Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 4.112
1.
Acta Neuropathol Commun ; 12(1): 72, 2024 May 06.
Article En | MEDLINE | ID: mdl-38711090

The RE1-silencing transcription factor (REST) acts either as a repressor or activator of transcription depending on the genomic and cellular context. REST is a key player in brain cell differentiation by inducing chromatin modifications, including DNA methylation, in a proximity of its binding sites. Its dysfunction may contribute to oncogenesis. Mutations in IDH1/2 significantly change the epigenome contributing to blockade of cell differentiation and glioma development. We aimed at defining how REST modulates gene activation and repression in the context of the IDH mutation-related phenotype in gliomas. We studied the effects of REST knockdown, genome wide occurrence of REST binding sites, and DNA methylation of REST motifs in IDH wild type and IDH mutant gliomas. We found that REST target genes, REST binding patterns, and TF motif occurrence proximal to REST binding sites differed in IDH wild-type and mutant gliomas. Among differentially expressed REST targets were genes involved in glial cell differentiation and extracellular matrix organization, some of which were differentially methylated at promoters or gene bodies. REST knockdown differently impacted invasion of the parental or IDH1 mutant glioma cells. The canonical REST-repressed gene targets showed significant correlation with the GBM NPC-like cellular state. Interestingly, results of REST or KAISO silencing suggested the interplay between these TFs in regulation of REST-activated and repressed targets. The identified gene regulatory networks and putative REST cooperativity with other TFs, such as KAISO, show distinct REST target regulatory networks in IDH-WT and IDH-MUT gliomas, without concomitant DNA methylation changes. We conclude that REST could be an important therapeutic target in gliomas.


Brain Neoplasms , DNA Methylation , Gene Regulatory Networks , Glioma , Isocitrate Dehydrogenase , Mutation , Isocitrate Dehydrogenase/genetics , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Humans , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Cell Line, Tumor , Repressor Proteins/genetics , Repressor Proteins/metabolism , Gene Expression Regulation, Neoplastic/genetics
2.
Radiology ; 311(2): e233120, 2024 May.
Article En | MEDLINE | ID: mdl-38713025

Background According to 2021 World Health Organization criteria, adult-type diffuse gliomas include glioblastoma, isocitrate dehydrogenase (IDH)-wildtype; oligodendroglioma, IDH-mutant and 1p/19q-codeleted; and astrocytoma, IDH-mutant, even when contrast enhancement is lacking. Purpose To develop and validate simple scoring systems for predicting IDH and subsequent 1p/19q codeletion status in gliomas without contrast enhancement using standard clinical MRI sequences. Materials and Methods This retrospective study included adult-type diffuse gliomas lacking contrast at contrast-enhanced MRI from two tertiary referral hospitals between January 2012 and April 2022 with diagnoses confirmed at pathology. IDH status was predicted primarily by using T2-fluid-attenuated inversion recovery (FLAIR) mismatch sign, followed by 1p/19q codeletion prediction. A visual rating of MRI features, apparent diffusion coefficient (ADC) ratio, and relative cerebral blood volume was measured. Scoring systems were developed through univariable and multivariable logistic regressions and underwent calibration and discrimination, including internal and external validation. Results For the internal validation cohort, 237 patients were included (mean age, 44.4 years ± 14.4 [SD]; 136 male patients; 193 patients in IDH prediction and 163 patients in 1p/19q prediction). For the external validation cohort, 35 patients were included (46.1 years ± 15.3; 20 male patients; 28 patients in IDH prediction and 24 patients in 1p/19q prediction). The T2-FLAIR mismatch sign demonstrated 100% specificity and 100% positive predictive value for IDH mutation. IDH status prediction scoring system for tumors without mismatch sign included age, ADC ratio, and morphologic characteristics, whereas 1p/19q codeletion prediction for IDH-mutant gliomas included ADC ratio, cortical involvement, and mismatch sign. For IDH status and 1p/19q codeletion prediction, bootstrap-corrected areas under the receiver operating characteristic curve were 0.86 (95% CI: 0.81, 0.90) and 0.73 (95% CI: 0.65, 0.81), respectively, whereas at external validation they were 0.99 (95% CI: 0.98, 1.0) and 0.88 (95% CI: 0.63, 1.0). Conclusion The T2-FLAIR mismatch sign and scoring systems using standard clinical MRI predicted IDH and 1p/19q codeletion status in gliomas lacking contrast enhancement. © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Badve and Hodges in this issue.


