Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Strahlenther Onkol ; 199(12): 1214-1224, 2023 12.
Article in English | MEDLINE | ID: mdl-37658922

ABSTRACT

PURPOSE: Radiotherapy is a major pillar in the treatment of solid tumors including breast cancer. However, epidemiological studies have revealed an increase in cardiac diseases approximately a decade after exposure of the thorax to ionizing irradiation, which might be related to vascular inflammation. Therefore, chronic inflammatory effects were examined in primary heart and lung endothelial cells (ECs) of mice after local heart irradiation. METHODS: Long-lasting effects on primary ECs of the heart and lung were studied 20-50 weeks after local irradiation of the heart of mice (8 and 16 Gy) in vivo by multiparameter flow cytometry using antibodies directed against cell surface markers related to proliferation, stemness, lipid metabolism, and inflammation, and compared to those induced by occlusion of the left anterior descending coronary artery. RESULTS: In vivo irradiation of the complete heart caused long-lasting persistent upregulation of inflammatory (HCAM, ICAM­1, VCAM-1), proliferation (CD105), and lipid (CD36) markers on primary heart ECs and an upregulation of ICAM­1 and VCAM­1 on primary ECs of the partially irradiated lung lobe. An artificially induced heart infarction induces similar effects with respect to inflammatory markers, albeit in a shorter time period. CONCLUSION: The long-lasting upregulation of prominent inflammatory markers on primary heart and lung ECs suggests that local heart irradiation induces chronic inflammation in the microvasculature of the heart and partially irradiated lung that leads to cardiac injury which might be related to altered lipid metabolism in the heart.


Subject(s)
Atherosclerosis , Intercellular Adhesion Molecule-1 , Mice , Animals , Endothelial Cells/metabolism , Endothelial Cells/pathology , Vascular Cell Adhesion Molecule-1 , Inflammation , Atherosclerosis/etiology , Thorax , Mice, Inbred C57BL
2.
Neuro Oncol ; 25(11): 2087-2097, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37075810

ABSTRACT

BACKGROUND: The international, multicenter registry LOGGIC Core BioClinical Data Bank aims to enhance the understanding of tumor biology in pediatric low-grade glioma (pLGG) and provide clinical and molecular data to support treatment decisions and interventional trial participation. Hence, the question arises whether implementation of RNA sequencing (RNA-Seq) using fresh frozen (FrFr) tumor tissue in addition to gene panel and DNA methylation analysis improves diagnostic accuracy and provides additional clinical benefit. METHODS: Analysis of patients aged 0 to 21 years, enrolled in Germany between April 2019 and February 2021, and for whom FrFr tissue was available. Central reference histopathology, immunohistochemistry, 850k DNA methylation analysis, gene panel sequencing, and RNA-Seq were performed. RESULTS: FrFr tissue was available in 178/379 enrolled cases. RNA-Seq was performed on 125 of these samples. We confirmed KIAA1549::BRAF-fusion (n = 71), BRAF V600E-mutation (n = 12), and alterations in FGFR1 (n = 14) as the most frequent alterations, among other common molecular drivers (n = 12). N = 16 cases (13%) presented rare gene fusions (eg, TPM3::NTRK1, EWSR1::VGLL1, SH3PXD2A::HTRA1, PDGFB::LRP1, GOPC::ROS1). In n = 27 cases (22%), RNA-Seq detected a driver alteration not otherwise identified (22/27 actionable). The rate of driver alteration detection was hereby increased from 75% to 97%. Furthermore, FGFR1 internal tandem duplications (n = 6) were only detected by RNA-Seq using current bioinformatics pipelines, leading to a change in analysis protocols. CONCLUSIONS: The addition of RNA-Seq to current diagnostic methods improves diagnostic accuracy, making precision oncology treatments (MEKi/RAFi/ERKi/NTRKi/FGFRi/ROSi) more accessible. We propose to include RNA-Seq as part of routine diagnostics for all pLGG patients, especially when no common pLGG alteration was identified.


