Your browser doesn't support javascript.
loading
Multiple Germline Events Contribute to Cancer Development in Patients with Li-Fraumeni Syndrome.
Subasri, Vallijah; Light, Nicholas; Kanwar, Nisha; Brzezinski, Jack; Luo, Ping; Hansford, Jordan R; Cairney, Elizabeth; Portwine, Carol; Elser, Christine; Finlay, Jonathan L; Nichols, Kim E; Alon, Noa; Brunga, Ledia; Anson, Jo; Kohlmann, Wendy; de Andrade, Kelvin C; Khincha, Payal P; Savage, Sharon A; Schiffman, Joshua D; Weksberg, Rosanna; Pugh, Trevor J; Villani, Anita; Shlien, Adam; Goldenberg, Anna; Malkin, David.
Afiliação
  • Subasri V; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
  • Light N; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.
  • Kanwar N; Vector Institute, Toronto, Ontario, Canada.
  • Brzezinski J; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.
  • Luo P; Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada.
  • Hansford JR; Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada.
  • Cairney E; Division of Haematology/Oncology, The Hospital for Sick Children, Department of Paediatrics, University of Toronto, Toronto, Ontario, Canada.
  • Portwine C; Princess Margaret Cancer Center, University Health Network, Toronto, Ontario, Canada.
  • Elser C; Children's Cancer Centre, Royal Children's Hospital, Melbourne, Victoria, Australia.
  • Finlay JL; Murdoch Children's Research Institute, Parkville, Victoria, Australia.
  • Nichols KE; Department of Pediatrics, University of Melbourne, Melbourne, Australia.
  • Alon N; Michael Rice Cancer Centre, Women's and Children's Hospital, North Adelaide, South Australia, Australia.
  • Brunga L; South Australia Health and Medical Research Institute, Adelaide, South Australia, Australia.
  • Anson J; South Australia Immunogenomics Cancer Institute, University of Adelaide, Adelaide, Australia.
  • Kohlmann W; Department of Paediatrics, London Health Sciences Centre and Western University, London, Ontario, Canada.
  • de Andrade KC; Department of Paediatrics, McMaster University, Hamilton, Ontario, Canada.
  • Khincha PP; Department of Medical Oncology, Princess Margaret Hospital and Mount Sinai Hospital, Toronto, Ontario, Canada.
  • Savage SA; Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
  • Schiffman JD; Neuro-Oncology Program, Nationwide Children's Hospital and The Ohio State University, Columbus, Ohio.
  • Weksberg R; Department of Oncology, St Jude Children's Research Hospital, Memphis, Tennessee.
  • Pugh TJ; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.
  • Villani A; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada.
  • Shlien A; Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
  • Goldenberg A; Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
  • Malkin D; Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, NCI, Bethesda, Maryland.
Cancer Res Commun ; 3(5): 738-754, 2023 05.
Article em En | MEDLINE | ID: mdl-37377903
ABSTRACT
Li-Fraumeni syndrome (LFS) is an autosomal dominant cancer-predisposition disorder. Approximately 70% of individuals who fit the clinical definition of LFS harbor a pathogenic germline variant in the TP53 tumor suppressor gene. However, the remaining 30% of patients lack a TP53 variant and even among variant TP53 carriers, approximately 20% remain cancer-free. Understanding the variable cancer penetrance and phenotypic variability in LFS is critical to developing rational approaches to accurate, early tumor detection and risk-reduction strategies. We leveraged family-based whole-genome sequencing and DNA methylation to evaluate the germline genomes of a large, multi-institutional cohort of patients with LFS (n = 396) with variant (n = 374) or wildtype TP53 (n = 22). We identified alternative cancer-associated genetic aberrations in 8/14 wildtype TP53 carriers who developed cancer. Among variant TP53 carriers, 19/49 who developed cancer harbored a pathogenic variant in another cancer gene. Modifier variants in the WNT signaling pathway were associated with decreased cancer incidence. Furthermore, we leveraged the noncoding genome and methylome to identify inherited epimutations in genes including ASXL1, ETV6, and LEF1 that confer increased cancer risk. Using these epimutations, we built a machine learning model that can predict cancer risk in patients with LFS with an area under the receiver operator characteristic curve (AUROC) of 0.725 (0.633-0.810).

Significance:

Our study clarifies the genomic basis for the phenotypic variability in LFS and highlights the immense benefits of expanding genetic and epigenetic testing of patients with LFS beyond TP53. More broadly, it necessitates the dissociation of hereditary cancer syndromes as single gene disorders and emphasizes the importance of understanding these diseases in a holistic manner as opposed to through the lens of a single gene.
Assuntos

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Síndrome de Li-Fraumeni Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Cancer Res Commun Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Canadá

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Síndrome de Li-Fraumeni Tipo de estudo: Prognostic_studies Limite: Humans Idioma: En Revista: Cancer Res Commun Ano de publicação: 2023 Tipo de documento: Article País de afiliação: Canadá