Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Oncologist ; 29(1): e47-e58, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37619245

ABSTRACT

The authors present a cohort of 661 young adult glioblastomas diagnosed using 2016 WHO World Health Organization Classification of Tumors of the Central Nervous System, utilizing comprehensive genomic profiling (CGP) to explore their genomic landscape and assess their relationship to currently defined disease entities. This analysis explored variants with evidence of pathogenic function, common copy number variants (CNVs), and several novel fusion events not described in literature. Tumor mutational burden (TMB) mutational signatures, anatomic location, and tumor recurrence are further explored. Using data collected from CGP, unsupervised machine-learning techniques were leveraged to identify 10 genomic classes in previously assigned young adult glioblastomas. The authors relate these molecular classes to current World Health Organization guidelines and reference current literature to give therapeutic and prognostic descriptions where possible.


Subject(s)
Central Nervous System Neoplasms , Glioblastoma , Humans , Young Adult , Glioblastoma/diagnosis , Glioblastoma/genetics , Retrospective Studies , Mutation , Neoplasm Recurrence, Local , Genomics/methods
3.
Hum Immunol ; 82(11): 859-870, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33648805

ABSTRACT

In the last decade, next-generation sequencing (NGS) has rapidly progressed from a research method to a core component of standard-of-care clinical testing. In oncology, tumor sequencing provides a critical tool to detect somatic driver mutations that not only characterize disease but also impact therapeutic decision-making. Here, we review the important role of NGS in the evaluation of hematopoietic neoplasms. We discusstechnical and practical considerations relevant in somatic mutation testing, emphasizing issues unique to blood cancers. Then, we describe how NGS data is being used to facilitate diagnosis, inform prognosis, guide therapy selection, and even monitor disease. This broad overview highlights the transformative impacts NGS data provides throughout the clinical course of patients with hematologic malignancies.


Subject(s)
Biomarkers, Tumor/genetics , DNA Mutational Analysis/methods , Hematologic Neoplasms/diagnosis , High-Throughput Nucleotide Sequencing , Hematologic Neoplasms/genetics , Hematologic Neoplasms/mortality , Hematologic Neoplasms/therapy , Humans , Mutation , Precision Medicine/methods , Prognosis , Risk Assessment/methods
4.
Front Neurol ; 11: 544680, 2020.
Article in English | MEDLINE | ID: mdl-33192972

ABSTRACT

Despite advances in systemic therapies for solid tumors, the development of brain metastases remains a significant contributor to overall cancer mortality and requires improved methods for diagnosing and treating these lesions. Similarly, the prognosis for malignant primary brain tumors remains poor with little improvement in overall survival over the last several decades. In both primary and metastatic central nervous system (CNS) tumors, the challenge from a clinical perspective centers on detecting CNS dissemination early and understanding how CNS lesions differ from the primary tumor, in order to determine potential treatment strategies. Acquiring tissue from CNS tumors has historically been accomplished through invasive neurosurgical procedures, which restricts the number of patients to those who can safely undergo a surgical procedure, and for which such interventions will add meaningful value to the care of the patient. In this review we discuss the potential of analyzing cell free DNA shed from tumor cells that is contained within the cerebrospinal fluid (CSF) as a sensitive and minimally invasive method to detect and characterize primary and metastatic tumors in the CNS.

5.
PLoS One ; 15(1): e0228356, 2020.
Article in English | MEDLINE | ID: mdl-31995621

ABSTRACT

Pediatric Low Grade Gliomas (PLGGs) display heterogeneity regarding morphology, genomic drivers and clinical outcomes. The treatment modality dictates the outcome and optimizing patient management can be challenging. In this study, we profiled a targeted panel of cancer-related genes in 37 Saudi Arabian patients with pLGGs to identify genetic abnormalities that can inform prognostic and therapeutic decision-making. We detected genetic alterations (GAs) in 97% (36/37) of cases, averaging 2.51 single nucleotide variations (SNVs) and 0.91 gene fusions per patient. The KIAA1549-BRAF fusion was the most common alteration (21/37 patients) followed by AFAP1-NTRK2 (2/37) and TBLXR-PI3KCA (2/37) fusions that were observed at much lower frequencies. The most frequently mutated) genes were NOTCH1-3 (7/37), ATM (4/37), RAD51C (3/37), RNF43 (3/37), SLX4 (3/37) and NF1 (3/37). Interestingly, we identified a GOPC-ROS1 fusion in an 8-year-old patient whose tumor lacked BRAF alterations and histologically classified as low grade glioma. The patient underwent gross total resection (GTR). The patient is currently disease free. To our knowledge this is the first report of GOPC-ROS1 fusion in PLGG. Taken together, we reveal the genetic characteristics of pLGG patients can enhance diagnostics and therapeutic decisions. In addition, we identified a GOPC-ROS1 fusion that may be a biomarker for pLGG.


