Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 152
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 185(12): 2184-2199.e16, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35649412

ABSTRACT

The factors driving therapy resistance in diffuse glioma remain poorly understood. To identify treatment-associated cellular and genetic changes, we analyzed RNA and/or DNA sequencing data from the temporally separated tumor pairs of 304 adult patients with isocitrate dehydrogenase (IDH)-wild-type and IDH-mutant glioma. Tumors recurred in distinct manners that were dependent on IDH mutation status and attributable to changes in histological feature composition, somatic alterations, and microenvironment interactions. Hypermutation and acquired CDKN2A deletions were associated with an increase in proliferating neoplastic cells at recurrence in both glioma subtypes, reflecting active tumor growth. IDH-wild-type tumors were more invasive at recurrence, and their neoplastic cells exhibited increased expression of neuronal signaling programs that reflected a possible role for neuronal interactions in promoting glioma progression. Mesenchymal transition was associated with the presence of a myeloid cell state defined by specific ligand-receptor interactions with neoplastic cells. Collectively, these recurrence-associated phenotypes represent potential targets to alter disease progression.


Subject(s)
Brain Neoplasms , Glioma , Tumor Microenvironment , Adult , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Evolution, Molecular , Genes, p16 , Glioma/genetics , Glioma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Mutation , Neoplasm Recurrence, Local
2.
Cell ; 156(5): 1002-16, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24581498

ABSTRACT

Brain metastasis is an ominous complication of cancer, yet most cancer cells that infiltrate the brain die of unknown causes. Here, we identify plasmin from the reactive brain stroma as a defense against metastatic invasion, and plasminogen activator (PA) inhibitory serpins in cancer cells as a shield against this defense. Plasmin suppresses brain metastasis in two ways: by converting membrane-bound astrocytic FasL into a paracrine death signal for cancer cells, and by inactivating the axon pathfinding molecule L1CAM, which metastatic cells express for spreading along brain capillaries and for metastatic outgrowth. Brain metastatic cells from lung cancer and breast cancer express high levels of anti-PA serpins, including neuroserpin and serpin B2, to prevent plasmin generation and its metastasis-suppressive effects. By protecting cancer cells from death signals and fostering vascular co-option, anti-PA serpins provide a unifying mechanism for the initiation of brain metastasis in lung and breast cancers.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Brain/metabolism , Fibrinolysin/metabolism , Neuropeptides/metabolism , Plasminogen Activator Inhibitor 2/metabolism , Serpins/metabolism , Adenocarcinoma/secondary , Animals , Astrocytes/metabolism , Brain/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma/secondary , Cell Line, Tumor , Cell Survival , Disease Models, Animal , Fas Ligand Protein/metabolism , Female , Humans , Lung Neoplasms/pathology , Mice , Mice, Nude , Neural Cell Adhesion Molecule L1/metabolism , Neuropeptides/genetics , Plasminogen Activator Inhibitor 2/genetics , Plasminogen Activators/metabolism , Serpins/genetics , Neuroserpin
3.
J Neurosci ; 43(47): 8043-8057, 2023 11 22.
Article in English | MEDLINE | ID: mdl-37722850

ABSTRACT

The malignant brain cancer glioblastoma (GBM) contains groups of highly invasive cells that drive tumor progression as well as recurrence after surgery and chemotherapy. The molecular mechanisms that enable these GBM cells to exit the primary mass and disperse throughout the brain remain largely unknown. Here we report using human tumor specimens and primary spheroids from male and female patients that glial cell adhesion molecule (GlialCAM), which has normal roles in brain astrocytes and is mutated in the developmental brain disorder megalencephalic leukoencephalopathy with subcortical cysts (MLC), is differentially expressed in subpopulations of GBM cells. High levels of GlialCAM promote cell-cell adhesion and a proliferative GBM cell state in the tumor core. In contrast, GBM cells with low levels of GlialCAM display diminished proliferation and enhanced invasion into the surrounding brain parenchyma. RNAi-mediated inhibition of GlialCAM expression leads to activation of proinvasive extracellular matrix adhesion and signaling pathways. Profiling GlialCAM-regulated genes combined with cross-referencing to single-cell transcriptomic datasets validates functional links among GlialCAM, Mlc1, and aquaporin-4 in the invasive cell state. Collectively, these results reveal an important adhesion and signaling axis comprised of GlialCAM and associated proteins including Mlc1 and aquaporin-4 that is critical for control of GBM cell proliferation and invasion status in the brain cancer microenvironment.SIGNIFICANCE STATEMENT Glioblastoma (GBM) contains heterogeneous populations of cells that coordinately drive proliferation and invasion. We have discovered that glial cell adhesion molecule (GlialCAM)/hepatocyte cell adhesion molecule (HepaCAM) is highly expressed in proliferative GBM cells within the tumor core. In contrast, GBM cells with low levels of GlialCAM robustly invade into surrounding brain tissue along blood vessels and white matter. Quantitative RNA sequencing identifies various GlialCAM-regulated genes with functions in cell-cell adhesion and signaling. These data reveal that GlialCAM and associated signaling partners, including Mlc1 and aquaporin-4, are key factors that determine proliferative and invasive cell states in GBM.