Subject(s)
Glioma , Proto-Oncogene Proteins B-raf , Child , Humans , Proto-Oncogene Proteins B-raf/genetics , Pathology, Molecular , Protein-Tyrosine Kinases , RNA-Seq , Proto-Oncogene Proteins/genetics , Precision Medicine , Glioma/pathology , DNA-Binding Proteins/genetics , Transcription Factors/genetics
4.
Acta Neuropathol ; 145(1): 49-69, 2023 01.
Article in English | MEDLINE | ID: mdl-36437415

ABSTRACT

Pediatric central nervous system (CNS) tumors represent the most common cause of cancer-related death in children aged 0-14 years. They differ from their adult counterparts, showing extensive clinical and molecular heterogeneity as well as a challenging histopathological spectrum that often impairs accurate diagnosis. Here, we use DNA methylation-based CNS tumor classification in combination with copy number, RNA-seq, and ChIP-seq analysis to characterize a newly identified CNS tumor type. In addition, we report histology, patient characteristics, and survival data in this tumor type. We describe a biologically distinct pediatric CNS tumor type (n = 31 cases) that is characterized by focal high-level amplification and resultant overexpression of either PLAGL1 or PLAGL2, and an absence of recurrent genetic alterations characteristic of other pediatric CNS tumor types. Both genes act as transcription factors for a regulatory subset of imprinted genes (IGs), components of the Wnt/ß-Catenin pathway, and the potential drug targets RET and CYP2W1, which are also specifically overexpressed in this tumor type. A derived PLAGL-specific gene expression signature indicates dysregulation of imprinting control and differentiation/development. These tumors occurred throughout the neuroaxis including the cerebral hemispheres, cerebellum, and brainstem, and were predominantly composed of primitive embryonal-like cells lacking robust expression of markers of glial or neuronal differentiation (e.g., GFAP, OLIG2, and synaptophysin). Tumors with PLAGL1 amplification were typically diagnosed during adolescence (median age 10.5 years), whereas those with PLAGL2 amplification were diagnosed during early childhood (median age 2 years). The 10-year overall survival was 66% for PLAGL1-amplified tumors, 25% for PLAGL2-amplified tumors, 18% for male patients, and 82% for female patients. In summary, we describe a new type of biologically distinct CNS tumor characterized by PLAGL1/2 amplification that occurs predominantly in infants and toddlers (PLAGL2) or adolescents (PLAGL1) which we consider best classified as a CNS embryonal tumor and which is associated with intermediate survival. The cell of origin and optimal treatment strategies remain to be defined.


Subject(s)
Central Nervous System Neoplasms , Neuroectodermal Tumors, Primitive , Child , Child, Preschool , Female , Humans , Infant , Male , Cell Cycle Proteins/genetics , Central Nervous System Neoplasms/genetics , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Neuroectodermal Tumors, Primitive/genetics , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Wnt Signaling Pathway/genetics
5.
Nat Commun ; 12(1): 5530, 2021 09 20.
Article in English | MEDLINE | ID: mdl-34545083

ABSTRACT

Long-term complications such as radiation-induced second malignancies occur in a subset of patients following radiation-therapy, particularly relevant in pediatric patients due to the long follow-up period in case of survival. Radiation-induced gliomas (RIGs) have been reported in patients after treatment with cranial irradiation for various primary malignancies such as acute lymphoblastic leukemia (ALL) and medulloblastoma (MB). We perform comprehensive (epi-) genetic and expression profiling of RIGs arising after cranial irradiation for MB (n = 23) and ALL (n = 9). Our study reveals a unifying molecular signature for the majority of RIGs, with recurrent PDGFRA amplification and loss of CDKN2A/B and an absence of somatic hotspot mutations in genes encoding histone 3 variants or IDH1/2, uncovering diagnostic markers and potentially actionable targets.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p15/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Amplification , Glioma/genetics , Neoplasm Recurrence, Local/pathology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Adolescent , Adult , Child , Chromosome Deletion , Cluster Analysis , DNA Methylation/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Rearrangement/genetics , Genome, Human , Glioma/pathology , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Radiation , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Transcription, Genetic , Young Adult
6.
Acta Neuropathol ; 142(5): 841-857, 2021 11.
Article in English | MEDLINE | ID: mdl-34417833

