BRAF Mutation and CDKN2A deletion define a clinically distinct subgroup of childhood secondary high-grade glioma

Detalhes bibliográficos
Autor(a) principal: Mistry, Matthew
Data de Publicação: 2015
Outros Autores: Zhukova, Nataliya, Merico, Daniele, Rakopoulos, Patricia, Krishnatry, Rahul, Shago, Mary, Stavropoulos, James, Alon, Noa, Pole, Jason D., Ray, Peter N., Navickiene, Vilma, Mangerel, Joshua, Remke, Marc, Buczkowicz, Pawel, Ramaswamy, Vijay, Stucklin, Ana Guerreiro, Li, Martin, Young, Edwin J., Zhang, Cindy, Castelo-Branco, Pedro, Bakry, Doua, Laughlin, Suzanne, Shlien, Adam, Chan, Jennifer, Ligon, Keith L., Rutka, James T., Dirks, Peter B., Taylor, Michael D., Greenberg, Mark, Malkin, David, Huang, Annie, Bouffet, Eric, Hawkins, Cynthia E., Tabori, Uri
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
Texto Completo: http://hdl.handle.net/10400.1/11546
Resumo: Purpose To uncover the genetic events leading to transformation of pediatric low-grade glioma (PLGG) to secondary high-grade glioma (sHGG). Patients and Methods We retrospectively identified patients with sHGG from a population-based cohort of 886 patients with PLGG with long clinical follow-up. Exome sequencing and array CGH were performed on available samples followed by detailed genetic analysis of the entire sHGG cohort. Clinical and outcome data of genetically distinct subgroups were obtained. Results sHGG was observed in 2.9% of PLGGs (26 of 886 patients). Patients with sHGG had a high frequency of nonsilent somatic mutations compared with patients with primary pediatric high-grade glioma (HGG; median, 25 mutations per exome; P = .0042). Alterations in chromatin-modifying genes and telomere-maintenance pathways were commonly observed, whereas no sHGG harbored the BRAF-KIAA1549 fusion. The most recurrent alterations were BRAF V600E and CDKN2A deletion in 39% and 57% of sHGGs, respectively. Importantly, all BRAF V600E and 80% of CDKN2A alterations could be traced back to their PLGG counterparts. BRAF V600E distinguished sHGG from primary HGG (P = .0023), whereas BRAF and CDKN2A alterations were less commonly observed in PLGG that did not transform (P < .001 and P < .001 respectively). PLGGs with BRAF mutations had longer latency to transformation than wild-type PLGG (median, 6.65 years [range, 3.5 to 20.3 years] v 1.59 years [range, 0.32 to 15.9 years], respectively; P = .0389). Furthermore, 5-year overall survival was 75% 15% and 29% +/- 12% for children with BRAF mutant and wild-type tumors, respectively (P = .024). Conclusion BRAF V600E mutations and CDKN2A deletions constitute a clinically distinct subtype of sHGG. The prolonged course to transformation for BRAF V600E PLGGs provides an opportunity for surgical interventions, surveillance, and targeted therapies to mitigate the outcome of sHGG. (C) 2015 by American Society of Clinical Oncology
id RCAP_a6f6f323c1c7f2ae1a7515fd0d73e0eb
oai_identifier_str oai:sapientia.ualg.pt:10400.1/11546
network_acronym_str RCAP
network_name_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository_id_str 7160
spelling BRAF Mutation and CDKN2A deletion define a clinically distinct subgroup of childhood secondary high-grade gliomaIntrinsic pontine gliomaTert promoter mutationsCentral-nervous-systemPilocytic astrocytomasGenomic landscapeHistone H3.3Fusion geneTumorsGlioblastomaDuplicationPurpose To uncover the genetic events leading to transformation of pediatric low-grade glioma (PLGG) to secondary high-grade glioma (sHGG). Patients and Methods We retrospectively identified patients with sHGG from a population-based cohort of 886 patients with PLGG with long clinical follow-up. Exome sequencing and array CGH were performed on available samples followed by detailed genetic analysis of the entire sHGG cohort. Clinical and outcome data of genetically distinct subgroups were obtained. Results sHGG was observed in 2.9% of PLGGs (26 of 886 patients). Patients with sHGG had a high frequency of nonsilent somatic mutations compared with patients with primary pediatric high-grade glioma (HGG; median, 25 mutations per exome; P = .0042). Alterations in chromatin-modifying genes and telomere-maintenance pathways were commonly observed, whereas no sHGG harbored the BRAF-KIAA1549 fusion. The most recurrent alterations were BRAF V600E and CDKN2A deletion in 39% and 57% of sHGGs, respectively. Importantly, all