Subject(s)
Brain Neoplasms/genetics , Gene Fusion , Genomics/methods , Glioma/genetics , Polymorphism, Single Nucleotide , Brain Neoplasms/surgery , Child , Child, Preschool , Clinical Decision-Making , Female , Glioma/surgery , High-Throughput Nucleotide Sequencing , Humans , Infant , Male , Prognosis , Retrospective Studies , Saudi Arabia , Treatment Outcome
6.
J Natl Cancer Inst ; 112(8): 855-858, 2020 08 01.
Article in English | MEDLINE | ID: mdl-31747025

ABSTRACT

PIK3CA is the most frequently mutated gene in human papillomavirus (HPV)-associated oropharyngeal squamous cell carcinoma (OPSCC). Prognostic implications of such mutations remain unknown. We sought to elucidate the clinical significance of PIK3CA mutations in HPV-associated OPSCC patients treated with definitive chemoradiation (CRT). Seventy-seven patients with HPV-associated OPSCC were enrolled on two phase II clinical trials of deintensified CRT (60 Gy intensity-modulated radiotherapy with concurrent weekly cisplatin). Targeted next-generation sequencing was performed. Of the 77 patients, nine had disease recurrence (two regional, four distant, three regional and distant). Thirty-four patients had mutation(s) identified; 16 had PIK3CA mutations. Patients with wild-type-PIK3CA had statistically significantly higher 3-year disease-free survival than PIK3CA-mutant patients (93.4%, 95% confidence interval [CI] = 85.0% to 99.9% vs 68.8%, 95% CI = 26.7% to 89.8%; P = .004). On multivariate analysis, PIK3CA mutation was the only variable statistically significantly associated with disease recurrence (hazard ratio = 5.71, 95% CI = 1.53 to 21.3; P = .01). PIK3CA mutation is associated with worse disease-free survival in a prospective cohort of newly diagnosed HPV-associated OPSCC patients treated with deintensified CRT.


Subject(s)
Alphapapillomavirus/physiology , Carcinoma, Squamous Cell , Chemoradiotherapy/methods , Class I Phosphatidylinositol 3-Kinases/genetics , Oropharyngeal Neoplasms , Adult , Aged , Aged, 80 and over , Alphapapillomavirus/pathogenicity , Carcinoma, Squamous Cell/diagnosis , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/therapy , Carcinoma, Squamous Cell/virology , Clinical Trials, Phase II as Topic/statistics & numerical data , Cohort Studies , DNA Mutational Analysis , Disease-Free Survival , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged , Mutation , Oropharyngeal Neoplasms/diagnosis , Oropharyngeal Neoplasms/genetics , Oropharyngeal Neoplasms/therapy , Oropharyngeal Neoplasms/virology , Prognosis , Prospective Studies , Radiotherapy Dosage , Treatment Outcome
7.
Curr Treat Options Oncol ; 19(8): 41, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29931654