Subject(s)
Aquaporins , Glioblastoma , Female , Humans , Male , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Cycle Proteins/metabolism , Glioblastoma/metabolism , Glioblastoma/pathology , Membrane Proteins/metabolism , Tumor Microenvironment , Cell Proliferation , Neoplasm Invasiveness
4.
Neuropathology ; 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38414131

ABSTRACT

Herein, we report a case of a collision tumor involving a multinodular and vacuolating neuronal tumor (MVNT) and a diffuse astrocytoma. A collision tumor between these two entities has not previously been reported. The patient is a 35-year-old woman who presented with new-onset hearing loss and ringing in her right ear. Magnetic resonance imaging identified a non-enhancing mass involving the gray matter and subcortical white matter of the left middle frontal gyrus. Additionally, tiny clustered nodules were noted along the underlying subcortical ribbon and superficial subcortical white matter of the left superior frontal gyrus. The patient underwent a left frontal craniotomy and complete resection of the mass. Histologic examination of the resected specimen demonstrated a collision tumor consisting of a diffuse astrocytoma (isocitrate dehydrogenase [IDH] mutant, central nervous system [CNS] World Health Organization [WHO] grade 2) and an MVNT, with the latter demonstrating characteristic morphologic and immunohistochemical features.

5.
Lancet Oncol ; 24(11): e438-e450, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37922934

ABSTRACT

Surgical resection represents the standard of care for people with newly diagnosed diffuse gliomas, and the neuropathological and molecular profile of the resected tissue guides clinical management and forms the basis for research. The Response Assessment in Neuro-Oncology (RANO) consortium is an international, multidisciplinary effort that aims to standardise research practice in neuro-oncology. These recommendations represent a multidisciplinary consensus from the four RANO groups: RANO resect, RANO recurrent glioblastoma, RANO radiotherapy, and RANO/PET for a standardised workflow to achieve a representative tumour evaluation in a disease characterised by intratumoural heterogeneity, including recommendations on which tumour regions should be surgically sampled, how to define those regions on the basis of preoperative imaging, and the optimal sample volume. Practical recommendations for tissue sampling are given for people with low-grade and high-grade gliomas, as well as for people with newly diagnosed and recurrent disease. Sampling of liquid biopsies is also addressed. A standardised workflow for subsequent handling of the resected tissue is proposed to avoid information loss due to decreasing tissue quality or insufficient clinical information. The recommendations offer a framework for prospective biobanking studies.


Subject(s)
Brain Neoplasms , Glioma , Humans , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/surgery , Prospective Studies , Biological Specimen Banks , Neoplasm Recurrence, Local/surgery , Glioma/diagnostic imaging , Glioma/surgery
6.
Lab Invest ; 102(7): 682-690, 2022 07.
Article in English | MEDLINE | ID: mdl-35152274

ABSTRACT

Comprehensive molecular profiling has dramatically transformed the diagnostic neuropathology of brain tumors. Diffuse gliomas, the most common and deadly brain tumor variants, are now classified by highly recurrent biomarkers instead of histomorphological characteristics. Several of the key molecular alterations driving glioma classification involve epigenetic dysregulation at a fundamental level, implicating fields of biology not previously thought to play major roles glioma pathogenesis. This article will review the major epigenetic alterations underlying malignant gliomas, their likely mechanisms of action, and potential strategies for their therapeutic targeting.


