Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 46
Filter
1.
Cancer Discov ; 11(6): 1454-1467, 2021 06.
Article in English | MEDLINE | ID: mdl-33563663

ABSTRACT

The RAS/MAPK pathway is an emerging targeted pathway across a spectrum of both adult and pediatric cancers. Typically, this is associated with a single, well-characterized point mutation in an oncogene. Hypermutant tumors that harbor many somatic mutations may obscure the interpretation of such targetable genomic events. We find that replication repair-deficient (RRD) cancers, which are universally hypermutant and affect children born with RRD cancer predisposition, are enriched for RAS/MAPK mutations (P = 10-8). These mutations are not random, exist in subclones, and increase in allelic frequency over time. The RAS/MAPK pathway is activated both transcriptionally and at the protein level in patient-derived RRD tumors, and these tumors responded to MEK inhibition in vitro and in vivo. Treatment of patients with RAS/MAPK hypermutant gliomas reveals durable responses to MEK inhibition. Our observations suggest that hypermutant tumors may be addicted to oncogenic pathways, resulting in favorable response to targeted therapies. SIGNIFICANCE: Tumors harboring a single RAS/MAPK driver mutation are targeted individually for therapeutic purposes. We find that in RRD hypermutant cancers, mutations in the RAS/MAPK pathway are enriched, highly expressed, and result in sensitivity to MEK inhibitors. Targeting an oncogenic pathway may provide therapeutic options for these hypermutant polyclonal cancers.This article is highlighted in the In This Issue feature, p. 1307.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Colorectal Neoplasms/drug therapy , Genetic Predisposition to Disease , Glioma/drug therapy , Mitogen-Activated Protein Kinase Kinases/genetics , Protein Kinase Inhibitors/therapeutic use , Adult , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Child , Colorectal Neoplasms/genetics , Female , Glioma/genetics , Global Health , Humans , Male , Mice , Mice, Inbred NOD , Mutation
2.
Neurooncol Adv ; 3(1): vdab002, 2021.
Article in English | MEDLINE | ID: mdl-33629064

ABSTRACT

BACKGROUND: Medulloblastoma (MB) comprises four subtypes of which group 3 MB are the most aggressive. Although overall survival for MB has improved, the outcome of group 3 MB remains dismal. C-MYC (MYC) amplification or MYC overexpression which characterizes group 3 MB is a strong negative prognostic factor and is frequently associated with metastases and relapses. We previously reported that MYC expression alone promotes highly aggressive MB phenotypes, in part via repression of thrombospondin-1 (TSP-1), a potent tumor suppressor. METHODS: In this study, we examined the potential role of TSP-1 and TSP-1 peptidomimetic ABT-898 in MYC-amplified human MB cell lines and two distinct murine models of MYC-driven group 3 MBs. RESULTS: We found that TSP-1 reconstitution diminished metastases and prolonged survival in orthotopic xenografts and promoted chemo- and radio-sensitivity via AKT signaling. Furthermore, we demonstrate that ABT-898 can recapitulate the effects of TSP-1 expression in MB cells in vitro and specifically induced apoptosis in murine group 3 MB tumor cells. CONCLUSION: Our data underscore the importance of TSP-1 as a critical tumor suppressor in MB and highlight TSP-1 peptidomimetics as promising novel therapeutics for the most lethal subtype of MB.

3.
Cancer Discov ; 11(5): 1176-1191, 2021 05.
Article in English | MEDLINE | ID: mdl-33355208

ABSTRACT

Although replication repair deficiency, either by mismatch repair deficiency (MMRD) and/or loss of DNA polymerase proofreading, can cause hypermutation in cancer, microsatellite instability (MSI) is considered a hallmark of MMRD alone. By genome-wide analysis of tumors with germline and somatic deficiencies in replication repair, we reveal a novel association between loss of polymerase proofreading and MSI, especially when both components are lost. Analysis of indels in microsatellites (MS-indels) identified five distinct signatures (MS-sigs). MMRD MS-sigs are dominated by multibase losses, whereas mutant-polymerase MS-sigs contain primarily single-base gains. MS deletions in MMRD tumors depend on the original size of the MS and converge to a preferred length, providing mechanistic insight. Finally, we demonstrate that MS-sigs can be a powerful clinical tool for managing individuals with germline MMRD and replication repair-deficient cancers, as they can detect the replication repair deficiency in normal cells and predict their response to immunotherapy. SIGNIFICANCE: Exome- and genome-wide MSI analysis reveals novel signatures that are uniquely attributed to mismatch repair and DNA polymerase. This provides new mechanistic insight into MS maintenance and can be applied clinically for diagnosis of replication repair deficiency and immunotherapy response prediction.This article is highlighted in the In This Issue feature, p. 995.


