Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Multicenter Study
. 2018 Jun;19(6):785-798.
doi: 10.1016/S1470-2045(18)30242-0. Epub 2018 May 9.

Spectrum and prevalence of genetic predisposition in medulloblastoma: a retrospective genetic study and prospective validation in a clinical trial cohort

Sebastian M Waszak  1 Paul A Northcott  2 Ivo Buchhalter  3 Giles W Robinson  4 Christian Sutter  5 Susanne Groebner  6 Kerstin B Grund  5 Laurence Brugières  7 David T W Jones  8 Kristian W Pajtler  9 A Sorana Morrissy  10 Marcel Kool  8 Dominik Sturm  9 Lukas Chavez  6 Aurelie Ernst  11 Sebastian Brabetz  9 Michael Hain  11 Thomas Zichner  1 Maia Segura-Wang  1 Joachim Weischenfeldt  12 Tobias Rausch  1 Balca R Mardin  1 Xin Zhou  13 Cristina Baciu  14 Christian Lawerenz  15 Jennifer A Chan  16 Pascale Varlet  17 Lea Guerrini-Rousseau  7 Daniel W Fults  18 Wiesława Grajkowska  19 Peter Hauser  20 Nada Jabado  21 Young-Shin Ra  22 Karel Zitterbart  23 Suyash S Shringarpure  24 Francisco M De La Vega  24 Carlos D Bustamante  24 Ho-Keung Ng  25 Arie Perry  26 Tobey J MacDonald  27 Pablo Hernáiz Driever  28 Anne E Bendel  29 Daniel C Bowers  30 Geoffrey McCowage  31 Murali M Chintagumpala  32 Richard Cohn  33 Timothy Hassall  34 Gudrun Fleischhack  35 Tone Eggen  36 Finn Wesenberg  37 Maria Feychting  38 Birgitta Lannering  39 Joachim Schüz  40 Christoffer Johansen  41 Tina V Andersen  42 Martin Röösli  43 Claudia E Kuehni  44 Michael Grotzer  45 Kristina Kjaerheim  36 Camelia M Monoranu  46 Tenley C Archer  47 Elizabeth Duke  48 Scott L Pomeroy  47 Redmond Shelagh  44 Stephan Frank  49 David Sumerauer  50 Wolfram Scheurlen  51 Marina V Ryzhova  52 Till Milde  53 Christian P Kratz  54 David Samuel  55 Jinghui Zhang  13 David A Solomon  26 Marco Marra  56 Roland Eils  57 Claus R Bartram  5 Katja von Hoff  58 Stefan Rutkowski  59 Vijay Ramaswamy  60 Richard J Gilbertson  61 Andrey Korshunov  62 Michael D Taylor  63 Peter Lichter  64 David Malkin  60 Amar Gajjar  4 Jan O Korbel  1 Stefan M Pfister  65
Affiliations
Multicenter Study

Spectrum and prevalence of genetic predisposition in medulloblastoma: a retrospective genetic study and prospective validation in a clinical trial cohort

Sebastian M Waszak et al. Lancet Oncol. 2018 Jun.

Abstract

Background: Medulloblastoma is associated with rare hereditary cancer predisposition syndromes; however, consensus medulloblastoma predisposition genes have not been defined and screening guidelines for genetic counselling and testing for paediatric patients are not available. We aimed to assess and define these genes to provide evidence for future screening guidelines.

Methods: In this international, multicentre study, we analysed patients with medulloblastoma from retrospective cohorts (International Cancer Genome Consortium [ICGC] PedBrain, Medulloblastoma Advanced Genomics International Consortium [MAGIC], and the CEFALO series) and from prospective cohorts from four clinical studies (SJMB03, SJMB12, SJYC07, and I-HIT-MED). Whole-genome sequences and exome sequences from blood and tumour samples were analysed for rare damaging germline mutations in cancer predisposition genes. DNA methylation profiling was done to determine consensus molecular subgroups: WNT (MBWNT), SHH (MBSHH), group 3 (MBGroup3), and group 4 (MBGroup4). Medulloblastoma predisposition genes were predicted on the basis of rare variant burden tests against controls without a cancer diagnosis from the Exome Aggregation Consortium (ExAC). Previously defined somatic mutational signatures were used to further classify medulloblastoma genomes into two groups, a clock-like group (signatures 1 and 5) and a homologous recombination repair deficiency-like group (signatures 3 and 8), and chromothripsis was investigated using previously established criteria. Progression-free survival and overall survival were modelled for patients with a genetic predisposition to medulloblastoma.