ABSTRACT

OPINION STATEMENT: In recent years, large-scale genomic studies have expanded our knowledge regarding genomic drivers in tumors of the central nervous system. While histopathologic analysis of brain tumors remains the primary method for tumor classification, the clinical utility of molecular and genomic testing to support and/or complement tumor classification continues to expand. This approach enhances diagnostic accuracy and provides clinicians with objective data to facilitate discussions regarding prognosis and treatment decisions, including selection of clinical trials. Ensuring accurate diagnoses is fundamental to the management of brain tumor patients. However, given the morphologic overlap among primary brain tumors, genomic data can be used to help distinguish tumor lineage. In its clearest form, we have embraced the concept of an integrated diagnosis, which combines traditional histopathology findings with molecular and genomic data. Patient prognosis varies significantly based on a tumor's genomic profile. For neuro-oncology patients, outcome studies linking diagnoses with genomic profiles show significant differences based on tumor biomarkers such as IDH1/2, H3F3A, BRAF, and CDKN2A and TERT status. Therefore, easy access to reliable genomic data is important in understanding a patient's disease and developing a clinical strategy wherein targeted molecular or immune therapies can be incorporated into the discussion.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Glioma/diagnosis , Glioma/therapy , Precision Medicine , Age Factors , Biomarkers, Tumor , Brain Neoplasms/genetics , Brain Neoplasms/mortality , Clinical Trials as Topic , Combined Modality Therapy , Disease Management , Genetic Predisposition to Disease , Genetic Testing , Genomics/methods , Glioma/genetics , Glioma/mortality , Humans , Immunotherapy , Molecular Targeted Therapy , Neoplasm Grading , Neoplasm Staging , Precision Medicine/methods , Prognosis , Treatment Outcome
8.
Neuro Oncol ; 19(7): 986-996, 2017 Jul 01.
Article in English | MEDLINE | ID: mdl-28104717

ABSTRACT

BACKGROUND: Clinical genomics platforms are needed to identify targetable alterations, but implementation of these technologies and best practices in routine clinical pediatric oncology practice are not yet well established. METHODS: Profile is an institution-wide prospective clinical research initiative that uses targeted sequencing to identify targetable alterations in tumors. OncoPanel, a multiplexed targeted exome-sequencing platform that includes 300 cancer-causing genes, was used to assess single nucleotide variants and rearrangements/indels. Alterations were annotated (Tiers 1-4) based on clinical significance, with Tier 1 alterations having well-established clinical utility. OncoCopy, a clinical genome-wide array comparative genomic hybridization (aCGH) assay, was also performed to evaluate copy number alterations and better define rearrangement breakpoints. RESULTS: Cancer genomes of 203 pediatric brain tumors were profiled across histological subtypes, including 117 samples analyzed by OncoPanel, 146 by OncoCopy, and 60 tumors subjected to both methodologies. OncoPanel revealed clinically relevant alterations in 56% of patients (44 cancer mutations and 20 rearrangements), including BRAF alterations that directed the use of targeted inhibitors. Rearrangements in MYB-QKI, MYBL1, BRAF, and FGFR1 were also detected. Furthermore, while copy number profiles differed across histologies, the combined use of OncoPanel and OncoCopy identified subgroup-specific alterations in 89% (17/19) of medulloblastomas. CONCLUSION: The combination of OncoPanel and OncoCopy multiplex genomic assays can identify critical diagnostic, prognostic, and treatment-relevant alterations and represents an effective precision medicine approach for clinical evaluation of pediatric brain tumors.


Subject(s)
Brain Neoplasms/genetics , DNA Copy Number Variations , Exome , Genomics/methods , Precision Medicine/methods , Brain Neoplasms/diagnosis , Child , Comparative Genomic Hybridization , Gene Dosage , Humans , Mutation , Polymorphism, Single Nucleotide , Sequence Analysis, DNA
9.
Neuro Oncol ; 19(6): 774-785, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28082416

ABSTRACT

Background: Activating mutations or structural rearrangements in BRAF are identified in roughly 75% of all pediatric low-grade astrocytomas (PLGAs). However, first-generation RAF inhibitors approved for adult melanoma have poor blood-brain penetrance and are only effective on tumors that express the canonical BRAFV600E oncoprotein, which functions as a monomer. These drugs (type I antagonists that target the "DFG-in" conformation of the kinase) fail to block signaling via KIAA1549:BRAF, a truncation/fusion BRAF oncoprotein which functions as a dimer and is found in the most common form of PLGA. Methods: A panel of small molecule RAF inhibitors (including type II inhibitors, targeting the "DFG-out" conformation of the kinase) was screened for drugs showing efficacy on murine models of PLGA and on authentic human PLGA cells expressing KIAA1549:BRAF. Results: We identify a type II RAF inhibitor that serves as an equipotent antagonist of BRAFV600E, KIAA1549:BRAF, and other noncanonical BRAF oncoproteins that function as dimers. This drug (MLN2480, also known as TAK-580) has good brain penetrance and is active on authentic human PLGA cells in brain organotypic cultures. Conclusion: MLN2480 may be an effective therapeutic for BRAF mutant pediatric astrocytomas.