ABSTRACT

Large-scale molecular profiling studies in recent years have shown that central nervous system (CNS) tumors display a much greater heterogeneity in terms of molecularly distinct entities, cellular origins and genetic drivers than anticipated from histological assessment. DNA methylation profiling has emerged as a useful tool for robust tumor classification, providing new insights into these heterogeneous molecular classes. This is particularly true for rare CNS tumors with a broad morphological spectrum, which are not possible to assign as separate entities based on histological similarity alone. Here, we describe a molecularly distinct subset of predominantly pediatric CNS neoplasms (n = 60) that harbor PATZ1 fusions. The original histological diagnoses of these tumors covered a wide spectrum of tumor types and malignancy grades. While the single most common diagnosis was glioblastoma (GBM), clinical data of the PATZ1-fused tumors showed a better prognosis than typical GBM, despite frequent relapses. RNA sequencing revealed recurrent MN1:PATZ1 or EWSR1:PATZ1 fusions related to (often extensive) copy number variations on chromosome 22, where PATZ1 and the two fusion partners are located. These fusions have individually been reported in a number of glial/glioneuronal tumors, as well as extracranial sarcomas. We show here that they are more common than previously acknowledged, and together define a biologically distinct CNS tumor type with high expression of neural development markers such as PAX2, GATA2 and IGF2. Drug screening performed on the MN1:PATZ1 fusion-bearing KS-1 brain tumor cell line revealed preliminary candidates for further study. In summary, PATZ1 fusions define a molecular class of histologically polyphenotypic neuroepithelial tumors, which show an intermediate prognosis under current treatment regimens.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Kruppel-Like Transcription Factors/genetics , Neoplasms, Neuroepithelial/genetics , Neoplasms, Neuroepithelial/pathology , Repressor Proteins/genetics , Biomarkers, Tumor/genetics , Child , Child, Preschool , Female , Humans , Male , Oncogene Fusion , Oncogene Proteins, Fusion/genetics
7.
Acta Neuropathol ; 141(4): 605-617, 2021 04.
Article in English | MEDLINE | ID: mdl-33585982

ABSTRACT

Low-grade gliomas (LGGs) are the most common childhood brain tumor in the general population and in individuals with the Neurofibromatosis type 1 (NF1) cancer predisposition syndrome. Surgical biopsy is rarely performed prior to treatment in the setting of NF1, resulting in a paucity of tumor genomic information. To define the molecular landscape of NF1-associated LGGs (NF1-LGG), we integrated clinical data, histological diagnoses, and multi-level genetic/genomic analyses on 70 individuals from 25 centers worldwide. Whereas, most tumors harbored bi-allelic NF1 inactivation as the only genetic abnormality, 11% had additional mutations. Moreover, tumors classified as non-pilocytic astrocytoma based on DNA methylation analysis were significantly more likely to harbor these additional mutations. The most common secondary alteration was FGFR1 mutation, which conferred an additional growth advantage in multiple complementary experimental murine Nf1 models. Taken together, this comprehensive characterization has important implications for the management of children with NF1-LGG, distinct from their sporadic counterparts.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Neurofibromatosis 1/complications , Adolescent , Animals , Child , Child, Preschool , Female , Humans , Infant , Male , Mice , Mutation
8.
Acta Neuropathol ; 139(2): 243-257, 2020 02.
Article in English | MEDLINE | ID: mdl-31768671

ABSTRACT

Tumors of the pineal region comprise several different entities with distinct clinical and histopathological features. Whereas some entities predominantly affect adults, pineoblastoma (PB) constitutes a highly aggressive malignancy of childhood with a poor outcome. PBs mainly arise sporadically, but may also occur in the context of cancer predisposition syndromes including DICER1 and RB1 germline mutation. With this study, we investigate clinico-pathological subgroups of pineal tumors and further characterize their biological features. We performed genome-wide DNA methylation analysis in 195 tumors of the pineal region and 20 normal pineal gland controls. Copy-number profiles were obtained from DNA methylation data; gene panel sequencing was added for 93 tumors and analysis was further complemented by miRNA sequencing for 22 tumor samples. Unsupervised clustering based on DNA methylation profiling separated known subgroups, like pineocytoma, pineal parenchymal tumor of intermediate differentiation, papillary tumor of the pineal region and PB, and further distinct subtypes within these groups, including three subtypes within the core PB subgroup. The novel molecular subgroup Pin-RB includes cases of trilateral retinoblastoma as well as sporadic pineal tumors with RB1 alterations, and displays similarities with retinoblastoma. Distinct clinical associations discriminate the second novel molecular subgroup PB-MYC from other PB cases. Alterations within the miRNA processing pathway (affecting DROSHA, DGCR8 or DICER1) are found in about two thirds of cases in the three core PB subtypes. Methylation profiling revealed biologically distinct groups of pineal tumors with specific clinical and molecular features. Our findings provide a foundation for further clinical as well as molecular and functional characterization of PB and other pineal tumors, including the role of miRNA processing defects in oncogenesis.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Pineal Gland , Pinealoma/genetics , Pinealoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Brain Neoplasms/metabolism , Case-Control Studies , Child , DNA Methylation , Female , Humans , Male , MicroRNAs , Middle Aged , Mutation/genetics , Pinealoma/metabolism , Young Adult
10.
Acta Neuropathol ; 136(2): 239-253, 2018 08.
Article in English | MEDLINE | ID: mdl-29766299