Subject(s)
Brain Neoplasms , Glioma , Biomarkers , Biomarkers, Tumor , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Epigenesis, Genetic , Glioma/genetics , Glioma/pathology , Humans
7.
Acta Neuropathol ; 141(2): 303-321, 2021 02.
Article in English | MEDLINE | ID: mdl-33394124

ABSTRACT

The deadly complication of brain metastasis (BM) is largely confined to a relatively narrow cross-section of systemic malignancies, suggesting a fundamental role for biological mechanisms shared across commonly brain metastatic tumor types. To identify and characterize such mechanisms, we performed genomic, transcriptional, and proteomic profiling using whole-exome sequencing, mRNA-seq, and reverse-phase protein array analysis in a cohort of the lung, breast, and renal cell carcinomas consisting of BM and patient-matched primary or extracranial metastatic tissues. While no specific genomic alterations were associated with BM, correlations with impaired cellular immunity, upregulated oxidative phosphorylation (OXPHOS), and canonical oncogenic signaling pathways including phosphoinositide 3-kinase (PI3K) signaling, were apparent across multiple tumor histologies. Multiplexed immunofluorescence analysis confirmed significant T cell depletion in BM, indicative of a fundamentally altered immune microenvironment. Moreover, functional studies using in vitro and in vivo modeling demonstrated heightened oxidative metabolism in BM along with sensitivity to OXPHOS inhibition in murine BM models and brain metastatic derivatives relative to isogenic parentals. These findings demonstrate that pathophysiological rewiring of oncogenic signaling, cellular metabolism, and immune microenvironment broadly characterizes BM. Further clarification of this biology will likely reveal promising targets for therapeutic development against BM arising from a broad variety of systemic cancers.


Subject(s)
Brain Neoplasms/pathology , Brain Neoplasms/secondary , DNA Fingerprinting/methods , Genomics/methods , Animals , Base Sequence , Brain Neoplasms/immunology , Cell Survival , Female , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Protein Array Analysis , Proteomics , Superoxide Dismutase/metabolism , Survival Analysis , Exome Sequencing
8.
Curr Neurol Neurosci Rep ; 21(12): 67, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34817712

ABSTRACT

PURPOSE OF REVIEW: The upcoming 2021 World Health Organization (WHO) Classification of Tumours of the Central Nervous System will feature numerous changes in classification, diagnostic criteria, nomenclature, and grading of diffuse gliomas. This article reviews these changes and the clinical and molecular findings underlying them. RECENT FINDINGS: Since the publication of the 2016 World Health Organization (WHO) Classification of Tumours of the Central Nervous System, research has led to new insights into how molecular changes impact both the classification and grading of CNS tumors. The continued integration of molecular and histopathological features has led to changes in diagnostic criteria and grading for various tumors. In the new 2021 WHO CNS tumor classification scheme, diffuse gliomas will be classified as either adult-type diffuse gliomas, pediatric-type diffuse high-grade gliomas, or pediatric-type diffuse low-grade gliomas. The upcoming changes in the classification of adult-type and pediatric-type diffuse gliomas allow for more effective communication of both diagnostic and prognostic information, and-particularly in the case of pediatric-type diffuse gliomas-may suggest possible targeted strategies for therapeutic intervention.


Subject(s)
Brain Neoplasms , Central Nervous System Neoplasms , Glioma , Adult , Brain Neoplasms/genetics , Central Nervous System Neoplasms/genetics , Child , Glioma/genetics , Humans , Mutation , Prognosis , World Health Organization
9.
Int J Cancer ; 146(3): 739-748, 2020 02 01.
Article in English | MEDLINE | ID: mdl-30963577

ABSTRACT

Glioma incidence is highest in non-Hispanic Whites, and to date, glioma genome-wide association studies (GWAS) to date have only included European ancestry (EA) populations. African Americans and Hispanics in the US have varying proportions of EA, African (AA) and Native American ancestries (NAA). It is unknown if identified GWAS loci or increased EA is associated with increased glioma risk. We assessed whether EA was associated with glioma in African Americans and Hispanics. Data were obtained for 832 cases and 675 controls from the Glioma International Case-Control Study and GliomaSE Case-Control Study previously estimated to have <80% EA, or self-identify as non-White. We estimated global and local ancestry using fastStructure and RFMix, respectively, using 1,000 genomes project reference populations. Within groups with ≥40% AA (AFR≥0.4 ), and ≥15% NAA (AMR≥0.15 ), genome-wide association between local EA and glioma was evaluated using logistic regression conditioned on global EA for all gliomas. We identified two regions (7q21.11, p = 6.36 × 10-4 ; 11p11.12, p = 7.0 × 10-4 ) associated with increased EA, and one associated with decreased EA (20p12.13, p = 0.0026) in AFR≥0.4 . In addition, we identified a peak at rs1620291 (p = 4.36 × 10-6 ) in 7q21.3. Among AMR≥0.15 , we found an association between increased EA in one region (12q24.21, p = 8.38 × 10-4 ), and decreased EA in two regions (8q24.21, p = 0. 0010; 20q13.33, p = 6.36 × 10-4 ). No other significant associations were identified. This analysis identified an association between glioma and two regions previously identified in EA populations (8q24.21, 20q13.33) and four novel regions (7q21.11, 11p11.12, 12q24.21 and 20p12.13). The identifications of novel association with EA suggest regions to target for future genetic association studies.