Findings: We included a total of 1022 patients with medulloblastoma from the retrospective cohorts (n=673) and the four prospective studies (n=349), from whom blood samples (n=1022) and tumour samples (n=800) were analysed for germline mutations in 110 cancer predisposition genes. In our rare variant burden analysis, we compared these against 53 105 sequenced controls from ExAC and identified APC, BRCA2, PALB2, PTCH1, SUFU, and TP53 as consensus medulloblastoma predisposition genes according to our rare variant burden analysis and estimated that germline mutations accounted for 6% of medulloblastoma diagnoses in the retrospective cohort. The prevalence of genetic predispositions differed between molecular subgroups in the retrospective cohort and was highest for patients in the MBSHH subgroup (20% in the retrospective cohort). These estimates were replicated in the prospective clinical cohort (germline mutations accounted for 5% of medulloblastoma diagnoses, with the highest prevalence [14%] in the MBSHH subgroup). Patients with germline APC mutations developed MBWNT and accounted for most (five [71%] of seven) cases of MBWNT that had no somatic CTNNB1 exon 3 mutations. Patients with germline mutations in SUFU and PTCH1 mostly developed infant MBSHH. Germline TP53 mutations presented only in childhood patients in the MBSHH subgroup and explained more than half (eight [57%] of 14) of all chromothripsis events in this subgroup. Germline mutations in PALB2 and BRCA2 were observed across the MBSHH, MBGroup3, and MBGroup4 molecular subgroups and were associated with mutational signatures typical of homologous recombination repair deficiency. In patients with a genetic predisposition to medulloblastoma, 5-year progression-free survival was 52% (95% CI 40-69) and 5-year overall survival was 65% (95% CI 52-81); these survival estimates differed significantly across patients with germline mutations in different medulloblastoma predisposition genes.

Interpretation: Genetic counselling and testing should be used as a standard-of-care procedure in patients with MBWNT and MBSHH because these patients have the highest prevalence of damaging germline mutations in known cancer predisposition genes. We propose criteria for routine genetic screening for patients with medulloblastoma based on clinical and molecular tumour characteristics.

Funding: German Cancer Aid; German Federal Ministry of Education and Research; German Childhood Cancer Foundation (Deutsche Kinderkrebsstiftung); European Research Council; National Institutes of Health; Canadian Institutes for Health Research; German Cancer Research Center; St Jude Comprehensive Cancer Center; American Lebanese Syrian Associated Charities; Swiss National Science Foundation; European Molecular Biology Organization; Cancer Research UK; Hertie Foundation; Alexander and Margaret Stewart Trust; V Foundation for Cancer Research; Sontag Foundation; Musicians Against Childhood Cancer; BC Cancer Foundation; Swedish Council for Health, Working Life and Welfare; Swedish Research Council; Swedish Cancer Society; the Swedish Radiation Protection Authority; Danish Strategic Research Council; Swiss Federal Office of Public Health; Swiss Research Foundation on Mobile Communication; Masaryk University; Ministry of Health of the Czech Republic; Research Council of Norway; Genome Canada; Genome BC; Terry Fox Research Institute; Ontario Institute for Cancer Research; Pediatric Oncology Group of Ontario; The Family of Kathleen Lorette and the Clark H Smith Brain Tumour Centre; Montreal Children's Hospital Foundation; The Hospital for Sick Children: Sonia and Arthur Labatt Brain Tumour Research Centre, Chief of Research Fund, Cancer Genetics Program, Garron Family Cancer Centre, MDT's Garron Family Endowment; BC Childhood Cancer Parents Association; Cure Search Foundation; Pediatric Brain Tumor Foundation; Brainchild; and the Government of Ontario.