ABSTRACT

Diffuse leptomeningeal glioneuronal tumors (DLGNT) represent rare CNS neoplasms which have been included in the 2016 update of the WHO classification. The wide spectrum of histopathological and radiological features can make this enigmatic tumor entity difficult to diagnose. In recent years, large-scale genomic and epigenomic analyses have afforded insight into key genetic alterations occurring in multiple types of brain tumors and provide unbiased, complementary tools to improve diagnostic accuracy. Through genome-wide DNA methylation screening of > 25,000 tumors, we discovered a molecularly distinct class comprising 30 tumors, mostly diagnosed histologically as DLGNTs. Copy-number profiles derived from the methylation arrays revealed unifying characteristics, including loss of chromosomal arm 1p in all cases. Furthermore, this molecular DLGNT class can be subdivided into two subgroups [DLGNT methylation class (MC)-1 and DLGNT methylation class (MC)-2], with all DLGNT-MC-2 additionally displaying a gain of chromosomal arm 1q. Co-deletion of 1p/19q, commonly seen in IDH-mutant oligodendroglioma, was frequently observed in DLGNT, especially in DLGNT-MC-1 cases. Both subgroups also had recurrent genetic alterations leading to an aberrant MAPK/ERK pathway, with KIAA1549:BRAF fusion being the most frequent event. Other alterations included fusions of NTRK1/2/3 and TRIM33:RAF1, adding up to an MAPK/ERK pathway activation identified in 80% of cases. In the DLGNT-MC-1 group, age at diagnosis was significantly lower (median 5 vs 14 years, p < 0.01) and clinical course less aggressive (5-year OS 100, vs 43% in DLGNT-MC-2). Our study proposes an additional molecular layer to the current histopathological classification of DLGNT, of particular use for cases without typical morphological or radiological characteristics, such as diffuse growth and radiologic leptomeningeal dissemination. Recurrent 1p deletion and MAPK/ERK pathway activation represent diagnostic biomarkers and therapeutic targets, respectively-laying the foundation for future clinical trials with, e.g., MEK inhibitors that may improve the clinical outcome of patients with DLGNT.


Subject(s)
Meningeal Neoplasms/classification , Meningeal Neoplasms/genetics , Oligodendroglioma/classification , Oligodendroglioma/genetics , Adolescent , Adult , Central Nervous System Neoplasms/diagnostic imaging , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/pathology , Child , Child, Preschool , DNA Copy Number Variations/genetics , DNA Methylation/genetics , Female , Genetic Testing , Humans , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Male , Meningeal Neoplasms/diagnostic imaging , Meningeal Neoplasms/pathology , Middle Aged , Mitogen-Activated Protein Kinase Kinases/metabolism , Oligodendroglioma/diagnostic imaging , Oligodendroglioma/pathology , Signal Transduction/genetics , Transcriptome , Young Adult
11.
Oncotarget ; 7(38): 61860-61873, 2016 Sep 20.
Article in English | MEDLINE | ID: mdl-27556362

ABSTRACT

Ependymomas in children can arise throughout all compartments of the central nervous system (CNS). Highly malignant paediatric ependymoma subtypes are Group A tumours of the posterior fossa (PF-EPN-A) and RELA-fusion positive (ST-EPN-RELA) tumours in the supratentorial compartment. It was repeatedly reported in smaller series that accumulation of p53 is frequently observed in ependymomas and that immunohistochemical staining correlates with poor clinical outcome, while TP53 mutations are rare. Our TP53 mutation analysis of 130 primary ependymomas identified a mutation rate of only 3%. Immunohistochemical analysis of 398 ependymomas confirmed previous results correlating the accumulation of p53 with inferior outcome. Among the p53-positive ependymomas, the vast majority exhibited a RELA fusion leading to the hypothesis that p53 inactivation might be linked to RELA positivity.In order to assess the potential of p53 reactivation through MDM2 inhibition in ependymoma, we evaluated the effects of Actinomycin-D and Nutlin-3 treatment in two preclinical ependymoma models representing the high-risk subtypes PF-EPN-A and ST-EPN-RELA. The IC-50 of the agent as determined by metabolic activity assays was in the lower nano-molar range (0.2-0.7 nM). Transcriptome analyses of high-dose (100 nM), low-dose (5 nM) and non-treated cells revealed re-expression of p53 dependent genes including p53 upregulated modulator of apoptosis (PUMA) after low-dose treatment. At the protein level, we validated the Actinomycin-D induced upregulation of PUMA, and of p53 interaction partners MDM2 and p21. Proapoptotic effects of low-dose application of the agent were confirmed by flow cytometry. Thus, Actinomycin-D could constitute a promising therapeutic option for ST-EPN-RELA ependymoma patients, whose tumours frequently exhibit p53 inactivation.


