Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
1.
Genet Med ; 24(6): 1336-1348, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35305867

RESUMO

PURPOSE: This study aimed to provide comprehensive diagnostic and candidate analyses in a pediatric rare disease cohort through the Genomic Answers for Kids program. METHODS: Extensive analyses of 960 families with suspected genetic disorders included short-read exome sequencing and short-read genome sequencing (srGS); PacBio HiFi long-read genome sequencing (HiFi-GS); variant calling for single nucleotide variants (SNV), structural variant (SV), and repeat variants; and machine-learning variant prioritization. Structured phenotypes, prioritized variants, and pedigrees were stored in PhenoTips database, with data sharing through controlled access the database of Genotypes and Phenotypes. RESULTS: Diagnostic rates ranged from 11% in patients with prior negative genetic testing to 34.5% in naive patients. Incorporating SVs from genome sequencing added up to 13% of new diagnoses in previously unsolved cases. HiFi-GS yielded increased discovery rate with >4-fold more rare coding SVs compared with srGS. Variants and genes of unknown significance remain the most common finding (58% of nondiagnostic cases). CONCLUSION: Computational prioritization is efficient for diagnostic SNVs. Thorough identification of non-SNVs remains challenging and is partly mitigated using HiFi-GS sequencing. Importantly, community research is supported by sharing real-time data to accelerate gene validation and by providing HiFi variant (SNV/SV) resources from >1000 human alleles to facilitate implementation of new sequencing platforms for rare disease diagnoses.


Assuntos
Genômica , Doenças Raras , Criança , Genoma , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Linhagem , Doenças Raras/diagnóstico , Doenças Raras/genética , Análise de Sequência de DNA
2.
Genome Res ; 27(1): 107-117, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27986820

RESUMO

The genomic alterations associated with cancers are numerous and varied, involving both isolated and large-scale complex genomic rearrangements (CGRs). Although the underlying mechanisms are not well understood, CGRs have been implicated in tumorigenesis. Here, we introduce CouGaR, a novel method for characterizing the genomic structure of amplified CGRs, leveraging both depth of coverage (DOC) and discordant pair-end mapping techniques. We applied our method to whole-genome sequencing (WGS) samples from The Cancer Genome Atlas and identify amplified CGRs in at least 5.2% (10+ copies) to 17.8% (6+ copies) of the samples. Furthermore, ∼95% of these amplified CGRs contain genes previously implicated in tumorigenesis, indicating the importance and widespread occurrence of CGRs in cancers. Additionally, CouGaR identified the occurrence of 'chromoplexy' in nearly 63% of all prostate cancer samples and 30% of all bladder cancer samples. To further validate the accuracy of our method, we experimentally tested 17 predicted fusions in two pediatric glioma samples and validated 15 of these (88%) with precise resolution of the breakpoints via qPCR experiments and Sanger sequencing, with nearly perfect copy count concordance. Additionally, to further help display and understand the structure of CGRs, we have implemented CouGaR-viz, a generic stand-alone tool for visualization of the copy count of regions, breakpoints, and relevant genes.


Assuntos
Carcinogênese/genética , Rearranjo Gênico/genética , Genoma Humano/genética , Neoplasias/genética , Biologia Computacional , Genômica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Neoplasias/patologia
3.
Acta Neuropathol ; 128(4): 573-81, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25047029