Subject(s)
Astrocytoma/drug therapy , Brain Neoplasms/drug therapy , Heterocyclic Compounds, 3-Ring/pharmacology , Oncogene Proteins, Fusion/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Multimerization/drug effects , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , raf Kinases/antagonists & inhibitors , Animals , Astrocytoma/metabolism , Astrocytoma/pathology , Blood-Brain Barrier/drug effects , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Child , Heterocyclic Compounds, 3-Ring/chemistry , High-Throughput Screening Assays , Humans , Male , Mice , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins B-raf/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , raf Kinases/genetics , raf Kinases/metabolism
10.
Nat Med ; 22(7): 723-6, 2016 07.
Article in English | MEDLINE | ID: mdl-27270588

ABSTRACT

Brain metastases represent the greatest clinical challenge in treating HER2-positive breast cancer. We report the development of orthotopic patient-derived xenografts (PDXs) of HER2-expressing breast cancer brain metastases (BCBM), and their use for the identification of targeted combination therapies. Combined inhibition of PI3K and mTOR resulted in durable tumor regressions in three of five PDXs, and therapeutic response was correlated with a reduction in the phosphorylation of 4EBP1, an mTORC1 effector. The two nonresponding PDXs showed hypermutated genomes with enrichment of mutations in DNA-repair genes, which suggests an association of genomic instability with therapeutic resistance. These findings suggest that a biomarker-driven clinical trial of PI3K inhibitor in combination with an mTOR inhibitor should be conducted for patients with HER2-positive BCBM.


Subject(s)
Aminopyridines/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Breast Neoplasms/drug therapy , Everolimus/pharmacology , Morpholines/pharmacology , Multiprotein Complexes/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adaptor Proteins, Signal Transducing , Animals , Brain Neoplasms/genetics , Brain Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carrier Proteins/drug effects , Carrier Proteins/metabolism , Caspase 3/drug effects , Caspase 3/metabolism , Cell Cycle Proteins , DNA Repair/genetics , Drug Resistance, Neoplasm/genetics , Drug Therapy, Combination , Eukaryotic Initiation Factors , Female , Gene Expression Profiling , Genomic Instability , Humans , Immunohistochemistry , Ki-67 Antigen/drug effects , Ki-67 Antigen/metabolism , Magnetic Resonance Imaging , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, SCID , Molecular Targeted Therapy , Neoplasm Transplantation , Phosphoproteins/drug effects , Phosphoproteins/metabolism , Phosphorylation , Receptor, ErbB-2/metabolism , Remission Induction , Xenograft Model Antitumor Assays
11.
Oncotarget ; 7(24): 37054-37063, 2016 Jun 14.
Article in English | MEDLINE | ID: mdl-27175596

ABSTRACT

Pituitary spindle cell oncocytoma (SCO) is an uncommon primary pituitary neoplasm that presents with mass effect on adjacent neurovascular structures, similar to non-hormone-producing pituitary adenomas. To determine the molecular etiology of SCO, we performed exome sequencing on four SCO cases, with matched normal controls, to assess somatic mutations and copy number alterations. Our analysis revealed a low mutation rate and a copy-neutral profile, consistent with the low-grade nature of this tumor. However, we identified a co-occurring somatic HRAS (p.Q61R) activating point mutation and MEN1 frameshift mutation (p.L117fs) present in a primary and recurrent tumor from one patient. Other SCOs demonstrated mutations in SND1 and FAT1, which are associated with MAPK pathway activation. Immunohistochemistry across the SCO cohort demonstrated robust MAPK activity in all cases (n=4), as evidenced by strong phospho-ERK staining, while phospho-AKT levels suggested only basal levels of PI3K pathway activation. Taken together, this identifies the MAPK signaling pathway as a novel therapeutic target for spindle cell oncocytoma, which may offer a powerful adjunct for aggressive tumors refractory to surgical resection.


