Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 25(22): 6788-6800, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31481512

ABSTRACT

PURPOSE: Diffuse intrinsic pontine gliomas (DIPG) are the most severe pediatric brain tumors. Although accepted as the standard therapeutic, radiotherapy is only efficient transiently and not even in every patient. The goal of the study was to identify the underlying molecular determinants of response to radiotherapy in DIPG. EXPERIMENTAL DESIGN: We assessed in vitro response to ionizing radiations in 13 different DIPG cellular models derived from treatment-naïve stereotactic biopsies reflecting the genotype variability encountered in patients at diagnosis and correlated it to their principal molecular alterations. Clinical and radiologic response to radiotherapy of a large cohort of 73 DIPG was analyzed according to their genotype. Using a kinome-wide synthetic lethality RNAi screen, we further identified target genes that can sensitize DIPG cells to ionizing radiations. RESULTS: We uncover TP53 mutation as the main driver of increased radioresistance and validated this finding in four isogenic pairs of TP53WT DIPG cells with or without TP53 knockdown. In an integrated clinical, radiological, and molecular study, we show that TP53MUT DIPG patients respond less to irradiation, relapse earlier after radiotherapy, and have a worse prognosis than their TP53WT counterparts. Finally, a kinome-wide synthetic lethality RNAi screen identifies CHK1 as a potential target, whose inhibition increases response to radiation specifically in TP53MUT cells. CONCLUSIONS: Here, we demonstrate that TP53 mutations are driving DIPG radioresistance both in patients and corresponding cellular models. We suggest alternative treatment strategies to mitigate radioresistance with CHK1 inhibitors. These findings will allow to consequently refine radiotherapy schedules in DIPG.


Subject(s)
Brain Stem Neoplasms/metabolism , Diffuse Intrinsic Pontine Glioma/metabolism , Radiation Tolerance , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Brain Stem Neoplasms/genetics , Brain Stem Neoplasms/mortality , Brain Stem Neoplasms/radiotherapy , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Survival/radiation effects , Diffuse Intrinsic Pontine Glioma/genetics , Diffuse Intrinsic Pontine Glioma/mortality , Diffuse Intrinsic Pontine Glioma/radiotherapy , Dose-Response Relationship, Radiation , Gene Knockdown Techniques , Histones/genetics , Histones/metabolism , Humans , Mutation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Prognosis , RNA Interference , RNA, Small Interfering/genetics , Radiation Tolerance/genetics , Tumor Suppressor Protein p53/genetics
2.
Oncogene ; 38(38): 6479-6490, 2019 09.
Article in English | MEDLINE | ID: mdl-31324890

ABSTRACT

Diffuse intrinsic pontine glioma (or DIPG) are pediatric high-grade gliomas associated with a dismal prognosis. They harbor specific substitution in histone H3 at position K27 that induces major epigenetic dysregulations. Most clinical trials failed so far to increase survival, and radiotherapy remains the most efficient treatment, despite only transient tumor control. We conducted the first lentiviral shRNA dropout screen in newly diagnosed DIPG to generate a cancer-lethal signature as a basis for the development of specific treatments with increased efficacy and reduced side effects compared to existing anticancer therapies. The analysis uncovered 41 DIPG essential genes among the 672 genes of human kinases tested, for which several distinct interfering RNAs impaired cell expansion of three different DIPG stem-cell cultures without deleterious effect on two control neural stem cells. Among them, PLK1, AURKB, CHEK1, EGFR, and GSK3A were previously identified by similar approach in adult GBM indicating common dependencies of these cancer cells and pediatric gliomas. As expected, we observed an enrichment of genes involved in proliferation and cell death processes with a significant number of candidates belonging to PTEN/PI3K/AKT and EGFR pathways already under scrutiny in clinical trials in this disease. We highlighted VRK3, a gene involved especially in cell cycle regulation, DNA repair, and neuronal differentiation, as a non-oncogenic addiction in DIPG. Its repression totally blocked DIPG cell growth in the four cellular models evaluated, and induced cell death in H3.3-K27M cells specifically but not in H3.1-K27M cells, supporting VRK3 as an interesting and promising target in DIPG.


Subject(s)
Brain Stem Neoplasms/genetics , Diffuse Intrinsic Pontine Glioma/genetics , Phosphotransferases/genetics , Protein Serine-Threonine Kinases/physiology , RNA, Small Interfering/physiology , Sequence Analysis, RNA/methods , Brain Stem Neoplasms/diagnosis , Brain Stem Neoplasms/pathology , Cell Survival/genetics , Cells, Cultured , Diffuse Intrinsic Pontine Glioma/diagnosis , Diffuse Intrinsic Pontine Glioma/pathology , Genes, Essential , HEK293 Cells , Humans , Phosphatidylinositol 3-Kinases/analysis , Phosphatidylinositol 3-Kinases/genetics , Phosphotransferases/analysis , Prognosis , Protein Serine-Threonine Kinases/analysis , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/analysis
3.
Acta Neuropathol Commun ; 6(1): 117, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30396367

