pediatric cancer susceptibility and, in turn, those who transition to survivorship with the risk of a second cancer (12,56,57). Our study includes long-term pediatric cancer survivors, which limits generalizability. Further studies are needed to validate and refine our observations, including functional investigation of the pathogenicity of novel variants in pediatric cancer. Characterization of pleiotropy is of central importance in cancer genetics to provide optimal genetic counseling and clinical surveillance. In pediatrics, germline genetic testing is frequently ordered in the context of children with phenotypes associated with a particular syndrome or a family history of cancer. Because of this ascertainment bias, the phenotypic cancer spectrum arising from pathogenic germline variants remains incomplete, even for iconic CSGs such as TP53. Our findings improve understanding of germline pediatric cancer genetic etiology and may lead to better pediatric cancer risk stratification at diagnosis, genetic counseling for patients and family members, and outcomes.

Funding

This work was supported by the Intramural Research Program of the Division of Cancer Epidemiology and Genetics of the National Cancer Institute, Bethesda, MD. We thank the CCSS participants and referring physicians for their valuable contributions. CCSS also is supported by the National Cancer Institute (CA55727, GT Armstrong, principal investigator) and St. Jude Children’s Research Hospital through the National Cancer Institute Cancer Center Support (CORE) grant (CA21765, C. Roberts, principal investigator) and the American Lebanese-Syrian Associated Charities (ALSAC).

Notes

Role of the funders: The funders had no role in the design of the study; the collection, analysis, and interpretation of the data; the writing of the manuscript; and the decision to submit the manuscript for publication.

Disclosures: DRS performs contract clinical telegenetics services for Genome Medical, Inc, in accordance with relevant National Cancer Institute ethics policies.

Prior presentation: This work was presented in part of the following meeting: Platform presentation at the 69th Annual Meeting of the American Society of Human Genetics, Houston, Texas, October 2019.

Disclaimer: The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does mention of trade names, commercial products or organizations imply endorsement by the US government.

Acknowledgment: This work used the computational resources of the NIH High Performance Computing Biowulf cluster.

Author Contribution: Conceptualization: JK, MG, DMK, SAS, LMM, DRS, LM; Formal analysis: JK, MG, WL, BZ, LS, JNS; Resources: SWH, LM, LLR, YYGTA, SB, WML, RD, LT, NF, BDH, BZ, MW, KJ, AAH, CD; Data curation: JK, MG, DMK, MNF, MD, MY, SAB, KCA, FPF, AMG, PPK, MJM, MLM, MLN, LO, AP, MP, MR, KS, TW; Writing - original draft: JK, MG, DRS, LM; Writing - review & editing: all authors; Supervision: SAS, LMM, DRS, LM.

Data Availability

The data for this project is publicly available (dbGAP accession: phs002072.v1.p1), as per funder policies.

References

1

Siegel
RL
,
Miller
KD
,
Jemal
A.
 
Cancer statistics, 2020
.
CA A Cancer J Clin
.
2020
;
70
(
1
):
7
30
.

2

Robison
LL
,
Hudson
MM.
 
Survivors of childhood and adolescent cancer: life-long risks and responsibilities
.
Nat Rev Cancer
.
2014
;
14
(
1
):
61
70
.

3

Bhakta
N
,
Liu
Q
,
Ness
KK
, et al.  
The cumulative burden of surviving childhood cancer: an initial report from the St Jude Lifetime Cohort Study (SJLIFE)
.
Lancet
.
2017
;
390
(
10112
):
2569
2582
.

4

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
(
9
):
833
842
.

5

Spector
LG
,
Pankratz
N
,
Marcotte
EL.
 
Genetic and nongenetic risk factors for childhood cancer
.
Pediatr Clin North Am
.
2015
;
62
(
1
):
11
25
.

6

Wang
Z
,
Wilson
CL
,
Easton
J
, et al.  
Genetic risk for subsequent neoplasms among long-term survivors of childhood cancer
.
J Clin Oncol
.
2018
;
36
(
20
):
2078
2087
.

7

Zhang
J
,
Walsh
MF
,
Wu
G
, et al.  
Germline mutations in predisposition genes in pediatric cancer
.
N Engl J Med
.
2015
;
373
(
24
):
2336
2346
.