Brain Neoplasms , Chromosomes, Human, Pair 1 , Glioma , Isocitrate Dehydrogenase , Magnetic Resonance Imaging , Mutation , Humans , Isocitrate Dehydrogenase/genetics , Male , Female , Adult , Glioma/genetics , Glioma/diagnostic imaging , Retrospective Studies , Magnetic Resonance Imaging/methods , Brain Neoplasms/genetics , Brain Neoplasms/diagnostic imaging , Chromosomes, Human, Pair 1/genetics , Middle Aged , Chromosomes, Human, Pair 19/genetics , Contrast Media , Chromosome Deletion
3.
Am Soc Clin Oncol Educ Book ; 44(3): e431450, 2024 Jun.
Article En | MEDLINE | ID: mdl-38723228

Low-grade gliomas present a formidable challenge in neuro-oncology because of the challenges imposed by the blood-brain barrier, predilection for the young adult population, and propensity for recurrence. In the past two decades, the systematic examination of genomic alterations in adults and children with primary brain tumors has uncovered profound new insights into the pathogenesis of these tumors, resulting in more accurate tumor classification and prognostication. It also identified several common recurrent genomic alterations that now define specific brain tumor subtypes and have provided a new opportunity for molecularly targeted therapeutic intervention. Adult-type diffuse low-grade gliomas are frequently associated with mutations in isocitrate dehydrogenase 1 and 2 (IDH1/2), resulting in production of 2-hydroxyglutarate, an oncometabolite important for tumorigenesis. Recent studies of IDH inhibitors have yielded promising results in patients at early stages of disease with prolonged progression-free survival (PFS) and delayed time to radiation and chemotherapy. Pediatric-type gliomas have high rates of alterations in BRAF, including BRAF V600E point mutations or BRAF-KIAA1549 rearrangements. BRAF inhibitors, often combined with MEK inhibitors, have resulted in radiographic response and improved PFS in these patients. This article reviews emerging approaches to the treatment of low-grade gliomas, including a discussion of targeted therapies and how they integrate with the current treatment modalities of surgical resection, chemotherapy, and radiation.


Brain Neoplasms , Glioma , Isocitrate Dehydrogenase , Neoplasm Grading , Humans , Glioma/genetics , Glioma/therapy , Glioma/drug therapy , Glioma/pathology , Isocitrate Dehydrogenase/genetics , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/drug therapy , Disease Management , Mutation , Molecular Targeted Therapy
4.
Nat Rev Dis Primers ; 10(1): 33, 2024 May 09.
Article En | MEDLINE | ID: mdl-38724526

Gliomas are primary brain tumours that are thought to develop from neural stem or progenitor cells that carry tumour-initiating genetic alterations. Based on microscopic appearance and molecular characteristics, they are classified according to the WHO classification of central nervous system (CNS) tumours and graded into CNS WHO grades 1-4 from a low to high grade of malignancy. Diffusely infiltrating gliomas in adults comprise three tumour types with distinct natural course of disease, response to treatment and outcome: isocitrate dehydrogenase (IDH)-mutant and 1p/19q-codeleted oligodendrogliomas with the best prognosis; IDH-mutant astrocytomas with intermediate outcome; and IDH-wild-type glioblastomas with poor prognosis. Pilocytic astrocytoma is the most common glioma in children and is characterized by circumscribed growth, frequent BRAF alterations and favourable prognosis. Diffuse gliomas in children are divided into clinically indolent low-grade tumours and high-grade tumours with aggressive behaviour, with histone 3 K27-altered diffuse midline glioma being the leading cause of glioma-related death in children. Ependymal tumours are subdivided into biologically and prognostically distinct types on the basis of histology, molecular biomarkers and location. Although surgery, radiotherapy and alkylating agent chemotherapy are the mainstay of glioma treatment, individually tailored strategies based on tumour-intrinsic dominant signalling pathways have improved outcome in subsets of patients.