BRAF V600E and 80% of CDKN2A alterations could be traced back to their PLGG counterparts. BRAF V600E distinguished sHGG from primary HGG (P = .0023), whereas BRAF and CDKN2A alterations were less commonly observed in PLGG that did not transform (P < .001 and P < .001 respectively). PLGGs with BRAF mutations had longer latency to transformation than wild-type PLGG (median, 6.65 years [range, 3.5 to 20.3 years] v 1.59 years [range, 0.32 to 15.9 years], respectively; P = .0389). Furthermore, 5-year overall survival was 75% 15% and 29% +/- 12% for children with BRAF mutant and wild-type tumors, respectively (P = .024). Conclusion BRAF V600E mutations and CDKN2A deletions constitute a clinically distinct subtype of sHGG. The prolonged course to transformation for BRAF V600E PLGGs provides an opportunity for surgical interventions, surveillance, and targeted therapies to mitigate the outcome of sHGG. (C) 2015 by American Society of Clinical Oncologyb.r.a.i.n. child Canada; JPA Foundation; Canadian Institute of Health Research Grant [MOP-123268]; Hospital for Sick Children RESTRA-COMP fund; Alex's Lemonade Stand Foundation; Society of Neuro-Oncology (International Research Development Fellowship Program); National Institutes of Health [P01CA142536]; Pediatric Low-Grade Astrocytoma FoundationAmerican Society of Clinical OncologySapientiaMistry, MatthewZhukova, NataliyaMerico, DanieleRakopoulos, PatriciaKrishnatry, RahulShago, MaryStavropoulos, JamesAlon, NoaPole, Jason D.Ray, Peter N.Navickiene, VilmaMangerel, JoshuaRemke, MarcBuczkowicz, PawelRamaswamy, VijayStucklin, Ana GuerreiroLi, MartinYoung, Edwin J.Zhang, CindyCastelo-Branco, PedroBakry, DouaLaughlin, SuzanneShlien, AdamChan, JenniferLigon, Keith L.Rutka, James T.Dirks, Peter B.Taylor, Michael D.Greenberg, MarkMalkin, DavidHuang, AnnieBouffet, EricHawkins, Cynthia E.Tabori, Uri2018-12-07T14:53:30Z20152015-01-01T00:00:00Zinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/articleapplication/pdfhttp://hdl.handle.net/10400.1/11546eng0732-183X10.1200/JCO.2014.58.3922info:eu-repo/semantics/openAccessreponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãoinstacron:RCAAP2023-07-24T10:23:22Zoai:sapientia.ualg.pt:10400.1/11546Portal AgregadorONGhttps://www.rcaap.pt/oai/openaireopendoar:71602024-03-19T20:03:02.383223Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos) - Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãofalse
dc.title.none.fl_str_mv BRAF Mutation and CDKN2A deletion define a clinically distinct subgroup of childhood secondary high-grade glioma
title BRAF Mutation and CDKN2A deletion define a clinically distinct subgroup of childhood secondary high-grade glioma
spellingShingle BRAF Mutation and CDKN2A deletion define a clinically distinct subgroup of childhood secondary high-grade glioma
Mistry, Matthew
Intrinsic pontine glioma
Tert promoter mutations
Central-nervous-system
Pilocytic astrocytomas
Genomic landscape
Histone H3.3
Fusion gene
Tumors
Glioblastoma
Duplication
title_short BRAF Mutation and CDKN2A deletion define a clinically distinct subgroup of childhood secondary high-grade glioma
title_full BRAF Mutation and CDKN2A deletion define a clinically distinct subgroup of childhood secondary high-grade glioma
title_fullStr BRAF Mutation and CDKN2A deletion define a clinically distinct subgroup of childhood secondary high-grade glioma
title_full_unstemmed BRAF Mutation and CDKN2A deletion define a clinically distinct subgroup of childhood secondary high-grade glioma
title_sort BRAF Mutation and CDKN2A deletion define a clinically distinct subgroup of childhood secondary high-grade glioma
author Mistry, Matthew
author_facet Mistry, Matthew
Zhukova, Nataliya
Merico, Daniele
Rakopoulos, Patricia
Krishnatry, Rahul
Shago, Mary
Stavropoulos, James
Alon, Noa
Pole, Jason D.
Ray, Peter N.
Navickiene, Vilma
Mangerel, Joshua
Remke, Marc
Buczkowicz, Pawel
Ramaswamy, Vijay
Stucklin, Ana Guerreiro
Li, Martin
Young, Edwin J.
Zhang, Cindy
Castelo-Branco, Pedro
Bakry, Doua
Laughlin, Suzanne
Shlien, Adam
Chan, Jennifer
Ligon, Keith L.
Rutka, James T.
Dirks, Peter B.
Taylor, Michael D.
Greenberg, Mark
Malkin, David
Huang, Annie
Bouffet, Eric
Hawkins, Cynthia E.
Tabori, Uri
author_role author
author2 Zhukova, Nataliya
Merico, Daniele
Rakopoulos, Patricia
Krishnatry, Rahul
Shago, Mary
Stavropoulos, James
Alon, Noa
Pole, Jason D.
Ray, Peter N.