RESUMO

Diffuse intrinsic pontine glioma (DIPG) is the main cause of brain tumour-related death in children. In the majority of cases diagnosis is based on clinical and MRI findings, resulting in the scarcity of pre-treatment specimens available to study. Our group has developed an autopsy-based protocol to investigate the histologic and biologic spectrum of DIPG. This has also allowed us to investigate the terminal pattern of disease and gain a better understanding of what challenges we are facing in treating DIPG. Here, we review 72 DIPG cases with well documented clinical history and molecular data and describe the pathological features of this disease in relation to clinical and genetic features. Fifty-three of the samples were autopsy material (7 pre-treatment) and 19 were pre-treatment biopsy/surgical specimens. Upon histological review, 62 patients had high-grade astrocytomas (18 WHO grade III and 44 WHO grade IV patients), 8 had WHO grade II astrocytomas, and 2 had features of primitive neuroectodermal tumour (PNET). K27M-H3 mutations were exclusively found in tumours with WHO grade II-IV astrocytoma histology. K27M-H3.1 and ACVR1 mutations as well as ALT phenotype were only found in WHO grade III-IV astrocytomas, while PIK3CA mutations and PDGFRA gains/amplifications were found in WHO grade II-IV astrocytomas. Approximately 1/3 of DIPG patients had leptomeningeal spread of their tumour. Further, diffuse invasion of the brainstem, spinal cord and thalamus was common with some cases showing spread as distant as the frontal lobes. These findings suggest that focal radiation may be inadequate for some of these patients. Importantly, we show that clinically classic DIPGs represent a diverse histologic spectrum, including multiple cases which would fit WHO criteria of grade II astrocytoma which nevertheless behave clinically as high-grade astrocytomas and harbour the histone K27M-H3.3 mutation. This suggests that the current WHO astrocytoma grading scheme may not appropriately predict outcome for paediatric brainstem gliomas.


Assuntos
Neoplasias do Tronco Encefálico/genética , Neoplasias do Tronco Encefálico/patologia , Glioma/genética , Glioma/patologia , Histonas/genética , Mutação/genética , Adolescente , Autopsia , Neoplasias do Tronco Encefálico/mortalidade , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Glioma/mortalidade , Humanos , Lactente , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino , Tumores Neuroectodérmicos Primitivos/genética , Tumores Neuroectodérmicos Primitivos/patologia , Modelos de Riscos Proporcionais , Proteína Supressora de Tumor p53/genética
4.
Acta Neuropathol ; 128(6): 853-62, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25315281

RESUMO

Although telomeres are maintained in most cancers by telomerase activation, a subset of tumors utilize alternative lengthening of telomeres (ALT) to sustain self-renewal capacity. In order to study the prevalence and significance of ALT in childhood brain tumors we screened 517 pediatric brain tumors using the novel C-circle assay. We examined the association of ALT with alterations in genes found to segregate with specific histological phenotypes and with clinical outcome. ALT was detected almost exclusively in malignant tumors (p = 0.001). ALT was highly enriched in primitive neuroectodermal tumors (12 %), choroid plexus carcinomas (23 %) and high-grade gliomas (22 %). Furthermore, in contrast to adult gliomas, pediatric low grade gliomas which progressed to high-grade tumors did not exhibit the ALT phenotype. Somatic but not germline TP53 mutations were highly associated with ALT (p = 1.01 × 10(-8)). Of the other alterations examined, only ATRX point mutations and reduced expression were associated with the ALT phenotype (p = 0.0005). Interestingly, ALT attenuated the poor outcome conferred by TP53 mutations in specific pediatric brain tumors. Due to very poor prognosis, one year overall survival was quantified in malignant gliomas, while in children with choroid plexus carcinoma, five year overall survival was investigated. For children with TP53 mutant malignant gliomas, one year overall survival was 63 ± 12 and 23 ± 10 % for ALT positive and negative tumors, respectively (p = 0.03), while for children with TP53 mutant choroid plexus carcinomas, 5 years overall survival was 67 ± 19 and 27 ± 13 % for ALT positive and negative tumors, respectively (p = 0.07). These observations suggest that the presence of ALT is limited to a specific group of childhood brain cancers which harbor somatic TP53 mutations and may influence the outcome of these patients. Analysis of ALT may contribute to risk stratification and targeted therapies to improve outcome for these children.


Assuntos
Neoplasias Encefálicas/genética , Carcinoma/genética , Neoplasias do Plexo Corióideo/genética , Glioma/genética , Tumores Neuroectodérmicos Primitivos/genética , Telômero , Proteína Supressora de Tumor p53/genética , Adolescente , Neoplasias Encefálicas/fisiopatologia , Carcinoma/fisiopatologia , Neoplasias do Plexo Corióideo/fisiopatologia , Estudos de Coortes , DNA Helicases/genética , Glioma/fisiopatologia , Humanos , Estimativa de Kaplan-Meier , Mutação , Gradação de Tumores , Tumores Neuroectodérmicos Primitivos/fisiopatologia , Proteínas Nucleares/genética , Fenótipo , Prognóstico , Telômero/metabolismo , Proteína Nuclear Ligada ao X
5.
Acta Neuropathol ; 128(6): 863-77, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25120190