Subject(s)
Dactinomycin/therapeutic use , Ependymoma/drug therapy , Transcription Factor RelA/genetics , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA Mutational Analysis , Ependymoma/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Imidazoles/metabolism , Immunohistochemistry , Medulloblastoma/metabolism , Mice , Neurons/metabolism , Piperazines/metabolism , Protein Synthesis Inhibitors/therapeutic use , Proto-Oncogene Proteins c-mdm2/genetics , Stem Cells/metabolism , Treatment Outcome
12.
Cancer Cell ; 29(3): 379-393, 2016 Mar 14.
Article in English | MEDLINE | ID: mdl-26923874

ABSTRACT

Atypical teratoid/rhabdoid tumor (ATRT) is one of the most common brain tumors in infants. Although the prognosis of ATRT patients is poor, some patients respond favorably to current treatments, suggesting molecular inter-tumor heterogeneity. To investigate this further, we genetically and epigenetically analyzed 192 ATRTs. Three distinct molecular subgroups of ATRTs, associated with differences in demographics, tumor location, and type of SMARCB1 alterations, were identified. Whole-genome DNA and RNA sequencing found no recurrent mutations in addition to SMARCB1 that would explain the differences between subgroups. Whole-genome bisulfite sequencing and H3K27Ac chromatin-immunoprecipitation sequencing of primary tumors, however, revealed clear differences, leading to the identification of subgroup-specific regulatory networks and potential therapeutic targets.


Subject(s)
Epigenesis, Genetic/genetics , Rhabdoid Tumor/genetics , Teratoma/genetics , Brain Neoplasms/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Humans , Mutation/genetics , SMARCB1 Protein , Transcription Factors/genetics
13.
Cancer Cell ; 27(5): 728-43, 2015 May 11.
Article in English | MEDLINE | ID: mdl-25965575

ABSTRACT

Ependymal tumors across age groups are currently classified and graded solely by histopathology. It is, however, commonly accepted that this classification scheme has limited clinical utility based on its lack of reproducibility in predicting patients' outcome. We aimed at establishing a uniform molecular classification using DNA methylation profiling. Nine molecular subgroups were identified in a large cohort of 500 tumors, 3 in each anatomical compartment of the CNS, spine, posterior fossa, supratentorial. Two supratentorial subgroups are characterized by prototypic fusion genes involving RELA and YAP1, respectively. Regarding clinical associations, the molecular classification proposed herein outperforms the current histopathological classification and thus might serve as a basis for the next World Health Organization classification of CNS tumors.


Subject(s)
Age Factors , Central Nervous System Neoplasms/pathology , Ependymoma/pathology , Adaptor Proteins, Signal Transducing/genetics , Adolescent , Adult , Aged , Central Nervous System Neoplasms/classification , Central Nervous System Neoplasms/genetics , Child , Child, Preschool , DNA Methylation , Ependymoma/classification , Ependymoma/genetics , Female , Gene Dosage , Gene Expression Profiling , Gene Fusion , Humans , Infant , Male , Middle Aged , Phosphoproteins/genetics , Transcription Factors , Transcription, Genetic , YAP-Signaling Proteins , Young Adult
14.
Nature ; 511(7510): 428-34, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25043047

ABSTRACT

Medulloblastoma is a highly malignant paediatric brain tumour currently treated with a combination of surgery, radiation and chemotherapy, posing a considerable burden of toxicity to the developing child. Genomics has illuminated the extensive intertumoral heterogeneity of medulloblastoma, identifying four distinct molecular subgroups. Group 3 and group 4 subgroup medulloblastomas account for most paediatric cases; yet, oncogenic drivers for these subtypes remain largely unidentified. Here we describe a series of prevalent, highly disparate genomic structural variants, restricted to groups 3 and 4, resulting in specific and mutually exclusive activation of the growth factor independent 1 family proto-oncogenes, GFI1 and GFI1B. Somatic structural variants juxtapose GFI1 or GFI1B coding sequences proximal to active enhancer elements, including super-enhancers, instigating oncogenic activity. Our results, supported by evidence from mouse models, identify GFI1 and GFI1B as prominent medulloblastoma oncogenes and implicate 'enhancer hijacking' as an efficient mechanism driving oncogene activation in a childhood cancer.