8

Parsons
DW
,
Roy
A
,
Yang
Y
, et al.  
Diagnostic yield of clinical tumor and germline whole-exome sequencing for children with solid tumors
.
JAMA Oncol
.
2016
;
2
(
5
):
616
624
.

9

Grobner
SN
,
Worst
BC
,
Weischenfeldt
J
, et al. ;
ICGC PedBrain-Seq Project
.
The landscape of genomic alterations across childhood cancers
.
Nature
.
2018
;
555
(
7696
):
321
327
.

10

Wilson
CL
,
Wang
Z
,
Liu
Q
, et al.  
Estimated number of adult survivors of childhood cancer in United States with cancer-predisposing germline variants
.
Pediatr Blood Cancer
.
2020
;
67
(
2
):10.1002/pbc.28047:e28047.

11

Qian
M
,
Cao
X
,
Devidas
M
, et al.  
TP53 germline variations influence the predisposition and prognosis of B-cell acute lymphoblastic leukemia in children
.
J Clin Oncol
.
2018
;
36
(
6
):
591
599
.

12

Mirabello
L
,
Zhu
B
,
Koster
R
, et al.  
Frequency of pathogenic germline variants in cancer-susceptibility genes in patients with osteosarcoma
.
JAMA Oncol
.
2020
;
6
(
5
):
724
. 10.1001/jamaoncol.2020.0197.

13

Robison
LL
,
Mertens
AC
,
Boice
JD
, et al.  
Study design and cohort characteristics of the Childhood Cancer Survivor Study: a multi-institutional collaborative project
.
Med Pediatr Oncol
.
2002
;
38
(
4
):
229
239
.

14

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
(
14
):
2308
2318
.

15

Leisenring
WM
,
Mertens
AC
,
Armstrong
GT
, et al.  
Pediatric cancer survivorship research: experience of the Childhood Cancer Survivor Study
.
J Clin Oncol
.
2009
;
27
(
14
):
2319
2327
.

16

Prorok
PC
,
Andriole
GL
,
Bresalier
RS
, et al.  
Design of the Prostate, Lung, Colorectal and Ovarian (PLCO) cancer screening trial
.
Control Clin Trials
.
2000
;
21
(
6
):
273S
309S
.

17

Calle
EE
,
Rodriguez
C
,
Jacobs
EJ
, et al.  
The American Cancer Society Cancer Prevention Study II Nutrition Cohort: rationale, study design, and baseline characteristics
.
Cancer
.
2002
;
94
(
2
):
500
511
.

18

Kim
J
,
Luo
W
,
Wang
M
, et al.  
Prevalence of pathogenic/likely pathogenic variants in the 24 cancer genes of the ACMG Secondary Findings v2.0 list in a large cancer cohort and ethnicity-matched controls
.
Genome Med
.
2018
;
10
(
1
):
99
.

19

Dagnall
CL
,
Morton
LM
,
Hicks
BD
, et al.  
Successful use of whole genome amplified DNA from multiple source types for high-density Illumina SNP microarrays
.
BMC Genomics
.
2018
;
19
(
1
):
182
.

20

Chen
CY
,
Pollack
S
,
Hunter
DJ
, et al.  
Improved ancestry inference using weights from external reference panels
.
Bioinformatics
.
2013
;
29
(
11
):
1399
1406
.

21

Rahman
N.
 
Realizing the promise of cancer predisposition genes
.
Nature
.
2014
;
505
(
7483
):
302
308
.

22

Forbes
SA
,
Beare
D
,
Boutselakis
H
, et al.  
COSMIC: somatic cancer genetics at high-resolution
.
Nucleic Acids Res
.
2017
;
45
(
D1
):
D777
D783
.

23

Ripperger
T
,
Bielack
SS
,
Borkhardt
A
, 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
;
173
(
4
):
1017
1037
.

24

Lek
M
,
Karczewski
KJ
,
Minikel
EV
, et al. ;
Exome Aggregation Consortium
.
Analysis of protein-coding genetic variation in 60,706 humans
.
Nature
.
2016
;
536
(
7616
):
285
291
.

25

Auton
A
,
Brooks
LD
,
Durbin
RM
, et al. ;
Genomes Project C
.
A global reference for human genetic variation
.
Nature
.
2015
;
526
(
7571
):
68
74
.

26

Exome Variant Server, NHLBI GO Exome Sequencing Project (ESP), Seattle
, WA. http://evs.gs.washington.edu/EVS/.