Subject(s)
Black or African American/genetics , Genetic Predisposition to Disease/genetics , Glioma/etiology , Glioma/genetics , Case-Control Studies , Female , Genetic Association Studies/methods , Genetic Loci/genetics , Genome-Wide Association Study/methods , Genotype , Hispanic or Latino , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Risk , White People/genetics
10.
Eur J Nucl Med Mol Imaging ; 47(6): 1446-1457, 2020 06.
Article in English | MEDLINE | ID: mdl-31865407

ABSTRACT

PURPOSE: Radiographic changes of brain metastases after stereotactic radiosurgery (SRS) can signify tumor recurrence and/or radiation necrosis (RN); however, standard imaging modalities cannot easily distinguish between these two entities. We investigated whether 18F-Fluorocholine uptake in surgical samples of the resected lesions correlates with pathologic evidence of recurrent tumor and PET imaging. METHODS: About 14 patients previously treated with SRS that developed radiographic changes were included. All patients underwent a preoperative 40-min dynamic PET/CT concurrent with 392 ± 11 MBq bolus injection of 18F-Fluorocholine. 18F-Fluorocholine pharmacokinetics were evaluated by standardized uptake value (SUV), graphical analysis (Patlak plot; KiP) and an irreversible two-compartment model (K1, k2, k3, and Ki). 12 out of 14 patients were administered an additional 72 ± 14 MBq injection of 18F-Fluorocholine 95 ± 26 minutes prior to surgical resection. About 113 resected samples from 12 patients were blindly reviewed by a neuropathologist to assess the viable tumor and necrotic content, microvascular proliferation, reactive gliosis, and mono- and polymorphonuclear inflammatory infiltrates. Correlation between these metrics 18F-Fluorocholine SUV was investigated with a linear mixed model. Comparison of survival distributions of two groups of patients (population median split of PET SUVmax) was performed with the log-rank test. RESULTS: Exactly 10 out of 12 patients for which surgical samples were acquired exhibited pathologic recurrence. Strong correlation was observed between SUVmax as measured from a surgically removed sample with highest uptake and by PET (Pearson's r = 0.66). Patients with 18F-Fluorocholine PET SUVmax > 6 experienced poor survival. Surgical samples with viable tumor had higher 18F-fluorocholine uptake (SUV) than those without tumor (4.5 ± 3.7 and 2.6 ± 3.0; p = 0.01). 18F-fluorocholine count data from surgical samples is driven not only by the percentage viable tumor but also by the degree of inflammation and reactive gliosis (p ≤ 0.02; multivariate regression). CONCLUSIONS: 18F-Fluorocholine accumulation is increased in viable tumor; however, inflammation and gliosis may also lead to elevated uptake. Higher 18F-Fluorocholine PET uptake portends worse prognosis. Kinetic analysis of dynamic 18F-Fluorocholine PET imaging supports the adequacy of the simpler static SUV metric.


Subject(s)
Brain Neoplasms , Radiosurgery , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/surgery , Choline/analogs & derivatives , Humans , Kinetics , Neoplasm Recurrence, Local , Positron Emission Tomography Computed Tomography , Positron-Emission Tomography
11.
J Neurooncol ; 150(1): 5-15, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32399739

ABSTRACT

INTRODUCTION: Recently discovered molecular alterations in pediatric low-grade glioma have helped to refine the classification of these tumors and offered novel targets for therapy. Genetic aberrations may combine with histopathology to offer new insights into glioma classification, gliomagenesis and prognosis. Therapies targeting common genetic aberrations in the MAPK pathway offer a novel mechanism of tumor control that is currently under study. METHODS: We have reviewed common molecular alterations found in pediatric low-grade glioma as well as recent clinical trials of MEK and BRAF inhibitors. RESULTS: In this topic review, we examine the current understanding of molecular alterations in pediatric low-grade glioma, as well as their role in diagnosis, prognosis and therapy. We summarize current data on the efficacy of targeted therapies in pediatric low-grade gliomas, as well as the many unanswered questions that these new discoveries and therapies raise. CONCLUSIONS: The identification of driver alterations in pediatric low-grade glioma and the development of targeted therapies have opened new therapeutic avenues for patients with low-grade gliomas.