Subject(s)
Cell Transformation, Neoplastic , DNA Mismatch Repair , DNA-Directed DNA Polymerase , Gene Expression Regulation, Neoplastic , Microsatellite Instability , Neoplasms/genetics , Humans , Exome Sequencing
4.
Cancer Res ; 80(24): 5606-5618, 2020 12 15.
Article in English | MEDLINE | ID: mdl-32938641

ABSTRACT

POLE mutations are a major cause of hypermutant cancers, yet questions remain regarding mechanisms of tumorigenesis, genotype-phenotype correlation, and therapeutic considerations. In this study, we establish mouse models harboring cancer-associated POLE mutations P286R and S459F, which cause rapid albeit distinct time to cancer initiation in vivo, independent of their exonuclease activity. Mouse and human correlates enabled novel stratification of POLE mutations into three groups based on clinical phenotype and mutagenicity. Cancers driven by these mutations displayed striking resemblance to the human ultrahypermutation and specific signatures. Furthermore, Pole-driven cancers exhibited a continuous and stochastic mutagenesis mechanism, resulting in intertumoral and intratumoral heterogeneity. Checkpoint blockade did not prevent Pole lymphomas, but rather likely promoted lymphomagenesis as observed in humans. These observations provide insights into the carcinogenesis of POLE-driven tumors and valuable information for genetic counseling, surveillance, and immunotherapy for patients. SIGNIFICANCE: Two mouse models of polymerase exonuclease deficiency shed light on mechanisms of mutation accumulation and considerations for immunotherapy.See related commentary by Wisdom and Kirsch p. 5459.


Subject(s)
DNA Polymerase II , Neoplasms , Animals , DNA Polymerase II/genetics , Humans , Immune Checkpoint Inhibitors , Mice , Mutation , Neoplasms/genetics , Poly-ADP-Ribose Binding Proteins/genetics
5.
J Neuropathol Exp Neurol ; 79(4): 437-447, 2020 04 01.
Article in English | MEDLINE | ID: mdl-32053195

ABSTRACT

The diagnosis of medulloblastoma incorporates the histologic and molecular subclassification of clinical medulloblastoma samples into wingless (WNT)-activated, sonic hedgehog (SHH)-activated, group 3 and group 4 subgroups. Accurate medulloblastoma subclassification has important prognostic and treatment implications. Immunohistochemistry (IHC)-based and nanoString-based subgrouping methodologies have been independently described as options for medulloblastoma subgrouping, however have not previously been directly compared. We describe our experience with nanoString-based subgrouping in a clinical setting and compare this with our IHC-based results. Study materials included FFPE tissue from 160 medulloblastomas. Clinical data and tumor histology were reviewed. Immunohistochemical-based subgrouping using ß-catenin, filamin A and p53 antibodies and nanoString-based gene expression profiling were performed. The sensitivity and specificity of IHC-based subgrouping of WNT and SHH-activated medulloblastomas was 91.5% and 99.54%, respectively. Filamin A immunopositivity highly correlated with SHH/WNT-activated subgroups (sensitivity 100%, specificity 92.7%, p < 0.001). Nuclear ß-catenin immunopositivity had a sensitivity of 76.2% and specificity of 99.23% for detection of WNT-activated tumors. Approximately 23.8% of WNT cases would have been missed using an IHC-based subgrouping method alone. nanoString could confidently predict medulloblastoma subgroup in 93% of cases and could distinguish group 3/4 subgroups in 96.3% of cases. nanoString-based subgrouping allows for a more prognostically useful classification of clinical medulloblastoma samples.