Brain Neoplasms , Glioma , Humans , Glioma/genetics , Glioma/physiopathology , Glioma/therapy , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/diagnosis , Brain Neoplasms/physiopathology , Prognosis , Child , Isocitrate Dehydrogenase/genetics , Mutation
5.
Nat Commun ; 15(1): 3785, 2024 May 06.
Article En | MEDLINE | ID: mdl-38710674

Mutations in human isocitrate dehydrogenase 1 (IDH1) drive tumor formation in a variety of cancers by replacing its conventional activity with a neomorphic activity that generates an oncometabolite. Little is understood of the mechanistic differences among tumor-driving IDH1 mutants. We previously reported that the R132Q mutant unusually preserves conventional activity while catalyzing robust oncometabolite production, allowing an opportunity to compare these reaction mechanisms within a single active site. Here, we employ static and dynamic structural methods and observe that, compared to R132H, the R132Q active site adopts a conformation primed for catalysis with optimized substrate binding and hydride transfer to drive improved conventional and neomorphic activity over R132H. This active site remodeling reveals a possible mechanism of resistance to selective mutant IDH1 therapeutic inhibitors. This work enhances our understanding of fundamental IDH1 mechanisms while pinpointing regions for improving inhibitor selectivity.


Catalytic Domain , Isocitrate Dehydrogenase , Mutation , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Humans , Kinetics , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/pharmacology
6.
BMC Med Imaging ; 24(1): 104, 2024 May 03.
Article En | MEDLINE | ID: mdl-38702613

BACKGROUND: The role of isocitrate dehydrogenase (IDH) mutation status for glioma stratification and prognosis is established. While structural magnetic resonance image (MRI) is a promising biomarker, it may not be sufficient for non-invasive characterisation of IDH mutation status. We investigated the diagnostic value of combined diffusion tensor imaging (DTI) and structural MRI enhanced by a deep radiomics approach based on convolutional neural networks (CNNs) and support vector machine (SVM), to determine the IDH mutation status in Central Nervous System World Health Organization (CNS WHO) grade 2-4 gliomas. METHODS: This retrospective study analyzed the DTI-derived fractional anisotropy (FA) and mean diffusivity (MD) images and structural images including fluid attenuated inversion recovery (FLAIR), non-enhanced T1-, and T2-weighted images of 206 treatment-naïve gliomas, including 146 IDH mutant and 60 IDH-wildtype ones. The lesions were manually segmented by experienced neuroradiologists and the masks were applied to the FA and MD maps. Deep radiomics features were extracted from each subject by applying a pre-trained CNN and statistical description. An SVM classifier was applied to predict IDH status using imaging features in combination with demographic data. RESULTS: We comparatively assessed the CNN-SVM classifier performance in predicting IDH mutation status using standalone and combined structural and DTI-based imaging features. Combined imaging features surpassed stand-alone modalities for the prediction of IDH mutation status [area under the curve (AUC) = 0.846; sensitivity = 0.925; and specificity = 0.567]. Importantly, optimal model performance was noted following the addition of demographic data (patients' age) to structural and DTI imaging features [area under the curve (AUC) = 0.847; sensitivity = 0.911; and specificity = 0.617]. CONCLUSIONS: Imaging features derived from DTI-based FA and MD maps combined with structural MRI, have superior diagnostic value to that provided by standalone structural or DTI sequences. In combination with demographic information, this CNN-SVM model offers a further enhanced non-invasive prediction of IDH mutation status in gliomas.


Brain Neoplasms , Diffusion Tensor Imaging , Glioma , Isocitrate Dehydrogenase , Mutation , Humans , Isocitrate Dehydrogenase/genetics , Glioma/diagnostic imaging , Glioma/genetics , Glioma/pathology , Diffusion Tensor Imaging/methods , Retrospective Studies , Male , Female , Middle Aged , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Adult , Aged , Neoplasm Grading , Support Vector Machine , Magnetic Resonance Imaging/methods , Neural Networks, Computer , Radiomics
7.
Folia Neuropathol ; 62(1): 96-101, 2024.
Article En | MEDLINE | ID: mdl-38741436

Gliosarcoma (GS) is a rare variant of IDH-wildtype glioblastoma. It is classified as grade 4 in the latest WHO CNS classification of both glial and mesenchymal components. Gliosarcoma may arise de novo or secondary from glioblastoma. It occurs in up to 2% of patients diagnosed with glioblastoma. We present a case report of a 51-year-old patient who was initially diagnosed with glioblastoma multiforme, which transformed into secondary gliosarcoma with an osteosarcoma component 16 months after the initial diagnosis. We believe that increasing reporting of secondary gliosarcoma (sGS) will be helpful in understanding, diagnosing and providing more effective treatment for this cancer.