ABSTRACT

Diffuse midline glioma (DMG), H3 K27M-mutant, is a new entity in the updated WHO classification grouping together diffuse intrinsic pontine gliomas and infiltrating glial neoplasms of the midline harboring the same canonical mutation at the Lysine 27 of the histones H3 tail.Two hundred and fifteen patients younger than 18 years old with centrally-reviewed pediatric high-grade gliomas (pHGG) were included in this study. Comprehensive transcriptomic (n = 140) and methylation (n = 80) profiling was performed depending on the material available, in order to assess the biological uniqueness of this new entity compared to other midline and hemispheric pHGG.Tumor classification based on gene expression (GE) data highlighted the similarity of K27M DMG independently of their location along the midline. T-distributed Stochastic Neighbor Embedding (tSNE) analysis of methylation profiling confirms the discrimination of DMG from other well defined supratentorial tumor subgroups. Patients with diffuse intrinsic pontine gliomas (DIPG) and thalamic DMG exhibited a similarly poor prognosis (11.1 and 10.8 months median overall survival, respectively). Interestingly, H3.1-K27M and H3.3-K27M primary tumor samples could be distinguished based both on their GE and DNA methylation profiles, suggesting that they might arise from a different precursor or from a different epigenetic reorganization.These differences in DNA methylation profiles were conserved in glioma stem-like cell culture models of DIPG which mimicked their corresponding primary tumor. ChIP-seq profiling of H3K27me3 in these models indicate that H3.3-K27M mutated DIPG stem cells exhibit higher levels of H3K27 trimethylation which are correlated with fewer genes expressed by RNAseq. When considering the global distribution of the H3K27me3 mark, we observed that intergenic regions were more trimethylated in the H3.3-K27M mutated cells compared to the H3.1-K27M mutated ones.H3 K27M-mutant DMG represent a homogenous group of neoplasms compared to other pediatric gliomas that could be further separated based on the type of histone H3 variant mutated and their respective epigenetic landscapes. As these characteristics drive different phenotypes, these findings may have important implication for the design of future trials in these specific types of neoplasms.


Subject(s)
Brain Neoplasms/genetics , Epigenomics , Glioma/genetics , Histones/genetics , Mutation/genetics , Transcriptome/physiology , Adolescent , Brain/pathology , Brain Neoplasms/pathology , Child , Child, Preschool , DNA Methylation/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Glioma/pathology , Humans , Lysine/genetics , Male , Methionine/genetics , Principal Component Analysis , Tumor Cells, Cultured
4.
Brain Pathol ; 28(1): 103-111, 2018 Jan.
Article in English | MEDLINE | ID: mdl-27984673

ABSTRACT

Ganglioglioma (GG) is a grade I tumor characterized by alterations in the MAPK pathway, including BRAF V600E mutation. Recently, diffuse midline glioma with an H3 K27M mutation was added to the WHO 2016 classification as a new grade IV entity. As co-occurrence of H3 K27M and BRAF V600E mutations has been reported in midline tumors and anaplastic GG, we searched for BRAF V600E and H3 K27M mutations in a series of 54 paediatric midline grade I GG (midline GG) to determine the frequency of double mutations and its relevance for prognosis. Twenty-seven patients (50%) possessed the BRAF V600E mutation. The frequency of the co-occurrence of H3F3A/BRAF mutations at diagnosis was 9.3%. No H3 K27M mutation was detected in the absence of the BRAF V600E mutation. Double-immunostaining revealed that BRAF V600E and H3 K27M mutant proteins were present in both the glial and neuronal components. Immunopositivity for the BRAF V600E mutant protein correlated with BRAF mutation status as detected by massARRAY or digital droplet PCR. The median follow-up of patients with double mutation was 4 years. One patient died of progressive disease 8 years after diagnosis, whereas the four other patients were all alive with stable disease at the last clinical follow-up (at 9 months, 1 year and 7 years) without adjuvant therapy. We demonstrate in this first series of midline GGs that the H3 K27M mutation can occur in association with the BRAF V600E mutation in grade I glioneuronal tumors. Despite the presence of H3 K27M mutations, these cases should not be graded and treated as grade IV tumors because they have a better spontaneous outcome than classic diffuse midline H3 K27M-mutant glioma. These data suggest that H3 K27M cannot be considered a specific hallmark of grade IV diffuse gliomas and highlight the importance of integrated histomolecular diagnosis in paediatric brain tumors.


Subject(s)
Brain Neoplasms/genetics , Ganglioglioma/genetics , Histones/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Spinal Cord Neoplasms/genetics , Adolescent , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Child , Child, Preschool , Female , Follow-Up Studies , Ganglioglioma/diagnostic imaging , Ganglioglioma/pathology , Ganglioglioma/therapy , Humans , Immunohistochemistry , Male , Neoplasm Grading , Spinal Cord Neoplasms/diagnostic imaging , Spinal Cord Neoplasms/pathology , Spinal Cord Neoplasms/therapy , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...