RESUMO

Pediatric ependymomas are highly recurrent tumors resistant to conventional chemotherapy. Telomerase, a ribonucleoprotein critical in permitting limitless replication, has been found to be critically important for the maintenance of tumor-initiating cells (TICs). These TICs are chemoresistant, repopulate the tumor from which they are identified, and are drivers of recurrence in numerous cancers. In this study, telomerase enzymatic activity was directly measured and inhibited to assess the therapeutic potential of targeting telomerase. Telomerase repeat amplification protocol (TRAP) (n = 36) and C-circle assay/telomere FISH/ATRX staining (n = 76) were performed on primary ependymomas to determine the prevalence and prognostic potential of telomerase activity or alternative lengthening of telomeres (ALT) as telomere maintenance mechanisms, respectively. Imetelstat, a phase 2 telomerase inhibitor, was used to elucidate the effect of telomerase inhibition on proliferation and tumorigenicity in established cell lines (BXD-1425EPN, R254), a primary TIC line (E520) and xenograft models of pediatric ependymoma. Over 60 % of pediatric ependymomas were found to rely on telomerase activity to maintain telomeres, while no ependymomas showed evidence of ALT. Children with telomerase-active tumors had reduced 5-year progression-free survival (29 ± 11 vs 64 ± 18 %; p = 0.03) and overall survival (58 ± 12 vs 83 ± 15 %; p = 0.05) rates compared to those with tumors lacking telomerase activity. Imetelstat inhibited proliferation and self-renewal by shortening telomeres and inducing senescence in vitro. In vivo, Imetelstat significantly reduced subcutaneous xenograft growth by 40 % (p = 0.03) and completely abolished the tumorigenicity of pediatric ependymoma TICs in an orthotopic xenograft model. Telomerase inhibition represents a promising therapeutic approach for telomerase-active pediatric ependymomas found to characterize high-risk ependymomas.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Inibidores Enzimáticos/farmacologia , Ependimoma/tratamento farmacológico , Indóis/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Niacinamida/análogos & derivados , Telomerase/antagonistas & inibidores , Animais , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/enzimologia , Carcinogênese/efeitos dos fármacos , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Pré-Escolar , Estudos de Coortes , Intervalo Livre de Doença , Ependimoma/diagnóstico , Ependimoma/enzimologia , Feminino , Humanos , Camundongos , Recidiva Local de Neoplasia/diagnóstico , Recidiva Local de Neoplasia/enzimologia , Transplante de Neoplasias , Células-Tronco Neoplásicas/enzimologia , Niacinamida/farmacologia , Oligonucleotídeos , Telomerase/metabolismo , Telômero/efeitos dos fármacos , Telômero/metabolismo
6.
Acta Neuropathol ; 124(3): 439-47, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22661320

RESUMO

Pediatric glioblastomas (GBM) including diffuse intrinsic pontine gliomas (DIPG) are devastating brain tumors with no effective therapy. Here, we investigated clinical and biological impacts of histone H3.3 mutations. Forty-two DIPGs were tested for H3.3 mutations. Wild-type versus mutated (K27M-H3.3) subgroups were compared for HIST1H3B, IDH, ATRX and TP53 mutations, copy number alterations and clinical outcome. K27M-H3.3 occurred in 71 %, TP53 mutations in 77 % and ATRX mutations in 9 % of DIPGs. ATRX mutations were more frequent in older children (p < 0.0001). No G34V/R-H3.3, IDH1/2 or H3.1 mutations were identified. K27M-H3.3 DIPGs showed specific copy number changes, including all gains/amplifications of PDGFRA and MYC/PVT1 loci. Notably, all long-term survivors were H3.3 wild type and this group of patients had better overall survival. K27M-H3.3 mutation defines clinically and biologically distinct subgroups and is prevalent in DIPG, which will impact future therapeutic trial design. K27M- and G34V-H3.3 have location-based incidence (brainstem/cortex) and potentially play distinct roles in pediatric GBM pathogenesis. K27M-H3.3 is universally associated with short survival in DIPG, while patients wild-type for H3.3 show improved survival. Based on prognostic and therapeutic implications, our findings argue for H3.3-mutation testing at diagnosis, which should be rapidly integrated into the clinical decision-making algorithm, particularly in atypical DIPG.