Navickiene, Vilma
Mangerel, Joshua
Remke, Marc
Buczkowicz, Pawel
Ramaswamy, Vijay
Stucklin, Ana Guerreiro
Li, Martin
Young, Edwin J.
Zhang, Cindy
Castelo-Branco, Pedro
Bakry, Doua
Laughlin, Suzanne
Shlien, Adam
Chan, Jennifer
Ligon, Keith L.
Rutka, James T.
Dirks, Peter B.
Taylor, Michael D.
Greenberg, Mark
Malkin, David
Huang, Annie
Bouffet, Eric
Hawkins, Cynthia E.
Tabori, Uri
author2_role author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
author
dc.contributor.none.fl_str_mv Sapientia
dc.contributor.author.fl_str_mv Mistry, Matthew
Zhukova, Nataliya
Merico, Daniele
Rakopoulos, Patricia
Krishnatry, Rahul
Shago, Mary
Stavropoulos, James
Alon, Noa
Pole, Jason D.
Ray, Peter N.
Navickiene, Vilma
Mangerel, Joshua
Remke, Marc
Buczkowicz, Pawel
Ramaswamy, Vijay
Stucklin, Ana Guerreiro
Li, Martin
Young, Edwin J.
Zhang, Cindy
Castelo-Branco, Pedro
Bakry, Doua
Laughlin, Suzanne
Shlien, Adam
Chan, Jennifer
Ligon, Keith L.
Rutka, James T.
Dirks, Peter B.
Taylor, Michael D.
Greenberg, Mark
Malkin, David
Huang, Annie
Bouffet, Eric
Hawkins, Cynthia E.
Tabori, Uri
dc.subject.por.fl_str_mv Intrinsic pontine glioma
Tert promoter mutations
Central-nervous-system
Pilocytic astrocytomas
Genomic landscape
Histone H3.3
Fusion gene
Tumors
Glioblastoma
Duplication
topic Intrinsic pontine glioma
Tert promoter mutations
Central-nervous-system
Pilocytic astrocytomas
Genomic landscape
Histone H3.3
Fusion gene
Tumors
Glioblastoma
Duplication
description Purpose To uncover the genetic events leading to transformation of pediatric low-grade glioma (PLGG) to secondary high-grade glioma (sHGG). Patients and Methods We retrospectively identified patients with sHGG from a population-based cohort of 886 patients with PLGG with long clinical follow-up. Exome sequencing and array CGH were performed on available samples followed by detailed genetic analysis of the entire sHGG cohort. Clinical and outcome data of genetically distinct subgroups were obtained. Results sHGG was observed in 2.9% of PLGGs (26 of 886 patients). Patients with sHGG had a high frequency of nonsilent somatic mutations compared with patients with primary pediatric high-grade glioma (HGG; median, 25 mutations per exome; P = .0042). Alterations in chromatin-modifying genes and telomere-maintenance pathways were commonly observed, whereas no sHGG harbored the BRAF-KIAA1549 fusion. The most recurrent alterations were BRAF V600E and CDKN2A deletion in 39% and 57% of sHGGs, respectively. Importantly, all BRAF V600E and 80% of CDKN2A alterations could be traced back to their PLGG counterparts. BRAF V600E distinguished sHGG from primary HGG (P = .0023), whereas BRAF and CDKN2A alterations were less commonly observed in PLGG that did not transform (P < .001 and P < .001 respectively). PLGGs with BRAF mutations had longer latency to transformation than wild-type PLGG (median, 6.65 years [range, 3.5 to 20.3 years] v 1.59 years [range, 0.32 to 15.9 years], respectively; P = .0389). Furthermore, 5-year overall survival was 75% 15% and 29% +/- 12% for children with BRAF mutant and wild-type tumors, respectively (P = .024). Conclusion BRAF V600E mutations and CDKN2A deletions constitute a clinically distinct subtype of sHGG. The prolonged course to transformation for BRAF V600E PLGGs provides an opportunity for surgical interventions, surveillance, and targeted therapies to mitigate the outcome of sHGG. (C) 2015 by American Society of Clinical Oncology
publishDate 2015
dc.date.none.fl_str_mv 2015
2015-01-01T00:00:00Z
2018-12-07T14:53:30Z
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
dc.type.driver.fl_str_mv info:eu-repo/semantics/article
format article
status_str publishedVersion
dc.identifier.uri.fl_str_mv http://hdl.handle.net/10400.1/11546
url http://hdl.handle.net/10400.1/11546
dc.language.iso.fl_str_mv eng
language eng
dc.relation.none.fl_str_mv 0732-183X
10.1200/JCO.2014.58.3922
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.format.none.fl_str_mv application/pdf
dc.publisher.none.fl_str_mv American Society of Clinical Oncology
publisher.none.fl_str_mv American Society of Clinical Oncology
dc.source.none.fl_str_mv reponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron:RCAAP
instname_str Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron_str RCAAP
institution RCAAP
reponame_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
collection Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository.name.fl_str_mv Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos) - Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
repository.mail.fl_str_mv
_version_ 1799133264402710528