27

Smith
RJH
,
Ranum
PT.
 Nonsyndromic hearing loss and deafness, DFNA3. In:
Adam
MP
,
Ardinger
HH
,
Pagon
RA
, et al. , eds.
GeneReviews
.
Seattle, WA
;
University of Washington
;
1993
. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.

28

Friedman
JM.
 Neurofibromatosis 1. In:
Adam
MP
,
Ardinger
HH
,
Pagon
RA
, et al. , eds.
GeneReviews
.
Seattle, WA
;
University of Washington
;
1993
. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.

29

Dome
JS
,
Huff
V.
 Wilms tumor predisposition. In:
Adam
MP
,
Ardinger
HH
,
Pagon
RA
, et al. , eds.
GeneReviews
.
Seattle, WA
;
University of Washington
;
1993
. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.

30

Schneider
K
,
Zelley
K
,
Nichols
KE
, et al.  Li-Fraumeni Syndrome. In:
Adam
MP
,
Ardinger
HH
,
Pagon
RA
, et al. , eds.
GeneReviews
.
Seattle, WA
;
University of Washington
;
1993
. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.

31

Kohlmann
W
,
Gruber
SB
, Lynch Syndrome. In:
Adam
MP
,
Ardinger
HH
,
Pagon
RA
, et al. , eds.
GeneReviews
.
Seattle, WA
;
University of Washington
;
1993
. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.

32

Mirabello
L
,
Yeager
M
,
Mai
PL
, et al.  
Germline TP53 variants and susceptibility to osteosarcoma
.
J Natl Cancer Inst
.
2015
;
107
(
7
).

33

Li
MM
,
Chao
E
,
Esplin
ED
, et al. ;
ACMG Professional Practice and Guidelines Committee
.
Points to consider for reporting of germline variation in patients undergoing tumor testing: a statement of the American College of Medical Genetics and Genomics (ACMG)
.
Genet Med
.
2020
;
22
(
7
):
1142
1148
.

34

Mandiwanza
T
,
Kaliaperumal
C
,
Khalil
A
, et al.  
Suprasellar pilocytic astrocytoma: one national centre’s experience
.
Childs Nerv Syst
.
2014
;
30
(
7
):
1243
1248
.

35

Crucis
A
,
Richer
W
,
Brugieres
L
, et al.  
Rhabdomyosarcomas in children with neurofibromatosis type I: a national historical cohort
.
Pediatr Blood Cancer
.
2015
;
62
(
10
):
1733
1738
.

36

Siegfried
A
,
Cances
C
,
Denuelle
M
, et al.  
Noonan syndrome, PTPN11 mutations, and brain tumors. A clinical report and review of the literature
.
Am J Med Genet A
.
2017
;
173
(
4
):
1061
1065
.

37

Mahamdallie
SS
,
Hanks
S
,
Karlin
KL
, et al.  
Mutations in the transcriptional repressor REST predispose to Wilms tumor
.
Nat Genet
.
2015
;
47
(
12
):
1471
1474
.

38

Taylor
MD
,
Liu
L
,
Raffel
C
, et al.  
Mutations in SUFU predispose to medulloblastoma
.
Nat Genet
.
2002
;
31
(
3
):
306
310
.

39

Diller
L
,
Sexsmith
E
,
Gottlieb
A
, et al.  
Germline p53 mutations are frequently detected in young children with rhabdomyosarcoma
.
J Clin Invest
.
1995
;
95
(
4
):
1606
1611
.

40

Chompret
A
,
Abel
A
,
Stoppa-Lyonnet
D
, et al.  
Sensitivity and predictive value of criteria for p53 germline mutation screening
.
J Med Genet
.
2001
;
38
(
1
):
43
47
.

41

Krishnatry
R
,
Zhukova
N
,
Guerreiro Stucklin
AS
, et al.  
Clinical and treatment factors determining long-term outcomes for adult survivors of childhood low-grade glioma: a population-based study
.
Cancer
.
2016
;
122
(
8
):
1261
1269
.

42

Else
T
,
Greenberg
S
,
Fishbein
L.
 Hereditary Paraganglioma-Pheochromocytoma Syndromes. In:
Adam
MP
,
Ardinger
HH
,
Pagon
RA
, et al. , eds.
GeneReviews
.
Seattle,WA
;
University of Washington
;
1993
. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved..