Subject(s)
DNA-Binding Proteins/genetics , Enhancer Elements, Genetic/genetics , Genomic Structural Variation/genetics , Medulloblastoma/genetics , Oncogenes/genetics , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Animals , Child , Chromosomes, Human, Pair 9/genetics , DNA-Binding Proteins/metabolism , Humans , Medulloblastoma/classification , Medulloblastoma/pathology , Mice , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism
15.
Cancer Cell ; 25(3): 393-405, 2014 Mar 17.
Article in English | MEDLINE | ID: mdl-24651015

ABSTRACT

Smoothened (SMO) inhibitors recently entered clinical trials for sonic-hedgehog-driven medulloblastoma (SHH-MB). Clinical response is highly variable. To understand the mechanism(s) of primary resistance and identify pathways cooperating with aberrant SHH signaling, we sequenced and profiled a large cohort of SHH-MBs (n = 133). SHH pathway mutations involved PTCH1 (across all age groups), SUFU (infants, including germline), and SMO (adults). Children >3 years old harbored an excess of downstream MYCN and GLI2 amplifications and frequent TP53 mutations, often in the germline, all of which were rare in infants and adults. Functional assays in different SHH-MB xenograft models demonstrated that SHH-MBs harboring a PTCH1 mutation were responsive to SMO inhibition, whereas tumors harboring an SUFU mutation or MYCN amplification were primarily resistant.


Subject(s)
Drug Resistance, Neoplasm/genetics , Hedgehog Proteins/genetics , Medulloblastoma/genetics , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Adolescent , Adult , Animals , Base Sequence , Biphenyl Compounds/therapeutic use , Cerebellar Neoplasms/drug therapy , Cerebellar Neoplasms/genetics , Child , Child, Preschool , DEAD-box RNA Helicases/genetics , DNA Copy Number Variations/genetics , Female , Gene Expression Profiling , High-Throughput Nucleotide Sequencing , Humans , Infant , Kruppel-Like Transcription Factors/genetics , Male , Medulloblastoma/drug therapy , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Sequence Data , N-Myc Proto-Oncogene Protein , Neoplasm Transplantation , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Patched Receptors , Patched-1 Receptor , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyridines/therapeutic use , Receptors, Cell Surface/genetics , Repressor Proteins/genetics , Signal Transduction/genetics , Smoothened Receptor , Telomerase/genetics , Tumor Suppressor Protein p53/genetics , Young Adult , Zinc Finger Protein Gli2
16.
Nature ; 488(7409): 100-5, 2012 Aug 02.
Article in English | MEDLINE | ID: mdl-22832583

ABSTRACT

Medulloblastoma is an aggressively growing tumour, arising in the cerebellum or medulla/brain stem. It is the most common malignant brain tumour in children, and shows tremendous biological and clinical heterogeneity. Despite recent treatment advances, approximately 40% of children experience tumour recurrence, and 30% will die from their disease. Those who survive often have a significantly reduced quality of life. Four tumour subgroups with distinct clinical, biological and genetic profiles are currently identified. WNT tumours, showing activated wingless pathway signalling, carry a favourable prognosis under current treatment regimens. SHH tumours show hedgehog pathway activation, and have an intermediate prognosis. Group 3 and 4 tumours are molecularly less well characterized, and also present the greatest clinical challenges. The full repertoire of genetic events driving this distinction, however, remains unclear. Here we describe an integrative deep-sequencing analysis of 125 tumour-normal pairs, conducted as part of the International Cancer Genome Consortium (ICGC) PedBrain Tumor Project. Tetraploidy was identified as a frequent early event in Group 3 and 4 tumours, and a positive correlation between patient age and mutation rate was observed. Several recurrent mutations were identified, both in known medulloblastoma-related genes (CTNNB1, PTCH1, MLL2, SMARCA4) and in genes not previously linked to this tumour (DDX3X, CTDNEP1, KDM6A, TBR1), often in subgroup-specific patterns. RNA sequencing confirmed these alterations, and revealed the expression of what are, to our knowledge, the first medulloblastoma fusion genes identified. Chromatin modifiers were frequently altered across all subgroups. These findings enhance our understanding of the genomic complexity and heterogeneity underlying medulloblastoma, and provide several potential targets for new therapeutics, especially for Group 3 and 4 patients.