Subject(s)
Adenoma, Oxyphilic/genetics , MAP Kinase Signaling System/physiology , Pituitary Neoplasms/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Adenoma, Oxyphilic/metabolism , Aged , DNA Mutational Analysis , Humans , Middle Aged , Mutation , Pituitary Neoplasms/metabolism
12.
Nat Genet ; 48(3): 273-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26829751

ABSTRACT

Angiocentric gliomas are pediatric low-grade gliomas (PLGGs) without known recurrent genetic drivers. We performed genomic analysis of new and published data from 249 PLGGs, including 19 angiocentric gliomas. We identified MYB-QKI fusions as a specific and single candidate driver event in angiocentric gliomas. In vitro and in vivo functional studies show that MYB-QKI rearrangements promote tumorigenesis through three mechanisms: MYB activation by truncation, enhancer translocation driving aberrant MYB-QKI expression and hemizygous loss of the tumor suppressor QKI. To our knowledge, this represents the first example of a single driver rearrangement simultaneously transforming cells via three genetic and epigenetic mechanisms in a tumor.


Subject(s)
Glioma/genetics , Oncogene Proteins v-myb/genetics , Oncogene Proteins, Fusion/genetics , RNA-Binding Proteins/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Child , Comparative Genomic Hybridization , Exome/genetics , Gene Expression Regulation, Neoplastic , Gene Rearrangement , Glioma/pathology , High-Throughput Nucleotide Sequencing , Humans , Mutation , Oncogene Proteins v-myb/biosynthesis , Oncogene Proteins, Fusion/biosynthesis , RNA-Binding Proteins/biosynthesis
13.
Neuro Oncol ; 17(10): 1344-55, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25754088

ABSTRACT

BACKGROUND: Multidimensional genotyping of formalin-fixed paraffin-embedded (FFPE) samples has the potential to improve diagnostics and clinical trials for brain tumors, but prospective use in the clinical setting is not yet routine. We report our experience with implementing a multiplexed copy number and mutation-testing program in a diagnostic laboratory certified by the Clinical Laboratory Improvement Amendments. METHODS: We collected and analyzed clinical testing results from whole-genome array comparative genomic hybridization (OncoCopy) of 420 brain tumors, including 148 glioblastomas. Mass spectrometry-based mutation genotyping (OncoMap, 471 mutations) was performed on 86 glioblastomas. RESULTS: OncoCopy was successful in 99% of samples for which sufficient DNA was obtained (n = 415). All clinically relevant loci for glioblastomas were detected, including amplifications (EGFR, PDGFRA, MET) and deletions (EGFRvIII, PTEN, 1p/19q). Glioblastoma patients ≤40 years old had distinct profiles compared with patients >40 years. OncoMap testing reliably identified mutations in IDH1, TP53, and PTEN. Seventy-seven glioblastoma patients enrolled on trials, of whom 51% participated in targeted therapeutic trials where multiplex data informed eligibility or outcomes. Data integration identified patients with complete tumor suppressor inactivation, albeit rarely (5% of patients) due to lack of whole-gene coverage in OncoMap. CONCLUSIONS: Combined use of multiplexed copy number and mutation detection from FFPE samples in the clinical setting can efficiently replace singleton tests for clinical diagnosis and prognosis in most settings. Our results support incorporation of these assays into clinical trials as integral biomarkers and their potential to impact interpretation of results. Limited tumor suppressor variant capture by targeted genotyping highlights the need for whole-gene sequencing in glioblastoma.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , DNA Copy Number Variations , Genome-Wide Association Study , Glioblastoma/diagnosis , Glioblastoma/genetics , Mutation , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Comparative Genomic Hybridization , Female , Gene Expression Profiling , Genotype , Humans , Infant , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , PTEN Phosphohydrolase/genetics , Prospective Studies , Tumor Suppressor Protein p53/genetics , Young Adult
14.
Neuro Oncol ; 17(5): 697-707, 2015 May.
Article in English | MEDLINE | ID: mdl-25366336