Subject(s)
Cerebellar Neoplasms/diagnosis , Gene Expression Profiling/methods , Hedgehog Proteins/genetics , Immunohistochemistry , Medulloblastoma/diagnosis , Wnt Proteins/genetics , Adolescent , Adult , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Child , Child, Preschool , Female , Gene Expression Regulation, Neoplastic , Humans , Infant , Kaplan-Meier Estimate , Male , Medulloblastoma/genetics , Medulloblastoma/pathology , Middle Aged , Sensitivity and Specificity , Young Adult
6.
Cancer Cell ; 36(1): 51-67.e7, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31287992

ABSTRACT

Embryonal tumors with multilayered rosettes (ETMRs) are highly lethal infant brain cancers with characteristic amplification of Chr19q13.41 miRNA cluster (C19MC) and enrichment of pluripotency factor LIN28A. Here we investigated C19MC oncogenic mechanisms and discovered a C19MC-LIN28A-MYCN circuit fueled by multiple complex regulatory loops including an MYCN core transcriptional network and super-enhancers resulting from long-range MYCN DNA interactions and C19MC gene fusions. Our data show that this powerful oncogenic circuit, which entraps an early neural lineage network, is potently abrogated by bromodomain inhibitor JQ1, leading to ETMR cell death.


Subject(s)
Brain Neoplasms/etiology , Chromosomes, Human, Pair 19 , MicroRNAs/genetics , Multigene Family , N-Myc Proto-Oncogene Protein/genetics , Neoplasms, Germ Cell and Embryonal/etiology , RNA-Binding Proteins/genetics , Biomarkers, Tumor , Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Cell Cycle/genetics , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 2 , DNA Copy Number Variations , Enhancer Elements, Genetic , Epigenesis, Genetic , Gene Expression Regulation , Gene Regulatory Networks , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Models, Biological , Neoplasms, Germ Cell and Embryonal/diagnosis , Neoplasms, Germ Cell and Embryonal/therapy , Oncogenes
8.
J Clin Invest ; 129(1): 223-229, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30358567

ABSTRACT

Replicative immortality is a hallmark of cancer cells governed by telomere maintenance. Approximately 90% of human cancers maintain their telomeres by activating telomerase, driven by the transcriptional upregulation of telomerase reverse transcriptase (TERT). Although TERT promoter mutations (TPMs) are a major cancer-associated genetic mechanism of TERT upregulation, many cancers exhibit TERT upregulation without TPMs. In this study, we describe the TERT hypermethylated oncological region (THOR), a 433-bp genomic region encompassing 52 CpG sites located immediately upstream of the TERT core promoter, as a cancer-associated epigenetic mechanism of TERT upregulation. Unmethylated THOR repressed TERT promoter activity regardless of TPM status, and hypermethylation of THOR counteracted this repressive function. THOR methylation analysis in 1,352 human tumors revealed frequent (>45%) cancer-associated DNA hypermethylation in 9 of 11 (82%) tumor types screened. Additionally, THOR hypermethylation, either independently or along with TPMs, accounted for how approximately 90% of human cancers can aberrantly activate telomerase. Thus, we propose that THOR hypermethylation is a prevalent telomerase-activating mechanism in cancer that can act independently of or in conjunction with TPMs, further supporting the utility of THOR hypermethylation as a prognostic biomarker.


Subject(s)
DNA Methylation , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/biosynthesis , Promoter Regions, Genetic , Telomerase/biosynthesis , Cell Line, Tumor , CpG Islands , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Humans , Neoplasm Proteins/genetics , Telomerase/genetics
10.
PLoS One ; 13(4): e0193565, 2018.
Article in English | MEDLINE | ID: mdl-29621254

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is a universally fatal childhood cancer of the brain. Despite the introduction of conventional chemotherapy and radiotherapy, improvements in survival have been marginal and long-term survivorship is uncommon. Thus, new targets for therapeutics are critically needed. Early phase clinical trials exploring molecularly-targeted therapies against the epidermal growth factor receptor (EGFR) and novel immunotherapies targeting interleukin receptor-13α2 (IL-13Rα2) have demonstrated activity in this disease. To identify additional therapeutic markers for cell surface receptors, we performed exome sequencing (16 new samples, 22 previously published samples, total 38 with 26 matched normal DNA samples), RNA deep sequencing (17 new samples, 11 previously published samples, total 28 with 18 matched normal RNA samples), and immunohistochemistry (17 DIPG tissue samples) to examine the expression of the interleukin-4 (IL-4) signaling axis components (IL-4, interleukin 13 (IL-13), and their respective receptors IL-4Rα, IL-13Rα1, and IL-13Rα2). In addition, we correlated cytokine and receptor expression with expression of the oncogenes EGFR and c-MET. In DIPG tissues, transcript-level analysis found significant expression of IL-4, IL-13, and IL-13Rα1/2, with strong differential expression of IL-13Rα1/2 in tumor versus normal brain. At the protein level, immunohistochemical studies revealed high content of IL-4 and IL-13Rα1/2 but notably low expression of IL-13. Additionally, a strong positive correlation was observed between c-Met and IL-4Rα. The genomic and transcriptional landscape across all samples was also summarized. These data create a foundation for the design of potential new immunotherapies targeting IL-13 cell surface receptors in DIPG.