Subject(s)
Brain Neoplasms , Glioma , Biomarkers, Tumor/genetics , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Child , Glioma/drug therapy , Glioma/genetics , Humans , Molecular Targeted Therapy , Prognosis
12.
N Engl J Med ; 372(26): 2499-508, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26061753

ABSTRACT

BACKGROUND: The prediction of clinical behavior, response to therapy, and outcome of infiltrative glioma is challenging. On the basis of previous studies of tumor biology, we defined five glioma molecular groups with the use of three alterations: mutations in the TERT promoter, mutations in IDH, and codeletion of chromosome arms 1p and 19q (1p/19q codeletion). We tested the hypothesis that within groups based on these features, tumors would have similar clinical variables, acquired somatic alterations, and germline variants. METHODS: We scored tumors as negative or positive for each of these markers in 1087 gliomas and compared acquired alterations and patient characteristics among the five primary molecular groups. Using 11,590 controls, we assessed associations between these groups and known glioma germline variants. RESULTS: Among 615 grade II or III gliomas, 29% had all three alterations (i.e., were triple-positive), 5% had TERT and IDH mutations, 45% had only IDH mutations, 7% were triple-negative, and 10% had only TERT mutations; 5% had other combinations. Among 472 grade IV gliomas, less than 1% were triple-positive, 2% had TERT and IDH mutations, 7% had only IDH mutations, 17% were triple-negative, and 74% had only TERT mutations. The mean age at diagnosis was lowest (37 years) among patients who had gliomas with only IDH mutations and was highest (59 years) among patients who had gliomas with only TERT mutations. The molecular groups were independently associated with overall survival among patients with grade II or III gliomas but not among patients with grade IV gliomas. The molecular groups were associated with specific germline variants. CONCLUSIONS: Gliomas were classified into five principal groups on the basis of three tumor markers. The groups had different ages at onset, overall survival, and associations with germline variants, which implies that they are characterized by distinct mechanisms of pathogenesis. (Funded by the National Institutes of Health and others.).


Subject(s)
Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 1 , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Mutation , Telomerase/genetics , Adult , Age of Onset , Biomarkers, Tumor , DNA Mutational Analysis , DNA, Neoplasm/analysis , Female , Germ-Line Mutation , Glioma/classification , Glioma/mortality , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Grading , Promoter Regions, Genetic , Proportional Hazards Models
13.
N Engl J Med ; 372(26): 2481-98, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26061751

ABSTRACT

BACKGROUND: Diffuse low-grade and intermediate-grade gliomas (which together make up the lower-grade gliomas, World Health Organization grades II and III) have highly variable clinical behavior that is not adequately predicted on the basis of histologic class. Some are indolent; others quickly progress to glioblastoma. The uncertainty is compounded by interobserver variability in histologic diagnosis. Mutations in IDH, TP53, and ATRX and codeletion of chromosome arms 1p and 19q (1p/19q codeletion) have been implicated as clinically relevant markers of lower-grade gliomas. METHODS: We performed genomewide analyses of 293 lower-grade gliomas from adults, incorporating exome sequence, DNA copy number, DNA methylation, messenger RNA expression, microRNA expression, and targeted protein expression. These data were integrated and tested for correlation with clinical outcomes. RESULTS: Unsupervised clustering of mutations and data from RNA, DNA-copy-number, and DNA-methylation platforms uncovered concordant classification of three robust, nonoverlapping, prognostically significant subtypes of lower-grade glioma that were captured more accurately by IDH, 1p/19q, and TP53 status than by histologic class. Patients who had lower-grade gliomas with an IDH mutation and 1p/19q codeletion had the most favorable clinical outcomes. Their gliomas harbored mutations in CIC, FUBP1, NOTCH1, and the TERT promoter. Nearly all lower-grade gliomas with IDH mutations and no 1p/19q codeletion had mutations in TP53 (94%) and ATRX inactivation (86%). The large majority of lower-grade gliomas without an IDH mutation had genomic aberrations and clinical behavior strikingly similar to those found in primary glioblastoma. CONCLUSIONS: The integration of genomewide data from multiple platforms delineated three molecular classes of lower-grade gliomas that were more concordant with IDH, 1p/19q, and TP53 status than with histologic class. Lower-grade gliomas with an IDH mutation either had 1p/19q codeletion or carried a TP53 mutation. Most lower-grade gliomas without an IDH mutation were molecularly and clinically similar to glioblastoma. (Funded by the National Institutes of Health.).