Brain Neoplasms , Glioblastoma , Gliosarcoma , Isocitrate Dehydrogenase , Osteosarcoma , Humans , Glioblastoma/genetics , Glioblastoma/pathology , Gliosarcoma/genetics , Gliosarcoma/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Osteosarcoma/genetics , Osteosarcoma/pathology , Middle Aged , Isocitrate Dehydrogenase/genetics , Male
9.
Cereb Cortex ; 34(4)2024 Apr 01.
Article En | MEDLINE | ID: mdl-38642107

Glioma is a systemic disease that can induce micro and macro alternations of whole brain. Isocitrate dehydrogenase and vascular endothelial growth factor are proven prognostic markers and antiangiogenic therapy targets in glioma. The aim of this study was to determine the ability of whole brain morphologic features and radiomics to predict isocitrate dehydrogenase status and vascular endothelial growth factor expression levels. This study recruited 80 glioma patients with isocitrate dehydrogenase wildtype and high vascular endothelial growth factor expression levels, and 102 patients with isocitrate dehydrogenase mutation and low vascular endothelial growth factor expression levels. Virtual brain grafting, combined with Freesurfer, was used to compute morphologic features including cortical thickness, LGI, and subcortical volume in glioma patient. Radiomics features were extracted from multiregional tumor. Pycaret was used to construct the machine learning pipeline. Among the radiomics models, the whole tumor model achieved the best performance (accuracy 0.80, Area Under the Curve 0.86), while, after incorporating whole brain morphologic features, the model had a superior predictive performance (accuracy 0.82, Area Under the Curve 0.88). The features contributed most in predicting model including the right caudate volume, left middle temporal cortical thickness, first-order statistics, shape, and gray-level cooccurrence matrix. Pycaret, based on morphologic features, combined with radiomics, yielded highest accuracy in predicting isocitrate dehydrogenase mutation and vascular endothelial growth factor levels, indicating that morphologic abnormalities induced by glioma were associated with tumor biology.


Brain Neoplasms , Glioma , Humans , Vascular Endothelial Growth Factor A/genetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Isocitrate Dehydrogenase/genetics , Magnetic Resonance Imaging , Glioma/diagnostic imaging , Glioma/genetics , Brain/diagnostic imaging , Brain/pathology , Mutation , Retrospective Studies
10.
Brain Tumor Pathol ; 41(2): 43-49, 2024 Apr.
Article En | MEDLINE | ID: mdl-38564040

Oligodendroglioma, IDH-mutant and 1p/19q-codeleted is known for their relative chemosensitivity and indolent clinical course among diffuse gliomas of adult type. Based on the data from phase 3 clinical trials, the standard of post-surgical care for those tumors is considered to be initial chemoradiotherapy regardless of histopathological grade, particularly with PCV. However, partly due to its renewed definition in late years, prognostic factors in patients with those tumors are not well established. Moreover, the survival rate declines over 15 years, with only a 37% OS rate at 20 years for grade 3 tumors, even with the current standard of care. Given that most of this disease occurs in young or middle-aged adults, further improvements in treatment and management are necessary. Here, we discuss prognostic factors, standard of care and chemotherapy, and future perspectives with neoadjuvant strategy in those tumors.


Brain Neoplasms , Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 1 , Isocitrate Dehydrogenase , Mutation , Neoadjuvant Therapy , Oligodendroglioma , Standard of Care , Humans , Oligodendroglioma/genetics , Oligodendroglioma/therapy , Oligodendroglioma/pathology , Isocitrate Dehydrogenase/genetics , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Prognosis , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 19/genetics , Adult , Chromosome Deletion , Survival Rate , Middle Aged
11.
Pathol Res Pract ; 257: 155272, 2024 May.
Article En | MEDLINE | ID: mdl-38631135

Glioblastoma, IDH-wild type, the most common malignant primary central nervous system tumor, represents a formidable challenge in clinical management due to its poor prognosis and limited therapeutic responses. With an evolving understanding of its underlying biology, there is an urgent need to identify prognostic molecular groups that can be subject to targeted therapy. This study established a cohort of 124 sequential glioblastomas from a tertiary hospital and aimed to find correlations between molecular features and survival outcomes. Comprehensive molecular characterization of the cohort revealed prevalent alterations as previously described, such as TERT promoter mutations and involvement of the PI3K-Akt-mTOR, CK4/6-CDKN2A/B-RB1, and p14ARF-MDM2-MDM4-p53 pathways. MGMT promoter methylation is a significant predictor of improved overall survival, aligned with previous data. Conversely, age showed a marginal association with higher mortality. Multivariate analysis to account for the effect of MGMT promoter methylation and age showed that, in contrast to other published series, this cohort demonstrated improved survival for tumors harboring PTEN mutations, and that there was no observed difference for most other molecular alterations, including EGFR amplification, RB1 loss, or the coexistence of EGFR amplification and deletion/exon skipping (EGFRvIII). Despite limitations in sample size, this study contributes data to the molecular landscape of glioblastomas, prompting further investigations to examine these findings more closely in larger cohorts.