Assuntos
Neoplasias do Tronco Encefálico/genética , Glioma/genética , Histonas/genética , Ponte/patologia , Adolescente , Neoplasias do Tronco Encefálico/mortalidade , Neoplasias do Tronco Encefálico/patologia , Criança , Pré-Escolar , Feminino , Perfilação da Expressão Gênica , Glioma/mortalidade , Glioma/patologia , Humanos , Lactente , Masculino , Mutação , Prognóstico , Taxa de Sobrevida
7.
Front Oncol ; 13: 1227854, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37469406
8.
Acta Neuropathol Commun ; 4(1): 93, 2016 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-27577993

RESUMO

Paediatric brain tumours arising in the thalamus present significant diagnostic and therapeutic challenges to physicians due to their sensitive midline location. As such, genetic analysis for biomarkers to aid in the diagnosis, prognosis and treatment of these tumours is needed. Here, we identified 64 thalamic gliomas with clinical follow-up and characterized targeted genomic alterations using newly optimized droplet digital and NanoString-based assays. The median age at diagnosis was 9.25 years (range, 0.63-17.55) and median survival was 6.43 (range, 0.01-27.63) years. Our cohort contained 42 and 22 tumours reviewed as low and high grade gliomas, respectively. Five (12 %) low grade and 11 (50 %) high grade gliomas were positive for the H3F3A/HIST1H3B K27M (H3K27M) mutation. Kaplan-Meier survival analysis revealed significantly worse overall survival for patients harbouring the H3K27M mutation versus H3F3A/HIST1H3B wild type (H3WT) samples (log-rank p < 0.0001) with a median survival of 1.02 vs. 9.12 years. Mitogen-activated protein kinase (MAPK) pathway activation via BRAF or FGFR1 hotspot mutations or fusion events were detected in 44 % of patients, and was associated with long-term survival in the absence of H3K27M (log-rank p < 0.0001). Multivariate analysis demonstrated H3K27M status and high grade histology to be the most significant independent predictors of poor overall survival with hazard ratios of 6.945 and 7.721 (p < 0.0001), respectively. In contrast, MAPK pathway activation is a predictor of favourable patient outcome, although not independent of other clinical factors. Importantly, we show that low grade malignancies may harbour H3K27M mutations and that these tumours show a dismal survival compared to low grade H3WT cases. Our data strongly supports the inclusion of targeted genetic testing in childhood thalamic tumours to most accurately stratify patients into appropriate risk groups.


Assuntos
Neoplasias Encefálicas/genética , Glioma/genética , Sistema de Sinalização das MAP Quinases/genética , Tálamo , Adolescente , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/cirurgia , Criança , Pré-Escolar , Feminino , Seguimentos , Glioma/patologia , Glioma/cirurgia , Humanos , Lactente , Estimativa de Kaplan-Meier , Masculino , Análise Multivariada , Mutação , Gradação de Tumores , Prognóstico , Modelos de Riscos Proporcionais , Tálamo/patologia , Tálamo/cirurgia
9.
Acta Neuropathol Commun ; 4: 13, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26860432

RESUMO

Through inadvertent oversight of the authors, the paper failed to acknowledge funding support from Genome Canada. The Acknowledgement section should include the text: "This work was supported by the Canadian Centre for Computational Genomics (C3G), part of the Genome Innovation Network (GIN), funded by Genome Canada through Genome Quebec and Ontario Genomics".