43

Nielsen
M
,
Infante
E
,
Brand
R.
 MUTYH Polyposis. In:
Adam
MP
,
Ardinger
HH
,
Pagon
RA
, et al. , eds.
GeneReviews
.
Seattle, WA
;
University of Washington
;
1993
. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.

44

Nelson
A
,
Myers
K.
 Shwachman-Diamond Syndrome. In:
Adam
MP
,
Ardinger
HH
,
Pagon
RA
, et al. , eds.
GeneReviews
.
Seattle, WA
:
University of Washington
;
1993
. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.

45

Nelson
AS
,
Myers
KC.
 
Diagnosis, treatment, and molecular pathology of Shwachman-Diamond Syndrome
.
Hematol Oncol Clin North Am
.
2018
;
32
(
4
):
687
700
.

46

Sfakianos
JP
,
Cha
EK
,
Iyer
G
, et al.  
Genomic characterization of upper tract urothelial carcinoma
.
Eur Urol
.
2015
;
68
(
6
):
970
977
.

47

Kruszka
P
,
Addissie
YA
,
Agochukwu
NB
, et al.  Muenke Syndrome. In:
Adam
MP
,
Ardinger
HH
,
Pagon
RA
, et al. , eds.
GeneReviews
.
Seattle, WA
;
University of Washington
;
1993
. GeneReviews is a registered trademark of the University of Washington, Seattle. All rights reserved.

48

Wang
Z
,
Wilson
CL
,
Armstrong
GT
, et al.  
Association of germline BRCA2 mutations with the risk of pediatric or adolescent non-Hodgkin lymphoma
.
JAMA Oncol
.
2019
;
5
(
9
):
1362
.

49

Walsh
MF
,
Kennedy
J
,
Harlan
M
, et al.  
Germline BRCA2 mutations detected in pediatric sequencing studies impact parents’ evaluation and care
.
Cold Spring Harb Mol Case Stud
.
2017
;
3
(
6
):
a001925
.

50

Robson
M
,
Im
SA
,
Senkus
E
, et al.  
Olaparib for metastatic breast cancer in patients with a germline BRCA mutation
.
N Engl J Med
.
2017
;
377
(
6
):
523
533
.

51

Litton
JK
,
Rugo
HS
,
Ettl
J
, et al.  
Talazoparib in patients with advanced breast cancer and a germline BRCA mutation
.
N Engl J Med
.
2018
;
379
(
8
):
753
763
.

52

Overman
MJ
,
Lonardi
S
,
Wong
KYM
, et al.  
Durable clinical benefit with nivolumab plus ipilimumab in DNA mismatch repair-deficient/microsatellite instability-high metastatic colorectal cancer
.
J Clin Oncol
.
2018
;
36
(
8
):
773
779
.

53

Caswell-Jin
JL
,
Zimmer
AD
,
Stedden
W
, et al.  
Cascade genetic testing of relatives for hereditary cancer risk: results of an online initiative
.
J Natl Cancer Inst
.
2019
;
111
(
1
):
95
98
.

54

Krawczak
M
,
Cooper
DN
,
Schmidtke
J.
 
Estimating the efficacy and efficiency of cascade genetic screening
.
Am J Hum Genet
.
2001
;
69
(
2
):
361
370
.

55

Richards
S
,
Aziz
N
,
Bale
S
, et al. ; on behalf of the ACMG Laboratory Quality Assurance Committee.
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
(
5
):
405
423
.

56

Qin
N
,
Wang
Z
,
Liu
Q
, et al.  
Pathogenic germline mutations in DNA repair genes in combination with cancer treatment exposures and risk of subsequent neoplasms among long-term survivors of childhood cancer
.
J Clin Oncol
.
2020
;
38
(
24
):
2728
1200
.

57

Morton
LM
,
Karyadi
DM
,
Hartley
S
, et al.  
Subsequent neoplasm risk associated with rare variants in DNA repair and clinical radiation sensitivity syndrome genes: a report from the Childhood Cancer Survivor Study
.
J Clin Oncol
.
2019
;
37
(
suppl 15
):
10028
.

Author notes

Jung Kim, Matthew Gianferante, Douglas R. Stewart, Lisa Mirabello, contributed equally to this work.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)

Supplementary data