Subject(s)
Cerebellar Neoplasms/genetics , Genome, Human/genetics , Medulloblastoma/genetics , Aging/genetics , Amino Acid Sequence , Cell Transformation, Neoplastic , Cerebellar Neoplasms/classification , Cerebellar Neoplasms/diagnosis , Cerebellar Neoplasms/pathology , Child , Chromatin/metabolism , Chromosomes, Human/genetics , DEAD-box RNA Helicases/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Genomics , Hedgehog Proteins/metabolism , High-Throughput Nucleotide Sequencing , Histone Demethylases/genetics , Humans , Medulloblastoma/classification , Medulloblastoma/diagnosis , Medulloblastoma/pathology , Methylation , Mutation/genetics , Mutation Rate , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Oncogene Proteins, Fusion/genetics , Patched Receptors , Patched-1 Receptor , Phosphoprotein Phosphatases/genetics , Polyploidy , Receptors, Cell Surface/genetics , Sequence Analysis, RNA , Signal Transduction , T-Box Domain Proteins/genetics , Transcription Factors/genetics , Wnt Proteins/metabolism , beta Catenin/genetics
17.
Acta Neuropathol ; 123(4): 515-27, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22160402

ABSTRACT

Focal high-level amplifications of MYC (or MYCC) define a subset of high-risk medulloblastoma patients. However, the prognostic role of MYCN oncogene amplification remains unresolved. We aimed to evaluate the prognostic value of this alteration alone and in combination with biological modifiers in 67 pediatric medulloblastomas with MYCN amplification (MYCN-MB). Twenty-one MYCN-MB were examined using gene expression profiling and array-CGH, whereas for 46 tumors immunohistochemical analysis and FISH were performed. All 67 tumors were further subjected to mutational analyses. We compared molecular, clinical, and prognostic characteristics both within biological MYCN-MB groups and with non-amplified tumors. Transcriptomic analysis revealed SHH-driven tumorigenesis in a subset of MYCN-MBs indicating a biological dichotomy of MYCN-MB. Activation of SHH was accompanied by variant-specific cytogenetic aberrations including deletion of 9q in SHH tumors. Non-SHH MB were associated with gain of 7q and isochromosome 17q/17q gain. Among clinically relevant variables, SHH subtype and 10q loss for non-SHH tumors comprised the most powerful markers of favorable prognosis in MYCN-MB. In conclusion, we demonstrate considerable heterogeneity within MYCN-MB in terms of genetics, tumor biology, and clinical outcome. Thus, assessment of disease group and 10q copy-number status may improve risk stratification of this group and may delineate MYCN-MB with the same dismal prognosis as MYC amplified tumors. Furthermore, based on the enrichment of MYCN and GLI2 amplifications in SHH-driven medulloblastoma, amplification of these downstream signaling intermediates should be taken into account before a patient is enrolled into a clinical trial using a smoothened inhibitor.


Subject(s)
Biomarkers, Tumor/genetics , Cerebellar Neoplasms/genetics , DNA Copy Number Variations/genetics , Medulloblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Cerebellar Neoplasms/mortality , Child, Preschool , Chromosome Aberrations , Chromosomes, Human, Pair 10 , Cluster Analysis , Computational Biology , DNA Mutational Analysis , Female , Gene Expression Profiling , Gene Regulatory Networks , Hedgehog Proteins/genetics , Humans , Longitudinal Studies , Male , Medulloblastoma/mortality , Microarray Analysis , N-Myc Proto-Oncogene Protein , Retrospective Studies , Risk Factors , Survival Analysis , Tumor Suppressor Protein p53/genetics , beta Catenin/metabolism
18.
J Clin Oncol ; 29(29): 3852-61, 2011 Oct 10.
Article in English | MEDLINE | ID: mdl-21911727