10.
Acta Neuropathol Commun ; 4: 1, 2016 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-26727948

RESUMO

INTRODUCTION: Diffuse intrinsic pontine glioma (DIPG) and midline high-grade glioma (mHGG) are lethal childhood brain tumors. Spatial genomic heterogeneity has been well-described in adult HGG but has not been comprehensively characterized in pediatric HGG. We performed whole exome sequencing on 38-matched primary, contiguous, and metastatic tumor sites from eight children with DIPG (n = 7) or mHGG (n = 1) collected using a unique MRI-guided autopsy protocol. Validation was performed using Sanger sequencing, Droplet Digital polymerase-chain reaction, immunohistochemistry, and fluorescent in-situ hybridization. RESULTS: Median age at diagnosis was 6.1 years (range: 2.9-23.3 years). Median overall survival was 13.2 months (range: 11.2-32.2 months). Contiguous tumor infiltration and distant metastases were observed in seven and six patients, respectively, including leptomeningeal dissemination in three DIPGs. Histopathological heterogeneity was evident in seven patients, including intra-pontine heterogeneity in two DIPGs, ranging from World Health Organization grade II to IV astrocytoma. We found conservation of heterozygous K27M mutations in H3F3A (n = 4) or HIST1H3B (n = 3) across all primary, contiguous, and metastatic tumor sites in all DIPGs. ACVR1 (n = 2), PIK3CA (n = 2), FGFR1 (n = 2), and MET (n = 1) were also intra-tumorally conserved. ACVR1 was co-mutated with HIST1H3B (n = 2). In contrast, PDGFRA amplification and mutation were spatially heterogeneous, as were mutations in BCOR (n = 1), ATRX (n = 2), and MYC (n = 1). TP53 aberrations (n = 3 patients) varied by type and location between primary and metastatic tumors sites but were intra-tumorally conserved. CONCLUSION: Spatial conservation of prognostically-relevant and therapeutically-targetable somatic mutations in DIPG and mHGG contrasts the significant heterogeneity of driver mutations seen in adult HGG and supports uniform implementation of diagnostic biopsy in DIPG and mHGG to classify molecular risk groups and guide therapeutic strategy.


Assuntos
Neoplasias do Tronco Encefálico/diagnóstico , Glioma/diagnóstico , Mutação/genética , Adolescente , Biópsia , Neoplasias do Tronco Encefálico/genética , Criança , Pré-Escolar , Feminino , Perfilação da Expressão Gênica , Glioma/genética , Humanos , Imageamento por Ressonância Magnética , Masculino , Índice de Gravidade de Doença , Adulto Jovem
11.
Front Oncol ; 5: 147, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26175967

RESUMO

Diffuse intrinsic pontine glioma (DIPG) is a devastating pediatric brain cancer with no effective therapy. Histological similarity of DIPG to supratentorial high-grade astrocytomas of adults has led to assumptions that these entities possess similar underlying molecular properties and therefore similar therapeutic responses to standard therapies. The failure of all clinical trials in the last 30 years to improve DIPG patient outcome has suggested otherwise. Recent studies employing next-generation sequencing and microarray technologies have provided a breadth of evidence highlighting the unique molecular genetics and epigenetics of this cancer, distinguishing it from both adult and pediatric cerebral high-grade astrocytomas. This review describes the most common molecular genetic and epigenetic signatures of DIPG in the context of molecular subgroups and histopathological diagnosis, including this tumor entity's unique mutational landscape, copy number alterations, and structural variants, as well as epigenetic changes on the global DNA and histone levels. The increased knowledge of DIPG biology and histopathology has opened doors to new diagnostic and therapeutic avenues.

12.
Mol Cancer Ther ; 14(11): 2560-8, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26351319

RESUMO

Pediatric high-grade astrocytomas (pHGA) and diffuse intrinsic pontine gliomas (DIPG) are devastating malignancies for which no effective therapies exist. We investigated the therapeutic potential of PARP1 inhibition in preclinical models of pHGA and DIPG. PARP1 levels were characterized in pHGA and DIPG patient samples and tumor-derived cell lines. The effects of PARP inhibitors veliparib, olaparib, and niraparib as monotherapy or as radiosensitizers on cell viability, DNA damage, and PARP1 activity were evaluated in a panel of pHGA and DIPG cell lines. Survival benefit of niraparib was examined in an orthotopic xenograft model of pHGA. About 85% of pHGAs and 76% of DIPG tissue microarray samples expressed PARP1. Six of 8 primary cell lines highly expressed PARP1. Interestingly, across multiple cell lines, some PARP1 protein expression was required for response to PARP inhibition; however, there was no correlation between protein level or PARP1 activity and sensitivity to PARP inhibitors. Niraparib was the most effective at reducing cell viability and proliferation (MTT and Ki67). Niraparib induced DNA damage (γH2AX foci) and induced growth arrest. Pretreatment of pHGA cells with a sublethal dose of niraparib (1 µmol/L) before 2 Gy of ionizing radiation (IR) decreased the rate of DNA damage repair, colony growth, and relative cell number. Niraparib (50 mg/kg) inhibited PARP1 activity in vivo and extended survival of mice with orthotopic pHGA xenografts, when administered before IR (20 Gy, fractionated), relative to control mice (40 vs. 25 days). Our data provide in vitro and in vivo evidence that niraparib may be an effective radiosensitizer for pHGA and DIPG.