Subject(s)
Brain Stem Neoplasms/drug therapy , Glioma/drug therapy , Receptors, Interleukin-13/drug effects , Brain Stem Neoplasms/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Glioma/genetics , Humans , Interleukin-4/metabolism , Point Mutation , Receptors, Interleukin-13/genetics , Receptors, Interleukin-4/genetics , Receptors, Interleukin-4/metabolism , Sequence Analysis, DNA
11.
Cell ; 172(5): 1050-1062.e14, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29474906

ABSTRACT

While the preponderance of morbidity and mortality in medulloblastoma patients are due to metastatic disease, most research focuses on the primary tumor due to a dearth of metastatic tissue samples and model systems. Medulloblastoma metastases are found almost exclusively on the leptomeningeal surface of the brain and spinal cord; dissemination is therefore thought to occur through shedding of primary tumor cells into the cerebrospinal fluid followed by distal re-implantation on the leptomeninges. We present evidence for medulloblastoma circulating tumor cells (CTCs) in therapy-naive patients and demonstrate in vivo, through flank xenografting and parabiosis, that medulloblastoma CTCs can spread through the blood to the leptomeningeal space to form leptomeningeal metastases. Medulloblastoma leptomeningeal metastases express high levels of the chemokine CCL2, and expression of CCL2 in medulloblastoma in vivo is sufficient to drive leptomeningeal dissemination. Hematogenous dissemination of medulloblastoma offers a new opportunity to diagnose and treat lethal disseminated medulloblastoma.


Subject(s)
Medulloblastoma/blood supply , Medulloblastoma/pathology , Meningeal Neoplasms/blood supply , Meningeal Neoplasms/secondary , Allografts , Animals , Cell Line, Tumor , Chemokine CCL2/metabolism , Chromosomes, Human, Pair 10/genetics , Female , Humans , Male , Medulloblastoma/genetics , Mice, SCID , Neoplastic Cells, Circulating , Parabiosis
12.
J Clin Oncol ; 36(1): 97, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29140773
13.
J Neuropathol Exp Neurol ; 76(7): 562-570, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28863456

ABSTRACT

Previous studies identified recurrent fusion and duplication events in pediatric low-grade glioma (pLGG). In addition to their role in diagnosis, the presence of these events aid in dictating therapy and predicting patient survival. Clinically, BRAF alterations are most commonly identified using fluorescent in situ hybridization (FISH). However, this method is costly, labor-intensive and does not identify nonBRAF events. Here, we evaluated the NanoString nCounter gene expression system for detecting 32 of the most commonly reported fusion/duplication events in pLGG. The assay was validated on 90 pLGG samples using FISH as the gold standard and showed sensitivity and specificity of 97% and 98%, respectively. We next profiled formalin-fixed paraffin-embedded preserved biopsy specimens from 429 pLGG cases. 171 (40%) of the cases within our cohort tested positive for a fusion or duplication event contained within our panel. These events, in order of prevalence, were KIAA1549-BRAF 16;9 (89/171, 52.0%), KIAA1549-BRAF 15;9 (42/171, 24.6%), KIAA1549-BRAF 16;11 (14/171, 8.2%), FGFR1-TACC1 17;7 (13/171, 7.6%), MYBL1 duplication (5/171, 2.9%), KIAA1549-BRAF 18;10 (4/171, 2.3%), KIAA1549-BRAF 15;11 (2/171, 1.2%), FAM131B-BRAF 2;9 (1/171, 0.6%), and RNF130-BRAF 3;9 (1/171, 0.6%). This work introduces NanoString as a viable clinical replacement for the detection of fusion and duplication events in pLGG.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins B-raf/genetics , Biomarkers, Tumor/genetics , Biopsy , Brain Neoplasms/diagnosis , Cohort Studies , Comparative Genomic Hybridization , Female , Glioma/diagnosis , Humans , In Situ Hybridization, Fluorescence , Male , Mutation/genetics , Pediatrics , Receptor, Fibroblast Growth Factor, Type 1/genetics
14.
Cancer Res ; 77(3): 696-706, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27899382