Brain Neoplasms , Glioblastoma , Isocitrate Dehydrogenase , Humans , Glioblastoma/genetics , Glioblastoma/mortality , Glioblastoma/pathology , Middle Aged , Male , Female , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Aged , Adult , Isocitrate Dehydrogenase/genetics , Mutation , Cohort Studies , Prognosis , Biomarkers, Tumor/genetics , DNA Methylation/genetics , Young Adult , Aged, 80 and over , Promoter Regions, Genetic/genetics , Survival Analysis
12.
Cancer Cell ; 42(5): 904-914.e9, 2024 May 13.
Article En | MEDLINE | ID: mdl-38579724

A subset of patients with IDH-mutant glioma respond to inhibitors of mutant IDH (IDHi), yet the molecular underpinnings of such responses are not understood. Here, we profiled by single-cell or single-nucleus RNA-sequencing three IDH-mutant oligodendrogliomas from patients who derived clinical benefit from IDHi. Importantly, the tissues were sampled on-drug, four weeks from treatment initiation. We further integrate our findings with analysis of single-cell and bulk transcriptomes from independent cohorts and experimental models. We find that IDHi treatment induces a robust differentiation toward the astrocytic lineage, accompanied by a depletion of stem-like cells and a reduction of cell proliferation. Furthermore, mutations in NOTCH1 are associated with decreased astrocytic differentiation and may limit the response to IDHi. Our study highlights the differentiating potential of IDHi on the cellular hierarchies that drive oligodendrogliomas and suggests a genetic modifier that may improve patient stratification.


Brain Neoplasms , Cell Differentiation , Isocitrate Dehydrogenase , Mutation , Oligodendroglioma , Oligodendroglioma/genetics , Oligodendroglioma/pathology , Oligodendroglioma/drug therapy , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/antagonists & inhibitors , Humans , Cell Differentiation/drug effects , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/drug therapy , Cell Lineage/drug effects , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Cell Proliferation/drug effects , Animals , Astrocytes/metabolism , Astrocytes/drug effects , Astrocytes/pathology , Mice , Single-Cell Analysis/methods
13.
Cancer Cell ; 42(5): 741-743, 2024 May 13.
Article En | MEDLINE | ID: mdl-38579726

In this issue of Cancer Cell, Spitzer and colleagues demonstrate the role of IDH inhibitors on IDHmutant gliomas in reducing proliferation and enhancing cell differentiation toward an astrocytic-like state, thus altering neurodevelopmental pathways. Despite clinical promise, unresolved questions regarding mechanisms of action and resistance underline the need for further research for treatment optimization.


Brain Neoplasms , Glioma , Isocitrate Dehydrogenase , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/antagonists & inhibitors , Glioma/drug therapy , Glioma/pathology , Glioma/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Mutation , Astrocytes/drug effects , Astrocytes/metabolism
14.
Cancer Cell ; 42(5): 815-832.e12, 2024 May 13.
Article En | MEDLINE | ID: mdl-38640932

Monocyte-derived tumor-associated macrophages (Mo-TAMs) intensively infiltrate diffuse gliomas with remarkable heterogeneity. Using single-cell transcriptomics, we chart a spatially resolved transcriptional landscape of Mo-TAMs across 51 patients with isocitrate dehydrogenase (IDH)-wild-type glioblastomas or IDH-mutant gliomas. We characterize a Mo-TAM subset that is localized to the peri-necrotic niche and skewed by hypoxic niche cues to acquire a hypoxia response signature. Hypoxia-TAM destabilizes endothelial adherens junctions by activating adrenomedullin paracrine signaling, thereby stimulating a hyperpermeable neovasculature that hampers drug delivery in glioblastoma xenografts. Accordingly, genetic ablation or pharmacological blockade of adrenomedullin produced by Hypoxia-TAM restores vascular integrity, improves intratumoral concentration of the anti-tumor agent dabrafenib, and achieves combinatorial therapeutic benefits. Increased proportion of Hypoxia-TAM or adrenomedullin expression is predictive of tumor vessel hyperpermeability and a worse prognosis of glioblastoma. Our findings highlight Mo-TAM diversity and spatial niche-steered Mo-TAM reprogramming in diffuse gliomas and indicate potential therapeutics targeting Hypoxia-TAM to normalize tumor vasculature.