Assuntos
Astrocitoma/tratamento farmacológico , Neoplasias do Tronco Encefálico/tratamento farmacológico , Glioma/tratamento farmacológico , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Poli(ADP-Ribose) Polimerases/metabolismo , Animais , Astrocitoma/enzimologia , Astrocitoma/radioterapia , Benzimidazóis/farmacologia , Western Blotting , Neoplasias do Tronco Encefálico/enzimologia , Neoplasias do Tronco Encefálico/radioterapia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Criança , Terapia Combinada , Glioma/enzimologia , Glioma/radioterapia , Humanos , Indazóis/farmacologia , Estimativa de Kaplan-Meier , Modelos Lineares , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Microscopia Confocal , Ftalazinas/farmacologia , Piperazinas/farmacologia , Piperidinas/farmacologia , Poli(ADP-Ribose) Polimerase-1 , Ponte/efeitos dos fármacos , Ponte/enzimologia , Ponte/efeitos da radiação , Radioterapia/métodos , Ensaios Antitumorais Modelo de Xenoenxerto
13.
J Clin Oncol ; 33(9): 1015-22, 2015 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-25667294

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.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/secundário , Inibidor p16 de Quinase Dependente de Ciclina/genética , Deleção de Genes , Glioma/genética , Glioma/secundário , Proteínas Proto-Oncogênicas B-raf/genética , Adolescente , Transformação Celular Neoplásica , Criança , Pré-Escolar , Cromatina/química , Progressão da Doença , Feminino , Seguimentos , Humanos , Lactente , Masculino , Mutação , Mutação Puntual , Estudos Retrospectivos , Telômero/ultraestrutura , Resultado do Tratamento
14.
Cancer Discov ; 4(10): 1198-213, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25100205

RESUMO

UNLABELLED: Alkylating agents are a first-line therapy for the treatment of several aggressive cancers, including pediatric glioblastoma, a lethal tumor in children. Unfortunately, many tumors are resistant to this therapy. We sought to identify ways of sensitizing tumor cells to alkylating agents while leaving normal cells unharmed, increasing therapeutic response while minimizing toxicity. Using an siRNA screen targeting over 240 DNA damage response genes, we identified novel sensitizers to alkylating agents. In particular, the base excision repair (BER) pathway, including 3-methylpurine-DNA glycosylase (MPG), as well as ataxia telangiectasia mutated (ATM), were identified in our screen. Interestingly, we identified MPG as a direct novel substrate of ATM. ATM-mediated phosphorylation of MPG was required for enhanced MPG function. Importantly, combined inhibition or loss of MPG and ATM resulted in increased alkylating agent-induced cytotoxicity in vitro and prolonged survival in vivo. The discovery of the ATM-MPG axis will lead to improved treatment of alkylating agent-resistant tumors. SIGNIFICANCE: Inhibition of ATM and MPG-mediated BER cooperate to sensitize tumor cells to alkylating agents, impairing tumor growth in vitro and in vivo with no toxicity to normal cells, providing an ideal therapeutic window.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , DNA Glicosilases/metabolismo , Resistencia a Medicamentos Antineoplásicos , Fatores Etários , Animais , Linhagem Celular Tumoral , Análise por Conglomerados , Variações do Número de Cópias de DNA , Reparo do DNA , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/genética , Ativação Enzimática , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/mortalidade , Glioblastoma/patologia , Humanos , Modelos Biológicos , Fosforilação , Interferência de RNA , RNA Interferente Pequeno/genética , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Nat Genet ; 46(5): 451-6, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24705254