ABSTRACT

PURPOSE: Integrated genomics approaches have revealed at least four distinct biologic variants of medulloblastoma: WNT (wingless), SHH (sonic hedgehog), group C, and group D. Because of the remarkable clinical heterogeneity of group D tumors and the dismal prognosis of group C patients, it is vital to identify molecular biomarkers that will allow early and effective treatment stratification in these non-WNT/non-SHH tumors. PATIENTS AND METHODS: We combined transcriptome and DNA copy-number analyses for 64 primary medulloblastomas. Bioinformatic tools were used to discover marker genes of molecular variants. Differentially expressed transcripts were evaluated for prognostic value in the screening cohort. The prognostic power of follistatin-like 5 (FSTL5) immunopositivity was tested for 235 nonoverlapping medulloblastoma samples on two independent tissue microarrays. RESULTS: Comprehensive analyses of transcriptomic and genetic alterations delineate four distinct variants of medulloblastoma. Stable subgroup separation was achieved by using the 300 transcripts that varied the most. Distinct expression patterns of FSTL5 in each molecular subgroup were confirmed by quantitative real-time polymerase chain reaction. Immunopositivity of FSTL5 identified a large cohort of patients (84 of 235 patients; 36%) at high risk for relapse and death. Importantly, more than 50% of non-WNT/non-SHH tumors displayed FSTL5 negativity, delineating a large patient cohort with a good prognosis who would otherwise be considered intermediate or high-risk on the basis of current molecular subgrouping. CONCLUSION: FSTL5 expression denoted a dismal prognosis both within and across medulloblastoma subgroups. The addition of FSTL5 immunohistochemistry to existing molecular stratification schemes constitutes a reliable and cost-effective tool for prognostication in future clinical trials of medulloblastoma.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Follistatin-Related Proteins/genetics , Medulloblastoma/genetics , Medulloblastoma/pathology , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/genetics , Brain Neoplasms/metabolism , Child , Cohort Studies , Disease-Free Survival , Follistatin-Related Proteins/biosynthesis , Gene Expression Profiling , Humans , Kaplan-Meier Estimate , Medulloblastoma/metabolism , Microarray Analysis , Prognosis , Real-Time Polymerase Chain Reaction , Survival Analysis
19.
Cancer Cell ; 20(2): 143-57, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21840481

ABSTRACT

Despite the histological similarity of ependymomas from throughout the neuroaxis, the disease likely comprises multiple independent entities, each with a distinct molecular pathogenesis. Transcriptional profiling of two large independent cohorts of ependymoma reveals the existence of two demographically, transcriptionally, genetically, and clinically distinct groups of posterior fossa (PF) ependymomas. Group A patients are younger, have laterally located tumors with a balanced genome, and are much more likely to exhibit recurrence, metastasis at recurrence, and death compared with Group B patients. Identification and optimization of immunohistochemical (IHC) markers for PF ependymoma subgroups allowed validation of our findings on a third independent cohort, using a human ependymoma tissue microarray, and provides a tool for prospective prognostication and stratification of PF ependymoma patients.


Subject(s)
Brain Neoplasms/classification , Cranial Fossa, Posterior/pathology , Ependymoma/classification , Adult , Brain Neoplasms/genetics , Chromosome Aberrations , Ependymoma/genetics , Female , Gene Expression Profiling , Humans , Male , Middle Aged
20.
J Clin Oncol ; 29(19): 2717-23, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21632505

ABSTRACT

PURPOSE: Medulloblastoma is a rare primary brain tumor in adults, whereas it constitutes the most common malignant brain tumor in children. Integrated genomics approaches revealed at least four distinct disease variants in children. The aim of this study was to investigate molecular subtypes and their prognostic implication in a large cohort of adult medulloblastomas as the biology in this age group remains poorly understood. PATIENTS AND METHODS: We combined transcriptome and DNA copy number analyses for 28 adult medulloblastomas. Statistical and bioinformatic tools were applied to discover distinct molecular variants. Clinical and molecular characteristics of each biologic subtype were validated using immunohistochemistry on a tissue microarray derived from an independent patient cohort of adult medulloblastomas (n = 103). RESULTS: Gene expression profiles revealed three distinct molecular variants with stable subtype separation using the 300 most varying transcripts. Distinct demographics, genetics, transcriptome, and prognosis were noted for each subtype of adult medulloblastoma. Immunohistochemistry revealed aberrant activation of the sonic hedgehog (SHH) pathway in over half of adult medulloblastomas constituting a promising molecular therapeutic target. In contrast, subtype C tumors, which comprise a robust subtype in childhood medulloblastoma are only exceptionally seen in adult cohorts. Notably, adult subtype D and Wnt/wingless tumors were associated with worse prognosis than pediatric cohorts, whereas survival for SHH tumors was similar for both age groups. CONCLUSION: The transcriptome of adult medulloblastomas differs considerably from pediatric counterparts, both in terms of tumor biology and prognostic impact. Therefore, age-specific classification is required and must be adapted for use in clinical trials of adult medulloblastoma.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Medulloblastoma/diagnosis , Medulloblastoma/genetics , Adolescent , Adult , Brain Neoplasms/classification , Cohort Studies , Computational Biology/methods , Female , Gene Dosage , Gene Expression Profiling , Hedgehog Proteins/metabolism , Humans , Immunohistochemistry/methods , In Situ Hybridization, Fluorescence , Male , Medulloblastoma/classification , Middle Aged , Oligonucleotide Array Sequence Analysis , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...