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is poorly responsive to current chemotherapy. The nuclear transporter exportin 1 (XPO1, CRM1) is often highly expressed in GBM, which may portend a poor prognosis. Here, we determine the efficacy of novel selective inhibitors of nuclear export (SINE) specific to XPO1 in preclinical models of GBM. METHODS: Seven patient-derived GBM lines were treated with 3 SINE compounds (KPT-251, KPT-276, and Selinexor) in neurosphere culture conditions. KPT-276 and Selinexor were also evaluated in a murine orthotopic patient-derived xenograft (PDX) model of GBM. Cell cycle effects were assayed by flow cytometry in vitro and immunohistochemistry in vivo. Apoptosis was determined by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and caspase 3/7 activity assays. RESULTS: Treatment of GBM neurosphere cultures with KPT-276, Selinexor, and KPT-251 revealed dose-responsive growth inhibition in all 7 GBM lines [range of half-maximal inhibitory concentration (IC50), 6-354 nM]. In an orthotopic PDX model, treatment with KPT-276 and Selinexor demonstrated pharmacodynamic efficacy, significantly suppressed tumor growth, and prolonged animal survival. Cellular proliferation was not altered with SINE treatment. Instead, induction of apoptosis was apparent both in vitro and in vivo with SINE treatment, without overt evidence of neurotoxicity. CONCLUSIONS: SINE compounds show preclinical efficacy utilizing in vitro and in vivo models of GBM, with induction of apoptosis as the mechanism of action. Selinexor is now in early clinical trials in solid and hematological malignancies. Based on these preclinical data and excellent brain penetration, we have initiated clinical trials of Selinexor in patients with relapsed GBM.


Subject(s)
Acrylamides/therapeutic use , Active Transport, Cell Nucleus/drug effects , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Hydrazines/therapeutic use , Karyopherins/antagonists & inhibitors , Oxadiazoles/pharmacology , Oxadiazoles/therapeutic use , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Thiazoles/therapeutic use , Triazoles/pharmacology , Triazoles/therapeutic use , Acrylamides/pharmacokinetics , Acrylamides/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Hydrazines/pharmacokinetics , Hydrazines/pharmacology , Karyopherins/metabolism , Macaca fascicularis , Male , Mice , Rats, Sprague-Dawley , Receptors, Cytoplasmic and Nuclear/metabolism , Thiazoles/pharmacokinetics , Thiazoles/pharmacology , Treatment Outcome , Triazoles/pharmacokinetics , Xenograft Model Antitumor Assays , Exportin 1 Protein
15.
Oncotarget ; 5(18): 8083-92, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25257301

ABSTRACT

Classifying adult gliomas remains largely a histologic diagnosis based on morphology; however astrocytic, oligodendroglial and mixed lineage tumors can display overlapping histologic features. We used multiplexed exome sequencing (OncoPanel) on 108 primary or recurrent adult gliomas, comprising 65 oligodendrogliomas, 28 astrocytomas and 15 mixed oligoastrocytomas to identify lesions that could enhance lineage classification. Mutations in TP53 (20/28, 71%) and ATRX (15/28, 54%) were enriched in astrocytic tumors compared to oligodendroglial tumors of which 4/65 (6%) had mutations in TP53 and 2/65 (3%) had ATRX mutations. We found that oligoastrocytomas harbored mutations in TP53 (80%, 12/15) and ATRX (60%, 9/15) at frequencies similar to pure astrocytic tumors, suggesting that oligoastrocytomas and astrocytomas may represent a single genetic or biological entity. p53 protein expression correlated with mutation status and showed significant increases in astrocytomas and oligoastrocytomas compared to oligodendrogliomas, a finding that also may facilitate accurate classification. Furthermore our OncoPanel analysis revealed that 15% of IDH1/2 mutant gliomas would not be detected by traditional IDH1 (p.R132H) antibody testing, supporting the use of genomic technologies in providing clinically relevant data. In all, our results demonstrate that multiplexed exome sequencing can support evaluation and classification of adult low-grade gliomas with a single clinical test.