Subject(s)
DNA, Neoplasm/analysis , Genes, p53 , Glioma/genetics , Mutation , Adolescent , Adult , Aged , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 19 , Cluster Analysis , Female , Glioblastoma/genetics , Glioma/metabolism , Glioma/mortality , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Grading , Proportional Hazards Models , Sequence Analysis, DNA , Signal Transduction
14.
Nature ; 483(7390): 479-83, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22343889

ABSTRACT

Both genome-wide genetic and epigenetic alterations are fundamentally important for the development of cancers, but the interdependence of these aberrations is poorly understood. Glioblastomas and other cancers with the CpG island methylator phenotype (CIMP) constitute a subset of tumours with extensive epigenomic aberrations and a distinct biology. Glioma CIMP (G-CIMP) is a powerful determinant of tumour pathogenicity, but the molecular basis of G-CIMP remains unresolved. Here we show that mutation of a single gene, isocitrate dehydrogenase 1 (IDH1), establishes G-CIMP by remodelling the methylome. This remodelling results in reorganization of the methylome and transcriptome. Examination of the epigenome of a large set of intermediate-grade gliomas demonstrates a distinct G-CIMP phenotype that is highly dependent on the presence of IDH mutation. Introduction of mutant IDH1 into primary human astrocytes alters specific histone marks, induces extensive DNA hypermethylation, and reshapes the methylome in a fashion that mirrors the changes observed in G-CIMP-positive lower-grade gliomas. Furthermore, the epigenomic alterations resulting from mutant IDH1 activate key gene expression programs, characterize G-CIMP-positive proneural glioblastomas but not other glioblastomas, and are predictive of improved survival. Our findings demonstrate that IDH mutation is the molecular basis of CIMP in gliomas, provide a framework for understanding oncogenesis in these gliomas, and highlight the interplay between genomic and epigenomic changes in human cancers.


Subject(s)
DNA Methylation/genetics , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Mutation/genetics , Phenotype , Astrocytes/cytology , Astrocytes/metabolism , Cell Survival/genetics , Cells, Cultured , CpG Islands/genetics , Epigenesis, Genetic , Epigenomics , Gene Expression Regulation , Glioblastoma/genetics , Glioblastoma/pathology , Glioma/pathology , HEK293 Cells , Histones/metabolism , Humans , Isocitrate Dehydrogenase/metabolism , Metabolome/genetics , Tumor Cells, Cultured
15.
Genes Dev ; 24(19): 2205-18, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20889717

ABSTRACT

Gene rearrangement in the form of an intragenic deletion is the primary mechanism of oncogenic mutation of the epidermal growth factor receptor (EGFR) gene in gliomas. However, the incidence of platelet-derived growth factor receptor-α (PDGFRA) gene rearrangement in these tumors is unknown. We investigated the PDGFRA locus in PDGFRA-amplified gliomas and identified two rearrangements, including the first case of a gene fusion between kinase insert domain receptor (KDR) (VEGFRII) and the PDGFRA gene, and six cases of PDGFRA(Δ8, 9), an intragenic deletion rearrangement. The PDGFRA(Δ8, 9) mutant was common, being present in 40% of the glioblastoma multiformes (GBMs) with PDGFRA amplification. Tumors with these two types of PDGFRA rearrangement displayed histologic features of oligodendroglioma, and the gene products of both rearrangements showed constitutively elevated tyrosine kinase activity and transforming potential that was reversed by PDGFR blockade. These results suggest the possibility that these PDGFRA mutants behave as oncogenes in this subset of gliomas, and that the prevalence of such rearrangements may have been considerably underestimated.