RESUMO

Diffuse intrinsic pontine glioma (DIPG) is a fatal brain cancer that arises in the brainstem of children, with no effective treatment and near 100% fatality. The failure of most therapies can be attributed to the delicate location of these tumors and to the selection of therapies on the basis of assumptions that DIPGs are molecularly similar to adult disease. Recent studies have unraveled the unique genetic makeup of this brain cancer, with nearly 80% found to harbor a p.Lys27Met histone H3.3 or p.Lys27Met histone H3.1 alteration. However, DIPGs are still thought of as one disease, with limited understanding of the genetic drivers of these tumors. To understand what drives DIPGs, we integrated whole-genome sequencing with methylation, expression and copy number profiling, discovering that DIPGs comprise three molecularly distinct subgroups (H3-K27M, silent and MYCN) and uncovering a new recurrent activating mutation affecting the activin receptor gene ACVR1 in 20% of DIPGs. Mutations in ACVR1 were constitutively activating, leading to SMAD phosphorylation and increased expression of the downstream activin signaling targets ID1 and ID2. Our results highlight distinct molecular subgroups and novel therapeutic targets for this incurable pediatric cancer.


Assuntos
Receptores de Ativinas Tipo I/genética , Neoplasias do Tronco Encefálico/genética , Regulação Neoplásica da Expressão Gênica/genética , Genoma Humano/genética , Glioma/genética , Animais , Neoplasias do Tronco Encefálico/classificação , Criança , Variações do Número de Cópias de DNA , Metilação de DNA , Perfilação da Expressão Gênica , Glioma/classificação , Humanos , Proteína 1 Inibidora de Diferenciação/metabolismo , Proteína 2 Inibidora de Diferenciação/metabolismo , Fosforilação , Análise de Sequência de DNA , Proteínas Smad/metabolismo , Peixe-Zebra
16.
Brain Pathol ; 23(3): 244-53, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-22971244

RESUMO

Pediatric high-grade astrocytomas (HGAs) account for 15-20% of all pediatric central nervous system tumors. These neoplasms predominantly involve the supratentorial hemispheres or the pons--diffuse intrinsic pontine gliomas (DIPG). Assumptions that pediatric HGAs are biologically similar to adult HGAs have recently been challenged, and the development of effective therapeutic modalities for DIPG and supratentorial HGA hinges on a better understanding of their biological properties. Here, 20 pediatric HGAs (9 DIPGs and 11 supratentorial HGAs) were subject to gene expression profiling following approval by the research ethics board at our institution. Many of these tumors showed expression signatures composed of genes that promote G1/S and G2/M cell cycle progression. In particular, Aurora kinase B (AURKB) was consistently and highly overexpressed in 6/9 DIPGs and 8/11 HGAs. Array data were validated using quantitative real-time PCR and immunohistochemistry, as well as cross-validation of our data set with previously published series. Inhibition of Aurora B activity in DIPG and in pediatric HGA cell lines resulted in growth arrest accompanied by morphological changes, cell cycle aberrations, nuclear fractionation and polyploidy as well as a reduction in colony formation. Our data highlight Aurora B as a potential therapeutic target in DIPG.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Neoplasias do Tronco Encefálico/tratamento farmacológico , Neoplasias do Tronco Encefálico/enzimologia , Glioma/tratamento farmacológico , Glioma/enzimologia , Proteínas Serina-Treonina Quinases/efeitos dos fármacos , Aurora Quinase B , Aurora Quinases , Western Blotting , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Criança , Imunofluorescência , Humanos , Imuno-Histoquímica , Reação em Cadeia da Polimerase em Tempo Real
17.
J Neuropathol Exp Neurol ; 70(6): 430-7, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21572341