PubMed Disclaimer

Figures

Figure 1
Figure 1
Discovery and replication of medulloblastoma predisposition genes (A) Study design and patient characteristics. (B) Rare variant burden analysis based on 673 patients with medulloblastoma and 53 105 controls from the Exome Aggregation Consortium. Diagnosis of medulloblastoma was the outcome variable. Proportion of patients with a genetic predisposition in our (C) retrospective cohort and (D) prospective cohort. (D) p values indicate statistical differences in the proportion of germline mutation carriers in the retrospective compared with prospective cohort. Infant=age 3 years or younger. Child=age 4–17 years. Adult=age 18 years or older. RR=relative risk. *p<0·001. †p<0·01. ‡p<0·05. §Patients with damaging germline mutations in APC, PTCH1, SUFU, TP53, PALB2, and BRCA2.
Figure 2
Figure 2
Clinical characteristics of patients with a genetic predisposition (A) Molecular tumour subgroups. (B) Age at diagnosis. (C) Family history of cancer. (D) Clinical signs of a genetic predisposition. p values indicate whether there is a difference between patients with different types of genetic predisposition. *Patients with compound heterozygous germline BRCA2 mutations. †Data are median (IQR). Circle sizes indicate the proportion of patients with a genetic predisposition.
Figure 3
Figure 3
Kaplan-Meier curves for survival outcomes of patients with a genetic predisposition (A) Progression-free survival. (B) Overall survival. Log-rank p values indicate differences across all patient groups.
Figure 4
Figure 4
Molecular medulloblastoma characteristics for patients with a genetic predisposition (A) Patterns of somatic inactivation of wild-type alleles for all patients with a germline mutation in one of the consensus medulloblastoma predisposition genes. Circle sizes indicate the proportion of patients with a genetic predisposition. (B) Somatic driver events in patients with medulloblastoma in the WNT subgroup (top panel) and age at diagnosis for all patients with germline APC mutations and patients in the WNT subgroup with somatic CTNNB1 mutations (bottom panel). (C) Association between germline PALB2 (n=5) and BRCA2 (n=7) mutation status and somatic mutational signatures 1, 3, 5, and 8. EXP=allele-specific gene expression. MUT=somatic inactivation via single nucleotide variation, insertion, or deletion. LOH=loss of heterozygosity. ND=no somatic alteration detected. NS=not significant. HRD=homologous recombination repair deficiency. *Present study and Hamilton and colleagues. †p<0·001. ‡p<0·01. §p<0·05.
Figure 5
Figure 5
Proposed clinical guidelines for genetic counselling and testing in medulloblastoma based on clinical and molecular tumour characteristics

Comment in

Similar articles

Cited by

References

    1. Johnson KJ, Cullen J, Barnholtz-Sloan JS. Childhood brain tumor epidemiology: a Brain Tumor Epidemiology Consortium review. Cancer Epidemiol Biomarkers Prev. 2014;23:2716–2736. - PMC - PubMed
    1. Northcott PA, Jones DTW, Kool M. Medulloblastomics: the end of the beginning. Nat Rev Cancer. 2012;12:818–834. - PMC - PubMed
    1. Wolter M, Reifenberger J, Sommer C, Ruzicka T, Reifenberger G. Mutations in the human homologue of the drosophila segment polarity gene patched (PTCH) in sporadic basal cell carcinomas of the skin and primitive neuroectodermal tumors of the central nervous system. Cancer Res. 1997;57:2581–2585. - PubMed
    1. Taylor MD, Liu L, Raffel C. Mutations in SUFU predispose to medulloblastoma. Nat Genet. 2002;31:306–310. - PubMed
    1. Li FP, Fraumeni JF. Rhabdomyosarcoma in children: epidemiologic study and identification of a familial cancer syndrome. J Natl Cancer Inst. 1969;43:1365–1373. - PubMed

Publication types

MeSH terms

Substances

-