Adrenomedullin , Brain Neoplasms , Glioblastoma , Tumor-Associated Macrophages , Humans , Glioblastoma/pathology , Glioblastoma/drug therapy , Glioblastoma/blood supply , Glioblastoma/genetics , Glioblastoma/metabolism , Animals , Adrenomedullin/genetics , Adrenomedullin/metabolism , Mice , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Brain Neoplasms/blood supply , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Tumor-Associated Macrophages/metabolism , Neovascularization, Pathologic/genetics , Tumor Microenvironment , Isocitrate Dehydrogenase/genetics , Xenograft Model Antitumor Assays , Cell Line, Tumor , Macrophages/metabolism , Cell Hypoxia
15.
J Neurooncol ; 167(3): 489-499, 2024 May.
Article En | MEDLINE | ID: mdl-38653957

PURPOSE: Glioblastoma is a malignant brain tumor with a poor prognosis. Genetic mutations associated with this disease are complex are not fully understood and require further elucidation for the development of new treatments. The purpose of this study was to comprehensively analyze genetic mutations in glioblastomas and evaluate the usefulness of RNA sequencing. PATIENTS AND METHODS: We analyzed 42 glioblastoma specimens that were resected in routine clinical practice and found wild-type variants of the IDH1 and IDH2 genes. RNA was extracted from frozen specimens and sequenced, and genetic analyses were performed using the CLC Genomics Workbench. RESULTS: The most common genetic alterations in the 42 glioblastoma specimens were TP53 mutation (28.6%), EGFR splicing variant (16.7%), EGFR mutation (9.5%), and FGFR3 fusion (9.5%). Novel genetic mutations were detected in 8 patients (19%). In 12 cases (28.6%), driver gene mutations were not detected, suggesting an association with PPP1R14A overexpression. Our findings suggest the transcription factors SOX10 and NKX6-2 are potential markers in glioblastoma. CONCLUSION: RNA sequencing is a promising approach for genotyping glioblastomas because it provides comprehensive information on gene expression and is relatively cost-effective.


Brain Neoplasms , Glioblastoma , Isocitrate Dehydrogenase , Mutation , Humans , Glioblastoma/genetics , Isocitrate Dehydrogenase/genetics , Male , Female , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Middle Aged , Aged , Adult , Sequence Analysis, RNA/methods , Biomarkers, Tumor/genetics , Genomics/methods , Young Adult , Aged, 80 and over , Prognosis
16.
Acta Neurochir (Wien) ; 166(1): 196, 2024 Apr 27.
Article En | MEDLINE | ID: mdl-38676720

BACKGROUND: The prognostic value of the extent of resection in the management of Glioblastoma is a long-debated topic, recently widened by the 2022 RANO-Resect Classification, which advocates for the resection of the non-enhancing disease surrounding the main core of tumors (supramaximal resection, SUPR) to achieve additional survival benefits. We conducted a retrospective analysis to corroborate the role of SUPR by the RANO-Resect Classification in a single center, homogenous cohort of patients. METHODS: Records of patients operated for WHO-2021 Glioblastomas at our institution between 2007 and 2018 were retrospectively reviewed; volumetric data of resected lesions were computed and classified by RANO-Resect criteria. Survival and correlation analyses were conducted excluding patients below near-total resection. RESULTS: 117 patients met the inclusion criteria, encompassing 45 near-total resections (NTR), 31 complete resections (CR), and 41 SUPR. Median progression-free and overall survival were 11 and 15 months for NTR, 13 and 17 months or CR, 20 and 24 months for SUPR, respectively (p < 0.001), with inverse correlation observed between survival and FLAIR residual volume (r -0.28). SUPR was not significantly associated with larger preoperative volumes or higher rates of postoperative deficits, although it was less associated with preoperative neurological deficits (OR 3.37, p = 0.003). The impact of SUPR on OS varied between MGMT unmethylated (HR 0.606, p = 0.044) and methylated (HR 0.273, p = 0.002) patient groups. CONCLUSIONS: Results of the present study support the validity of supramaximal resection by the new RANO-Resect classification, also highlighting a possible surgical difference between tumors with methylated and unmethylated MGMT promoter.