RESUMO

To determine whether the zinc finger transcription factors GLI1 to GLI3 and suppressor of fused (SUFU) components of the Sonic hedgehog signaling pathway may be prognostic markers and potential therapeutic targets in pediatric medulloblastoma (MB), we investigated the relationship of the expression of these proteins to prognosis in the MB of 124 patients who had undergone surgery at the Hospital for Sick Children (Toronto, Ontario, Canada). The expressions of GLI1 (p = 0.011) and GLI2 (p = 0.003), but not of GLI3 (p = 0.774) or SUFU (p = 0.137), in the MB were associated with a worse overall survival by Kaplan-Meier analysis. Overall survival of patients positive for GLI1 and GLI2 was 6.01 ± 0.85 years and 5.27 ± 1.44 years, respectively, versus 10.11 ± 1.52 years and 10.18 ± 0.22 years for patients negative for GLI1 and GLI2, respectively. Knockdown of GLI2 in 3 MB cell lines resulted in decreased cell number and viability, as determined by the MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay; knockdown of GLI1 had no effect. The decrease in cell number with GLI2 knockdown was caused by G0 cell cycle arrest; there was no induction of apoptosis. These results suggest that targeting the Sonic hedgehog pathway in positive patients may be a useful adjuvant therapeutic strategy for MB.


Assuntos
Apoptose/fisiologia , Neoplasias Cerebelares/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Meduloblastoma/metabolismo , Proteínas Nucleares/metabolismo , Adolescente , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Neoplasias Cerebelares/patologia , Criança , Pré-Escolar , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Marcação In Situ das Extremidades Cortadas/métodos , Lactente , Fatores de Transcrição Kruppel-Like/genética , Masculino , Meduloblastoma/patologia , Proteínas Nucleares/genética , Pediatria , RNA Interferente Pequeno/farmacologia , Estudos Retrospectivos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transdução Genética/métodos , Proteína GLI1 em Dedos de Zinco , Proteína Gli2 com Dedos de Zinco
18.
J Clin Oncol ; 28(8): 1337-44, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20142589

RESUMO

PURPOSE: Diffuse intrinsic pontine glioma (DIPG) is one of the most devastating of pediatric malignancies and one for which no effective therapy exists. A major contributor to the failure of therapeutic trials is the assumption that biologic properties of brainstem tumors in children are identical to cerebral high-grade gliomas of adults. A better understanding of the biology of DIPG itself is needed in order to develop agents targeted more specifically to these children's disease. Herein, we address this lack of knowledge by performing the first high-resolution single nucleotide polymorphism (SNP) -based DNA microarray analysis of a series of DIPGs. PATIENTS AND METHODS: Eleven samples (nine postmortem and two pretreatment surgical samples), the largest series thus far examined, were hybridized to SNP arrays (250 k or 6.0). The study was approved by the research ethics board at our institution. All array findings were validated using quantitative polymerase chain reaction, fluorescence in situ hybridization, immunohistochemistry, and/or microsatellite analysis. RESULTS: Analysis of DIPG copy number alterations showed recurrent changes distinct from those of pediatric supratentorial high-grade astrocytomas. Thirty-six percent of DIPGs had gains in platelet-derived growth factor receptor alpha (PDGFRA; 4 to 18 copies) and all showed PDGFR-alpha expression. Low-level gains in poly (ADP-ribose) polymerase (PARP) -1 were identified in three cases. Pathway analysis revealed genes with loss of heterozygosity were enriched for DNA repair pathways. CONCLUSION: To our knowledge, our data provides the first, comprehensive high-resolution genomic analysis of pediatric DIPG. Our findings of recurrent involvement of the PDGFR pathway as well as defects in DNA repair pathways coupled with gain of PARP-1 highlight two potential, biologically based, therapeutic targets directed specifically at this devastating disease.


Assuntos
Neoplasias do Tronco Encefálico/genética , Glioma/genética , Poli(ADP-Ribose) Polimerases/genética , Polimorfismo de Nucleotídeo Único , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Autopsia , Neoplasias do Tronco Encefálico/patologia , Estudos de Casos e Controles , Criança , Pré-Escolar , Reparo do DNA/genética , DNA de Neoplasias/análise , Feminino , Dosagem de Genes , Glioma/patologia , Humanos , Lactente , Recém-Nascido , Perda de Heterozigosidade , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Poli(ADP-Ribose) Polimerase-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA