Show simple item record

dc.contributor.authorKoschmann, C
dc.contributor.authorZamler, D
dc.contributor.authorMacKay, A
dc.contributor.authorRobinson, D
dc.contributor.authorWu, Y-M
dc.contributor.authorDoherty, R
dc.contributor.authorMarini, B
dc.contributor.authorTran, D
dc.contributor.authorGarton, H
dc.contributor.authorMuraszko, K
dc.contributor.authorRobertson, P
dc.contributor.authorLeonard, M
dc.contributor.authorZhao, L
dc.contributor.authorBixby, D
dc.contributor.authorPeterson, L
dc.contributor.authorCamelo-Piragua, S
dc.contributor.authorJones, C
dc.contributor.authorMody, R
dc.contributor.authorLowenstein, PR
dc.contributor.authorCastro, MG
dc.date.accessioned2016-09-28T09:35:43Z
dc.date.issued2016-10-04
dc.identifier.citationOncotarget, 2016, 7 (40), pp. 65696 - 65706
dc.identifier.issn1949-2553
dc.identifier.urihttps://repository.icr.ac.uk/handle/internal/123
dc.identifier.eissn1949-2553
dc.identifier.doi10.18632/oncotarget.11602
dc.description.abstractPediatric high-grade glioma (HGG, WHO Grade III and IV) is a devastating brain tumor with a median survival of less than two years. PDGFRA is frequently mutated/ amplified in pediatric HGG, but the significance of this finding has not been fully characterized. We hypothesize that alterations of PDGFRA will promote distinct prognostic and treatment implications in pediatric HGG. In order to characterize the impact of PDGFR pathway alterations, we integrated genomic data from pediatric HGG patients (n=290) from multiple pediatric datasets and sequencing platforms. Integration of multiple human datasets showed that PDGFRA mutation, but not amplification, was associated with older age in pediatric HGG (P= <0.0001). In multivariate analysis, PDGFRA mutation was correlated with worse prognosis (P = 0.026), while PDGFRA amplification was not (P = 0.11). By Kaplan-Meier analysis, non-brainstem HGG with PDGFRA amplification carried a worse prognosis than non-brainstem HGG without PDGFRA amplification (P = 0.021). There were no pediatric patients with PDGFRA-amplified HGG that survived longer than two years. Additionally, we performed paired molecular profiling (germline / tumor / primary cell culture) and targeting of an infant thalamic HGG with amplification and outlier increased expression of PDGFRA. Dasatinib inhibited proliferation most effectively. In summary, integration of the largest genomic dataset of pediatric HGG to date, allowed us to highlight that PDGFRA mutation is found in older pediatric patients and that PDGFRA amplification is prognostic in non-brainstem HGG. Future precision-medicine based clinical trials for pediatric patients with PDGFRA-altered HGG should consider the optimized delivery of dasatinib.
dc.formatPrint
dc.format.extent65696 - 65706
dc.languageeng
dc.language.isoeng
dc.publisherIMPACT JOURNALS LLC
dc.rights.urihttps://creativecommons.org/licenses/by/4.0
dc.subjectTumor Cells, Cultured
dc.subjectHumans
dc.subjectGlioma
dc.subjectBrain Neoplasms
dc.subjectReceptor, Platelet-Derived Growth Factor alpha
dc.subjectAntineoplastic Agents
dc.subjectSurvival Rate
dc.subjectFollow-Up Studies
dc.subjectAge Factors
dc.subjectApoptosis
dc.subjectCell Proliferation
dc.subjectGene Amplification
dc.subjectMutation
dc.subjectAdolescent
dc.subjectAdult
dc.subjectChild
dc.subjectChild, Preschool
dc.subjectInfant
dc.subjectFemale
dc.subjectMale
dc.subjectYoung Adult
dc.subjectNeoplasm Grading
dc.subjectBiomarkers, Tumor
dc.titleCharacterizing and targeting PDGFRA alterations in pediatric high-grade glioma.
dc.typeJournal Article
dcterms.dateAccepted2016-08-13
rioxxterms.versionofrecord10.18632/oncotarget.11602
rioxxterms.licenseref.urihttps://creativecommons.org/licenses/by/4.0
rioxxterms.licenseref.startdate2016-10
rioxxterms.typeJournal Article/Review
dc.relation.isPartOfOncotarget
pubs.issue40
pubs.notesNot known
pubs.organisational-group/ICR
pubs.organisational-group/ICR/Primary Group
pubs.organisational-group/ICR/Primary Group/ICR Divisions
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Cancer Therapeutics
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Cancer Therapeutics/Glioma Team
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Molecular Pathology
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Molecular Pathology/Glioma Team
pubs.organisational-group/ICR
pubs.organisational-group/ICR/Primary Group
pubs.organisational-group/ICR/Primary Group/ICR Divisions
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Cancer Therapeutics
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Cancer Therapeutics/Glioma Team
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Molecular Pathology
pubs.organisational-group/ICR/Primary Group/ICR Divisions/Molecular Pathology/Glioma Team
pubs.publication-statusPublished
pubs.volume7
pubs.embargo.termsNot known
icr.researchteamGlioma Team
dc.contributor.icrauthorMackay, Alan
dc.contributor.icrauthorJones, Chris


Files in this item

Thumbnail
Thumbnail

This item appears in the following collection(s)

Show simple item record

https://creativecommons.org/licenses/by/4.0
Except where otherwise noted, this item's license is described as https://creativecommons.org/licenses/by/4.0