Brain Neoplasms , Glioblastoma , Isocitrate Dehydrogenase , Humans , Glioblastoma/surgery , Glioblastoma/pathology , Glioblastoma/genetics , Glioblastoma/mortality , Retrospective Studies , Middle Aged , Male , Brain Neoplasms/surgery , Brain Neoplasms/pathology , Brain Neoplasms/mortality , Brain Neoplasms/diagnostic imaging , Female , Aged , Adult , Isocitrate Dehydrogenase/genetics , Neurosurgical Procedures/methods
17.
Zhonghua Bing Li Xue Za Zhi ; 53(5): 439-445, 2024 May 08.
Article Zh | MEDLINE | ID: mdl-38678323

Objective: To examine whether immunohistochemistry of methylthioadenosine phosphorylase (MTAP) and p16 could be used to predict the CDKN2A status in various brain tumors. Methods: A total of 118 cases of IDH-mutant astrocytomas, 16 IDH-wildtype glioblastoma, 17 polymorphic xanthoastrocytoma (PXA) and 20 meningiomas diagnosed at Xuanwu Hospital, Capital Medical University, Beijing, China from November 2017 to October 2023 were collected and analyzed. The CDKN2A status was detected by using fluorescence in situ hybridization or next-generation sequencing. Expression of MTAP and p16 proteins was detected with immunohistochemistry. The association of loss of MTAP/p16 expression with CDKN2A homozygous/heterozygous deletion was examined. Results: Among the 118 cases of IDH-mutant astrocytoma, 13 cases showed homozygous deletion of CDKN2A. All of them had no expression of MTAP while 9 cases had no expression of p16. Among the 16 cases of IDH wild-type glioblastoma, 6 cases showed homozygous deletion of CDKN2A. All 6 cases had no expression of MTAP, while 3 of these cases had no expression of p16 expression. Among the 17 PXA cases, 4 cases showed homozygous deletion of CDKN2A, and the expression of MTAP and p16 was also absent in these 4 cases. Among the 20 cases of meningiomas, 4 cases showed homozygous deletion of CDKN2A. Their expression of MTAP and p16 was also absent. Among the four types of brain tumors, MTAP was significantly correlated with CDKN2A homozygous deletion (P<0.05), with a sensitivity of 100%. However, it was only significantly correlated with the loss of heterozygosity (LOH) of CDKN2A in astrocytomas (P<0.001). P16 was associated with CDKN2A homozygous deletion in IDH-mutant astrocytoma and PXA (P<0.001), but not with the LOH of CDKN2A. Its sensitivity and specificity were lower than that of MTAP. Conclusions: MTAP could serve as a predictive surrogate for CDKN2A homozygous deletion in adult IDH-mutant astrocytoma, PXA, adult IDH-wildtype glioblastoma and meningioma. However, p16 could only be used in the first two tumor types, and its specificity and sensitivity are lower than that of MTAP.


Biomarkers, Tumor , Brain Neoplasms , Cyclin-Dependent Kinase Inhibitor p16 , Homozygote , Purine-Nucleoside Phosphorylase , Humans , Purine-Nucleoside Phosphorylase/genetics , Purine-Nucleoside Phosphorylase/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Biomarkers, Tumor/metabolism , Biomarkers, Tumor/genetics , Astrocytoma/genetics , Astrocytoma/metabolism , Meningioma/genetics , Meningioma/metabolism , Meningioma/pathology , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Immunohistochemistry , In Situ Hybridization, Fluorescence , Gene Deletion , Meningeal Neoplasms/genetics , Meningeal Neoplasms/metabolism , Mutation , Male , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Female , Adult , High-Throughput Nucleotide Sequencing
18.
Acta Neuropathol Commun ; 12(1): 57, 2024 Apr 11.
Article En | MEDLINE | ID: mdl-38605367

Li-Fraumeni syndrome (LFS) is an autosomal dominant tumor predisposition syndrome caused by heterozygous germline mutations or deletions in the TP53 tumor suppressor gene. Central nervous system tumors, such as choroid plexus tumors, medulloblastomas, and diffuse gliomas, are frequently found in patients with LFS. Although molecular profiles of diffuse gliomas that develop in pediatric patients with LFS have been elucidated, those in adults are limited. Recently, diffuse gliomas have been divided into pediatric- and adult-type gliomas, based on their distinct molecular profiles. In the present study, we investigated the molecular profiles of high-grade gliomas in three adults with LFS. These tumors revealed characteristic histopathological findings of high-grade glioma or glioblastoma and harbored wild-type IDH1/2 according to whole exome sequencing (WES). However, these tumors did not exhibit the key molecular alterations of glioblastoma, IDH-wildtype such as TERT promoter mutation, EGFR amplification, or chromosome 7 gain and 10 loss. Although WES revealed no other characteristic gene mutations or copy number alterations in high-grade gliomas, such as those in histone H3 genes, PDGFRA amplification was found in all three cases together with uniparental disomy of chromosome 17p, where the TP53 gene is located. DNA methylation analyses revealed that all tumors exhibited DNA methylation profiles similar to those of pediatric-type high-grade glioma H3-wildtype and IDH-wildtype (pHGG H3-/IDH-wt), RTK1 subtype. These data suggest that high-grade gliomas developed in adult patients with LFS may be involved in pHGG H3-/IDH-wt. PDGFRA and homozygous alterations in TP53 may play pivotal roles in the development of this type of glioma in adult patients with LFS.