ABSTRACT

Metastatic relapse is the major cause of death in pediatric neuroblastoma, where there remains a lack of therapies to target this stage of disease. To understand the molecular mechanisms mediating neuroblastoma metastasis, we developed a mouse model using intracardiac injection and in vivo selection to isolate malignant cell subpopulations with a higher propensity for metastasis to bone and the central nervous system. Gene expression profiling revealed primary and metastatic cells as two distinct cell populations defined by differential expression of 412 genes and of multiple pathways, including CADM1, SPHK1, and YAP/TAZ, whose expression independently predicted survival. In the metastatic subpopulations, a gene signature was defined (MET-75) that predicted survival of neuroblastoma patients with metastatic disease. Mechanistic investigations demonstrated causal roles for CADM1, SPHK1, and YAP/TAZ in mediating metastatic phenotypes in vitro and in vivo Notably, pharmacologic targeting of SPHK1 or YAP/TAZ was sufficient to inhibit neuroblastoma metastasis in vivo Overall, we identify gene expression signatures and candidate therapeutics that could improve the treatment of metastatic neuroblastoma. Cancer Res; 77(3); 696-706. ©2017 AACR.


Subject(s)
Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neuroblastoma/genetics , Neuroblastoma/pathology , Transcriptome , Animals , Cell Line, Tumor , Disease Models, Animal , Gene Expression Profiling , Heterografts , Immunoblotting , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred NOD , Mice, SCID , Oligonucleotide Array Sequence Analysis , X-Ray Microtomography
15.
J Clin Oncol ; 34(29): 3537-3543, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27573663

ABSTRACT

Purpose Vinblastine monotherapy has shown promising activity and a low-toxicity profile in patients with pediatric low-grade glioma (PLGG) who experienced treatment failure after initial treatment with chemotherapy and/or radiation. The aim of this study was to assess the activity of vinblastine in therapy-naïve children. Patients and Methods Patients < 18 years old with unresectable and/or progressive therapy-naïve PLGG were eligible. Vinblastine was administered once per week at a dose of 6 mg/m2 intravenously over a period of 70 weeks. Vision, quality of life, neurofibromatosis type 1 (NF1) status, and BRAF mutation/fusion status were also determined and correlated with outcome. Results Fifty-four patients were enrolled onto the study, with a median age of 8 years (range, 0.7 to 17.2 years). Most patients had chiasmatic/hypothalamic tumors (55.5%), and 13 patients (24.1%) had NF1. The most common histology was pilocytic astrocytoma (46.3%). Seventeen patients were diagnosed using radiologic criteria alone. Best response to chemotherapy was centrally reviewed with a response rate (complete, partial, or minor response) of 25.9%. Disease stabilization (complete, partial, or minor response or stable disease) was achieved in 47 patients (87.0%). Visual improvement was observed in 20% of patients with optic pathway glioma. Five-year overall survival and progression-free survival (PFS) rates were 94.4% (95% CI, 88.5% to 100%) and 53.2% (95% CI, 41.3% to 68.5%), respectively, for the entire cohort. Patients with NF1 had a significantly better PFS (85.1%; 95% CI, 68.0% to 100%) when compared with patients without NF1 (42.0%; 95% CI, 29.1% to 60.7%; P = .012). Age< 3 years or > 10 years was not associated with poor outcome. Treatment was well tolerated, and quality of life was not affected during treatment. In this trial, there was no correlation between BRAF alterations and outcome. Conclusion Vinblastine administered once per week is well tolerated in children with treatment naïve PLGG. Overall survival and PFS are comparable to current therapies, with a favorable toxicity profile and a maintained quality of life.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Astrocytoma/drug therapy , Brain Neoplasms/drug therapy , Neurofibromatosis 1/drug therapy , Vinblastine/administration & dosage , Adolescent , Antineoplastic Agents, Phytogenic/adverse effects , Astrocytoma/complications , Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/complications , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Canada , Child , Child, Preschool , Disease-Free Survival , Female , Humans , Infant , Male , Mutation , Neoplasm Grading , Neurofibromatosis 1/pathology , Oncogene Fusion , Proto-Oncogene Proteins B-raf/genetics , Quality of Life , Survival Rate , Vinblastine/adverse effects , Vision Disorders/etiology
16.
J Clin Oncol ; 34(19): 2206-11, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27001570