Subject(s)
Astrocytoma/genetics , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Cell Lineage , DNA Mutational Analysis , Exome , Genetic Testing/methods , Mutation , Oligodendroglioma/genetics , Astrocytoma/chemistry , Astrocytoma/classification , Astrocytoma/pathology , Biomarkers, Tumor/analysis , Brain Neoplasms/chemistry , Brain Neoplasms/classification , Brain Neoplasms/pathology , DNA Helicases/genetics , Diagnosis, Differential , ErbB Receptors/genetics , Gene Expression Profiling , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Immunohistochemistry , Isocitrate Dehydrogenase/genetics , Nuclear Proteins/genetics , Oligodendroglioma/chemistry , Oligodendroglioma/classification , Oligodendroglioma/pathology , PTEN Phosphohydrolase/genetics , Phenotype , Predictive Value of Tests , Reproducibility of Results , Tumor Suppressor Protein p53/genetics , X-linked Nuclear Protein
16.
Nat Genet ; 46(5): 462-6, 2014 May.
Article in English | MEDLINE | ID: mdl-24705250

ABSTRACT

Pediatric midline high-grade astrocytomas (mHGAs) are incurable with few treatment targets identified. Most tumors harbor mutations encoding p.Lys27Met in histone H3 variants. In 40 treatment-naive mHGAs, 39 analyzed by whole-exome sequencing, we find additional somatic mutations specific to tumor location. Gain-of-function mutations in ACVR1 occur in tumors of the pons in conjunction with histone H3.1 p.Lys27Met substitution, whereas FGFR1 mutations or fusions occur in thalamic tumors associated with histone H3.3 p.Lys27Met substitution. Hyperactivation of the bone morphogenetic protein (BMP)-ACVR1 developmental pathway in mHGAs harboring ACVR1 mutations led to increased levels of phosphorylated SMAD1, SMAD5 and SMAD8 and upregulation of BMP downstream early-response genes in tumor cells. Global DNA methylation profiles were significantly associated with the p.Lys27Met alteration, regardless of the mutant histone H3 variant and irrespective of tumor location, supporting the role of this substitution in driving the epigenetic phenotype. This work considerably expands the number of potential treatment targets and further justifies pretreatment biopsy in pediatric mHGA as a means to orient therapeutic efforts in this disease.


Subject(s)
Activin Receptors, Type I/genetics , Astrocytoma/genetics , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , Genome, Human/genetics , Mutation/genetics , Animals , Base Sequence , Bone Morphogenetic Proteins/metabolism , Child , DNA Copy Number Variations/genetics , DNA Methylation/genetics , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Sequence Analysis, DNA , Smad Proteins/metabolism
17.
Proc Natl Acad Sci U S A ; 110(20): 8188-93, 2013 May 14.
Article in English | MEDLINE | ID: mdl-23633565

ABSTRACT

Pediatric low-grade gliomas (PLGGs) are among the most common solid tumors in children but, apart from BRAF kinase mutations or duplications in specific subclasses, few genetic driver events are known. Diffuse PLGGs comprise a set of uncommon subtypes that exhibit invasive growth and are therefore especially challenging clinically. We performed high-resolution copy-number analysis on 44 formalin-fixed, paraffin-embedded diffuse PLGGs to identify recurrent alterations. Diffuse PLGGs exhibited fewer such alterations than adult low-grade gliomas, but we identified several significantly recurrent events. The most significant event, 8q13.1 gain, was observed in 28% of diffuse astrocytoma grade IIs and resulted in partial duplication of the transcription factor MYBL1 with truncation of its C-terminal negative-regulatory domain. A similar recurrent deletion-truncation breakpoint was identified in two angiocentric gliomas in the related gene v-myb avian myeloblastosis viral oncogene homolog (MYB) on 6q23.3. Whole-genome sequencing of a MYBL1-rearranged diffuse astrocytoma grade II demonstrated MYBL1 tandem duplication and few other events. Truncated MYBL1 transcripts identified in this tumor induced anchorage-independent growth in 3T3 cells and tumor formation in nude mice. Truncated transcripts were also expressed in two additional tumors with MYBL1 partial duplication. Our results define clinically relevant molecular subclasses of diffuse PLGGs and highlight a potential role for the MYB family in the biology of low-grade gliomas.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Proto-Oncogene Proteins/genetics , Trans-Activators/genetics , 3T3 Cells , Alleles , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Child , Child, Preschool , Cohort Studies , Comparative Genomic Hybridization , Glioma/pathology , Humans , Male , Mice , Mice, Nude , Multigene Family , Mutation , Protein Structure, Tertiary , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...