Brain Neoplasms , Glioma , Li-Fraumeni Syndrome , Adult , Humans , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Genes, p53 , Glioblastoma/genetics , Glioblastoma/pathology , Glioma/genetics , Glioma/pathology , Isocitrate Dehydrogenase/genetics , Li-Fraumeni Syndrome/genetics , Mutation/genetics
19.
Nat Commun ; 15(1): 3445, 2024 Apr 24.
Article En | MEDLINE | ID: mdl-38658533

Mutations in isocitrate dehydrogenases (IDH) are oncogenic events due to the generation of oncogenic metabolite 2-hydroxyglutarate. However, the role of wild-type IDH in cancer development remains elusive. Here we show that wild-type IDH2 is highly expressed in triple negative breast cancer (TNBC) cells and promotes their proliferation in vitro and tumor growth in vivo. Genetic silencing or pharmacological inhibition of wt-IDH2 causes a significant increase in α-ketoglutarate (α-KG), indicating a suppression of reductive tricarboxylic acid (TCA) cycle. The aberrant accumulation of α-KG due to IDH2 abrogation inhibits mitochondrial ATP synthesis and promotes HIF-1α degradation, leading to suppression of glycolysis. Such metabolic double-hit results in ATP depletion and suppression of tumor growth, and renders TNBC cells more sensitive to doxorubicin treatment. Our study reveals a metabolic property of TNBC cells with active utilization of glutamine via reductive TCA metabolism, and suggests that wild-type IDH2 plays an important role in this metabolic process and could be a potential therapeutic target for TNBC.


Cell Proliferation , Citric Acid Cycle , Isocitrate Dehydrogenase , Ketoglutaric Acids , Triple Negative Breast Neoplasms , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Humans , Female , Animals , Cell Line, Tumor , Citric Acid Cycle/drug effects , Ketoglutaric Acids/metabolism , Mice , Cell Proliferation/drug effects , Glycolysis/drug effects , Adenosine Triphosphate/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mitochondria/metabolism , Mitochondria/drug effects , Glutamine/metabolism , Xenograft Model Antitumor Assays , Mutation
20.
NPJ Syst Biol Appl ; 10(1): 38, 2024 Apr 09.
Article En | MEDLINE | ID: mdl-38594351

Acute myeloid leukemia (AML) is characterized by uncontrolled proliferation of poorly differentiated myeloid cells, with a heterogenous mutational landscape. Mutations in IDH1 and IDH2 are found in 20% of the AML cases. Although much effort has been made to identify genes associated with leukemogenesis, the regulatory mechanism of AML state transition is still not fully understood. To alleviate this issue, here we develop a new computational approach that integrates genomic data from diverse sources, including gene expression and ATAC-seq datasets, curated gene regulatory interaction databases, and mathematical modeling to establish models of context-specific core gene regulatory networks (GRNs) for a mechanistic understanding of tumorigenesis of AML with IDH mutations. The approach adopts a new optimization procedure to identify the top network according to its accuracy in capturing gene expression states and its flexibility to allow sufficient control of state transitions. From GRN modeling, we identify key regulators associated with the function of IDH mutations, such as DNA methyltransferase DNMT1, and network destabilizers, such as E2F1. The constructed core regulatory network and outcomes of in-silico network perturbations are supported by survival data from AML patients. We expect that the combined bioinformatics and systems-biology modeling approach will be generally applicable to elucidate the gene regulation of disease progression.


Leukemia, Myeloid, Acute , Nucleophosmin , Humans , Gene Regulatory Networks/genetics , Isocitrate Dehydrogenase/genetics , Leukemia, Myeloid, Acute/genetics , Carcinogenesis
...