ABSTRACT

PURPOSE: Recurrent glioblastoma multiforme (GBM) is incurable with current therapies. Biallelic mismatch repair deficiency (bMMRD) is a highly penetrant childhood cancer syndrome often resulting in GBM characterized by a high mutational burden. Evidence suggests that high mutation and neoantigen loads are associated with response to immune checkpoint inhibition. PATIENTS AND METHODS: We performed exome sequencing and neoantigen prediction on 37 bMMRD cancers and compared them with childhood and adult brain neoplasms. Neoantigen prediction bMMRD GBM was compared with responsive adult cancers from multiple tissues. Two siblings with recurrent multifocal bMMRD GBM were treated with the immune checkpoint inhibitor nivolumab. RESULTS: All malignant tumors (n = 32) were hypermutant. Although bMMRD brain tumors had the highest mutational load because of secondary polymerase mutations (mean, 17,740 ± standard deviation, 7,703), all other high-grade tumors were hypermutant (mean, 1,589 ± standard deviation, 1,043), similar to other cancers that responded favorably to immune checkpoint inhibitors. bMMRD GBM had a significantly higher mutational load than sporadic pediatric and adult gliomas and all other brain tumors (P < .001). bMMRD GBM harbored mean neoantigen loads seven to 16 times higher than those in immunoresponsive melanomas, lung cancers, or microsatellite-unstable GI cancers (P < .001). On the basis of these preclinical data, we treated two bMMRD siblings with recurrent multifocal GBM with the anti-programmed death-1 inhibitor nivolumab, which resulted in clinically significant responses and a profound radiologic response. CONCLUSION: This report of initial and durable responses of recurrent GBM to immune checkpoint inhibition may have implications for GBM in general and other hypermutant cancers arising from primary (genetic predisposition) or secondary MMRD.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Brain Neoplasms/genetics , Colorectal Neoplasms/genetics , Glioblastoma/genetics , Mutation , Neoplastic Syndromes, Hereditary/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adult , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/drug therapy , Child , Child, Preschool , Female , Glioblastoma/diagnostic imaging , Glioblastoma/drug therapy , Humans , Magnetic Resonance Imaging , Male , Nivolumab
17.
Acta Neuropathol Commun ; 4: 13, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26860432

ABSTRACT

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".

18.
Acta Neuropathol Commun ; 4: 1, 2016 Jan 04.
Article in English | MEDLINE | ID: mdl-26727948

ABSTRACT

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.


Subject(s)
Brain Stem Neoplasms/diagnosis , Glioma/diagnosis , Mutation/genetics , Adolescent , Biopsy , Brain Stem Neoplasms/genetics , Child , Child, Preschool , Female , Gene Expression Profiling , Glioma/genetics , Humans , Magnetic Resonance Imaging , Male , Severity of Illness Index , Young Adult
20.
Acta Neuropathol ; 129(6): 789-808, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25975377

ABSTRACT

Diffusely infiltrating astrocytomas include diffuse astrocytomas WHO grade II and anaplastic astrocytomas WHO grade III and are classified under astrocytic tumours according to the current WHO Classification. Although the patients generally have longer survival as compared to those with glioblastoma, the timing of inevitable malignant progression ultimately determines the prognosis. Recent advances in molecular genetics have uncovered that histopathologically diagnosed astrocytomas may consist of two genetically different groups of tumours. The majority of diffusely infiltrating astrocytomas regardless of WHO grade have concurrent mutations of IDH1 or IDH2, TP53 and ATRX. Among these astrocytomas, no other genetic markers that may distinguish grade II and grade III tumours have been identified. Those astrocytomas without IDH mutation tend to have a distinct genotype and a poor prognosis comparable to that of glioblastomas. On the other hand, diffuse astrocytomas that arise in children do not harbour IDH/TP53 mutations, but instead display mutations of BRAF or structural alterations involving MYB/MYBL1 or FGFR1. A molecular classification may thus help delineate diffusely infiltrating astrocytomas into distinct pathogenic and prognostic groups, which could aid in determining individualised therapeutic strategies.


Subject(s)
Astrocytoma , Biomarkers, Tumor/metabolism , Brain Neoplasms , Astrocytoma/genetics , Astrocytoma/pathology , Astrocytoma/therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Humans , Neuroimaging
SELECTION OF CITATIONS
SEARCH DETAIL
...