Subject(s)
Gene Rearrangement , Glioblastoma/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Amino Acid Sequence , Base Sequence , Benzamides , Gene Dosage , Gene Fusion/genetics , Glioblastoma/pathology , Humans , Imatinib Mesylate , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Mutation/genetics , Oligodendroglioma/genetics , Oligodendroglioma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Phthalazines/pharmacology , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction , Transformation, Genetic/drug effects
16.
Acta Neuropathol ; 133(3): 417-429, 2017 03.
Article in English | MEDLINE | ID: mdl-27812792

ABSTRACT

Epileptogenic tumors affecting children and young adults are a morphologically diverse collection of neuroepithelial neoplasms that, as a group, exhibit varying levels of glial and/or neuronal differentiation. Recent advances in molecular profiling technology, including comprehensive DNA sequencing and methylation analysis, have enabled the application of more precise and biologically relevant classification schemes to these tumors. In this report, we describe a morphologically and molecularly distinct epileptogenic neoplasm, the polymorphous low-grade neuroepithelial tumor of the young (PLNTY), which likely accounts for a sizable portion of oligodendroglioma-like tumors affecting the pediatric population. Characteristic microscopic findings most notably include infiltrative growth, the invariable presence of oligodendroglioma-like cellular components, and intense immunolabeling for cluster of differentiation 34 (CD34). Moreover, integrative molecular profiling reveals a distinct DNA methylation signature for PLNTYs, along with frequent genetic abnormalities involving either B-Raf proto-oncogene (BRAF) or fibroblast growth factor receptors 2 and 3 (FGFR2, FGFR3). These findings suggest that PLNTY represents a distinct biological entity within the larger spectrum of pediatric, low-grade neuroepithelial tumors.


Subject(s)
Antigens, CD34/metabolism , Brain Neoplasms/complications , Brain Neoplasms/genetics , Epilepsy/etiology , Gene Expression Regulation, Neoplastic/genetics , Mutation , Neoplasms, Neuroepithelial/complications , Signal Transduction/physiology , Adolescent , Adult , Antigens, CD34/genetics , Brain Neoplasms/diagnostic imaging , Child , Child, Preschool , Epilepsy/genetics , Female , Glial Fibrillary Acidic Protein/metabolism , Humans , Male , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Neoplasms, Neuroepithelial/diagnostic imaging , Neoplasms, Neuroepithelial/genetics , Neuroglia/pathology , Oligodendroglioma/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins B-raf/genetics , Receptors, Fibroblast Growth Factor/genetics , Young Adult
17.
Pediatr Blood Cancer ; 64(7)2017 Jul.
Article in English | MEDLINE | ID: mdl-28035748

ABSTRACT

BACKGROUND: The PI3K/AKT/mTOR pathway is aberrantly activated in many pediatric solid tumors including gliomas and medulloblastomas. Preclinical data in a pediatric glioma model demonstrated that the combination of perifosine (AKT inhibitor) and temsirolimus (mTOR inhibitor) is more potent at inhibiting the axis than either agent alone. We conducted this study to assess pharmacokinetics and identify the maximum tolerated dose for the combination. PROCEDURE: We performed a standard 3+3 phase I, open-label, dose-escalation study in patients with recurrent/refractory pediatric solid tumors. Four dose levels of perifosine (25-75 mg/m2 /day) and temsirolimus (25-75 mg/m2 IV weekly) were investigated. RESULTS: Twenty-three patients (median age 8.5 years) with brain tumors (diffuse intrinsic pontine glioma [DIPG] n = 8, high-grade glioma n = 6, medulloblastoma n = 2, ependymoma n = 1), neuroblastoma (n = 4), or rhabdomyosarcoma (n = 2) were treated. The combination was generally well tolerated and no dose-limiting toxicity was encountered. The most common grade 3 or 4 toxicities (at least possibly related) were thrombocytopenia (38.1%), neutropenia (23.8%), lymphopenia (23.8%), and hypercholesterolemia (19.0%). Pharmacokinetic findings for temsirolimus were similar to those observed in the temsirolimus single-agent phase II pediatric study and pharmacokinetic findings for perifosine were similar to those in adults. Stable disease was seen in 9 of 11 subjects with DIPG or high-grade glioma; no partial or complete responses were achieved. CONCLUSIONS: The combination of these AKT and mTOR inhibitors was safe and feasible in patients with recurrent/refractory pediatric solid tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasm Recurrence, Local/drug therapy , Neoplasms/drug therapy , Phosphorylcholine/analogs & derivatives , Sirolimus/analogs & derivatives , Adolescent , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Humans , Male , Maximum Tolerated Dose , Phosphorylcholine/administration & dosage , Phosphorylcholine/adverse effects , Phosphorylcholine/pharmacokinetics , Sirolimus/administration & dosage , Sirolimus/adverse effects , Sirolimus/pharmacokinetics , Young Adult
18.
Proc Natl Acad Sci U S A ; 111(22): 8149-54, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24843164

ABSTRACT

PTPRD, which encodes the protein tyrosine phosphatase receptor-δ, is one of the most frequently inactivated genes across human cancers, including glioblastoma multiforme (GBM). PTPRD undergoes both deletion and mutation in cancers, with copy number loss comprising the primary mode of inactivation in GBM. However, it is unknown whether loss of PTPRD promotes tumorigenesis in vivo, and the mechanistic basis of PTPRD function in tumors is unclear. Here, using genomic analysis and a glioma mouse model, we demonstrate that loss of Ptprd accelerates tumor formation and define the oncogenic context in which Ptprd loss acts. Specifically, we show that in human GBMs, heterozygous loss of PTPRD is the predominant type of lesion and that loss of PTPRD and the CDKN2A/p16(INK4A) tumor suppressor frequently co-occur. Accordingly, heterozygous loss of Ptprd cooperates with p16 deletion to drive gliomagenesis in mice. Moreover, loss of the Ptprd phosphatase resulted in phospho-Stat3 accumulation and constitutive activation of Stat3-driven genetic programs. Surprisingly, the consequences of Ptprd loss are maximal in the heterozygous state, demonstrating a tight dependence on gene dosage. Ptprd loss did not increase cell proliferation but rather altered pathways governing the macrophage response. In total, we reveal that PTPRD is a bona fide tumor suppressor, pinpoint PTPRD loss as a cause of aberrant STAT3 activation in gliomas, and establish PTPRD loss, in the setting of CDKN2A/p16(INK4A) deletion, as a driver of glioma progression.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , STAT3 Transcription Factor/metabolism , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/metabolism , Cell Proliferation , Chickens , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Models, Animal , Gene Expression Regulation, Neoplastic/physiology , Genes, Tumor Suppressor/physiology , Glioblastoma/immunology , Glioblastoma/pathology , Heterozygote , Humans , Mice , Mice, Knockout , Neoplasm Transplantation , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Tumor Microenvironment/immunology
19.
Genes Dev ; 23(11): 1327-37, 2009 Jun 01.
Article in English | MEDLINE | ID: mdl-19487573

ABSTRACT

Activated oncogenic signaling is central to the development of nearly all forms of cancer, including the most common class of primary brain tumor, glioma. Research over the last two decades has revealed the particular importance of the Akt pathway, and its molecular antagonist PTEN (phosphatase and tensin homolog), in the process of gliomagenesis. Recent studies have also demonstrated that microRNAs (miRNAs) may be responsible for the modulation of cancer-implicated genes in tumors. Here we report the identification miR-26a as a direct regulator of PTEN expression. We also show that miR-26a is frequently amplified at the DNA level in human glioma, most often in association with monoallelic PTEN loss. Finally, we demonstrate that miR-26a-mediated PTEN repression in a murine glioma model both enhances de novo tumor formation and precludes loss of heterozygosity and the PTEN locus. Our results document a new epigenetic mechanism for PTEN regulation in glioma and further highlight dysregulation of Akt signaling as crucial to the development of these tumors.


Subject(s)
Gene Expression Regulation, Neoplastic , Glioma/physiopathology , PTEN Phosphohydrolase/metabolism , Animals , Cells, Cultured , DNA Helicases/metabolism , Disease Models, Animal , Kaplan-Meier Estimate , Loss of Heterozygosity , Mice , MicroRNAs/metabolism , NIH 3T3 Cells , PTEN Phosphohydrolase/genetics
20.
Annu Rev Med ; 64: 59-70, 2013.
Article in English | MEDLINE | ID: mdl-23043492

ABSTRACT

Glioblastoma, the most common malignant primary brain tumor, carries an invariably poor prognosis. Targeting underlying biological foundations of the disease will be crucial to developing more effective treatment strategies. Although increasing evidence clearly indicates that glioblastoma is a molecularly heterogeneous disorder, recent large-scale expression profiling has provided a framework for categorizing the tumor into 3 to 4 distinct subclasses, each with its own characteristic genomic alterations. As such, there remains the enticing possibility that glioblastoma subclasses themselves might represent predictive biomarkers, particularly in the context of specific targeted agents. This review focuses on how best to ascertain the functional relevance of molecular subclass in glioblastoma through both preclinical and clinical investigations. The availability of appropriate mouse modeling systems along with expanded molecular profiling capabilities in the clinical setting should aid such efforts. However, significant systematic challenges remain, particularly in the setting of clinical trials.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/analysis , Brain Neoplasms , Genetic Therapy/methods , Genomics/methods , Glioblastoma , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL