Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Cell ; 164(5): 1060-1072, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26919435

ABSTRACT

Primitive neuroectodermal tumors of the central nervous system (CNS-PNETs) are highly aggressive, poorly differentiated embryonal tumors occurring predominantly in young children but also affecting adolescents and adults. Herein, we demonstrate that a significant proportion of institutionally diagnosed CNS-PNETs display molecular profiles indistinguishable from those of various other well-defined CNS tumor entities, facilitating diagnosis and appropriate therapy for patients with these tumors. From the remaining fraction of CNS-PNETs, we identify four new CNS tumor entities, each associated with a recurrent genetic alteration and distinct histopathological and clinical features. These new molecular entities, designated "CNS neuroblastoma with FOXR2 activation (CNS NB-FOXR2)," "CNS Ewing sarcoma family tumor with CIC alteration (CNS EFT-CIC)," "CNS high-grade neuroepithelial tumor with MN1 alteration (CNS HGNET-MN1)," and "CNS high-grade neuroepithelial tumor with BCOR alteration (CNS HGNET-BCOR)," will enable meaningful clinical trials and the development of therapeutic strategies for patients affected by poorly differentiated CNS tumors.


Subject(s)
Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/pathology , DNA Methylation , Neuroectodermal Tumors/genetics , Neuroectodermal Tumors/pathology , Amino Acid Sequence , Central Nervous System Neoplasms/classification , Central Nervous System Neoplasms/diagnosis , Child , Forkhead Transcription Factors/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Molecular Sequence Data , Neuroectodermal Tumors/classification , Neuroectodermal Tumors/diagnosis , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Repressor Proteins/chemistry , Repressor Proteins/genetics , Signal Transduction , Trans-Activators , Tumor Suppressor Proteins/genetics
2.
Cell ; 148(1-2): 59-71, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22265402

ABSTRACT

Genomic rearrangements are thought to occur progressively during tumor development. Recent findings, however, suggest an alternative mechanism, involving massive chromosome rearrangements in a one-step catastrophic event termed chromothripsis. We report the whole-genome sequencing-based analysis of a Sonic-Hedgehog medulloblastoma (SHH-MB) brain tumor from a patient with a germline TP53 mutation (Li-Fraumeni syndrome), uncovering massive, complex chromosome rearrangements. Integrating TP53 status with microarray and deep sequencing-based DNA rearrangement data in additional patients reveals a striking association between TP53 mutation and chromothripsis in SHH-MBs. Analysis of additional tumor entities substantiates a link between TP53 mutation and chromothripsis, and indicates a context-specific role for p53 in catastrophic DNA rearrangements. Among these, we observed a strong association between somatic TP53 mutations and chromothripsis in acute myeloid leukemia. These findings connect p53 status and chromothripsis in specific tumor types, providing a genetic basis for understanding particularly aggressive subtypes of cancer.


Subject(s)
Brain Neoplasms/genetics , Gene Rearrangement , Medulloblastoma/genetics , Tumor Suppressor Protein p53/genetics , Animals , Child , Chromosome Aberrations , DNA Copy Number Variations , DNA Mutational Analysis , Disease Models, Animal , Humans , Leukemia, Myeloid, Acute/genetics , Li-Fraumeni Syndrome/physiopathology , Mice , Middle Aged
3.
Nature ; 555(7697): 469-474, 2018 03 22.
Article in English | MEDLINE | ID: mdl-29539639

ABSTRACT

Accurate pathological diagnosis is crucial for optimal management of patients with cancer. For the approximately 100 known tumour types of the central nervous system, standardization of the diagnostic process has been shown to be particularly challenging-with substantial inter-observer variability in the histopathological diagnosis of many tumour types. Here we present a comprehensive approach for the DNA methylation-based classification of central nervous system tumours across all entities and age groups, and demonstrate its application in a routine diagnostic setting. We show that the availability of this method may have a substantial impact on diagnostic precision compared to standard methods, resulting in a change of diagnosis in up to 12% of prospective cases. For broader accessibility, we have designed a free online classifier tool, the use of which does not require any additional onsite data processing. Our results provide a blueprint for the generation of machine-learning-based tumour classifiers across other cancer entities, with the potential to fundamentally transform tumour pathology.


Subject(s)
Central Nervous System Neoplasms/diagnosis , Central Nervous System Neoplasms/genetics , DNA Methylation , Adolescent , Adult , Aged , Aged, 80 and over , Central Nervous System Neoplasms/classification , Central Nervous System Neoplasms/pathology , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Male , Middle Aged , Reproducibility of Results , Unsupervised Machine Learning , Young Adult
4.
Nature ; 510(7506): 537-41, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24847876

ABSTRACT

Epigenetic alterations, that is, disruption of DNA methylation and chromatin architecture, are now acknowledged as a universal feature of tumorigenesis. Medulloblastoma, a clinically challenging, malignant childhood brain tumour, is no exception. Despite much progress from recent genomics studies, with recurrent changes identified in each of the four distinct tumour subgroups (WNT-pathway-activated, SHH-pathway-activated, and the less-well-characterized Group 3 and Group 4), many cases still lack an obvious genetic driver. Here we present whole-genome bisulphite-sequencing data from thirty-four human and five murine tumours plus eight human and three murine normal controls, augmented with matched whole-genome, RNA and chromatin immunoprecipitation sequencing data. This comprehensive data set allowed us to decipher several features underlying the interplay between the genome, epigenome and transcriptome, and its effects on medulloblastoma pathophysiology. Most notable were highly prevalent regions of hypomethylation correlating with increased gene expression, extending tens of kilobases downstream of transcription start sites. Focal regions of low methylation linked to transcription-factor-binding sites shed light on differential transcriptional networks between subgroups, whereas increased methylation due to re-normalization of repressed chromatin in DNA methylation valleys was positively correlated with gene expression. Large, partially methylated domains affecting up to one-third of the genome showed increased mutation rates and gene silencing in a subgroup-specific fashion. Epigenetic alterations also affected novel medulloblastoma candidate genes (for example, LIN28B), resulting in alternative promoter usage and/or differential messenger RNA/microRNA expression. Analysis of mouse medulloblastoma and precursor-cell methylation demonstrated a somatic origin for many alterations. Our data provide insights into the epigenetic regulation of transcription and genome organization in medulloblastoma pathogenesis, which are probably also of importance in a wider developmental and disease context.


Subject(s)
DNA Methylation/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Medulloblastoma/genetics , Sequence Analysis, DNA/methods , Animals , Binding Sites , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Chromatin Immunoprecipitation , Female , Genome/genetics , Histones/metabolism , Humans , Medulloblastoma/pathology , Mice , Promoter Regions, Genetic/genetics , RNA-Binding Proteins/genetics , Transcription Factors/metabolism , Transcription, Genetic
5.
BMC Pediatr ; 20(1): 48, 2020 01 31.
Article in English | MEDLINE | ID: mdl-32005112

ABSTRACT

BACKGROUND: About 2000 children and adolescents under the age of 18 are diagnosed with cancer each year in Germany. Because of current medical treatment methods, a high survival rate can be reached for many types of the disease. Nevertheless, patients face a number of long-term effects related to the treatment. As a result, physical and psychological consequences have increasingly become the focus of research in recent years. Social dimensions of health have received little attention in health services research in oncology so far. Yet, there are no robust results that allow an estimation of whether and to what extent the disease and treatment impair the participation of children and adolescents and which factors mediate this effect. Social participation is of great importance especially because interactions with peers and experiences in different areas of life are essential for the development of children and adolescents. METHODS: Data are collected in a longitudinal, prospective, observational multicenter study. For this purpose, all patients and their parents who are being treated for cancer in one of the participating clinics throughout Germany will be interviewed within the first month after diagnosis (t1), after completion of intensive treatment (t2) and half a year after the end of intensive treatment (t3) using standardized questionnaires. Analysis will be done by descriptive and multivariate methods. DISCUSSION: The results can be used to identify children and adolescents in high-risk situations at an early stage in order to be able to initiate interventions tailored to the needs. Such tailored interventions will finally reduce the risk of impairments in the participation of children and adolescents and increase quality of life. TRIAL REGISTRATION: ClinicalTrials.gov: NCT04101123.


Subject(s)
Brain Neoplasms , Leukemia , Sarcoma , Adolescent , Child , Germany , Humans , Multicenter Studies as Topic , Observational Studies as Topic , Prospective Studies , Quality of Life , Socioeconomic Factors
6.
Nature ; 482(7384): 226-31, 2012 Jan 29.
Article in English | MEDLINE | ID: mdl-22286061

ABSTRACT

Glioblastoma multiforme (GBM) is a lethal brain tumour in adults and children. However, DNA copy number and gene expression signatures indicate differences between adult and paediatric cases. To explore the genetic events underlying this distinction, we sequenced the exomes of 48 paediatric GBM samples. Somatic mutations in the H3.3-ATRX-DAXX chromatin remodelling pathway were identified in 44% of tumours (21/48). Recurrent mutations in H3F3A, which encodes the replication-independent histone 3 variant H3.3, were observed in 31% of tumours, and led to amino acid substitutions at two critical positions within the histone tail (K27M, G34R/G34V) involved in key regulatory post-translational modifications. Mutations in ATRX (α-thalassaemia/mental retardation syndrome X-linked) and DAXX (death-domain associated protein), encoding two subunits of a chromatin remodelling complex required for H3.3 incorporation at pericentric heterochromatin and telomeres, were identified in 31% of samples overall, and in 100% of tumours harbouring a G34R or G34V H3.3 mutation. Somatic TP53 mutations were identified in 54% of all cases, and in 86% of samples with H3F3A and/or ATRX mutations. Screening of a large cohort of gliomas of various grades and histologies (n = 784) showed H3F3A mutations to be specific to GBM and highly prevalent in children and young adults. Furthermore, the presence of H3F3A/ATRX-DAXX/TP53 mutations was strongly associated with alternative lengthening of telomeres and specific gene expression profiles. This is, to our knowledge, the first report to highlight recurrent mutations in a regulatory histone in humans, and our data suggest that defects of the chromatin architecture underlie paediatric and young adult GBM pathogenesis.


Subject(s)
Chromatin Assembly and Disassembly/genetics , Chromatin/genetics , Glioblastoma/genetics , Histones/genetics , Mutation/genetics , Adaptor Proteins, Signal Transducing/genetics , Base Sequence , Child , Chromatin/metabolism , Co-Repressor Proteins , DNA Helicases/genetics , DNA Mutational Analysis , Exome/genetics , Gene Expression Profiling , Histones/metabolism , Humans , Molecular Chaperones , Molecular Sequence Data , Nuclear Proteins/genetics , Telomere/genetics , Tumor Suppressor Protein p53/genetics , X-linked Nuclear Protein
7.
Neuro Oncol ; 25(12): 2273-2286, 2023 12 08.
Article in English | MEDLINE | ID: mdl-37379234

ABSTRACT

BACKGROUND: The prognosis for Li-Fraumeni syndrome (LFS) patients with medulloblastoma (MB) is poor. Comprehensive clinical data for this patient group is lacking, challenging the development of novel therapeutic strategies. Here, we present clinical and molecular data on a retrospective cohort of pediatric LFS MB patients. METHODS: In this multinational, multicenter retrospective cohort study, LFS patients under 21 years with MB and class 5 or class 4 constitutional TP53 variants were included. TP53 mutation status, methylation subgroup, treatment, progression free- (PFS) and overall survival (OS), recurrence patterns, and incidence of subsequent neoplasms were evaluated. RESULTS: The study evaluated 47 LFS individuals diagnosed with MB, mainly classified as DNA methylation subgroup "SHH_3" (86%). The majority (74%) of constitutional TP53 variants represented missense variants. The 2- and 5-year (y-) PFS were 36% and 20%, and 2- and 5y-OS were 53% and 23%, respectively. Patients who received postoperative radiotherapy (RT) (2y-PFS: 44%, 2y-OS: 60%) or chemotherapy before RT (2y-PFS: 32%, 2y-OS: 48%) had significantly better clinical outcome then patients who were not treated with RT (2y-PFS: 0%, 2y-OS: 25%). Patients treated according to protocols including high-intensity chemotherapy and patients who received only maintenance-type chemotherapy showed similar outcomes (2y-PFS: 42% and 35%, 2y-OS: 68% and 53%, respectively). CONCLUSIONS: LFS MB patients have a dismal prognosis. In the presented cohort use of RT significantly increased survival rates, whereas chemotherapy intensity did not influence their clinical outcome. Prospective collection of clinical data and development of novel treatments are required to improve the outcome of LFS MB patients.


Subject(s)
Cerebellar Neoplasms , Li-Fraumeni Syndrome , Medulloblastoma , Child , Humans , Li-Fraumeni Syndrome/complications , Li-Fraumeni Syndrome/genetics , Li-Fraumeni Syndrome/therapy , Medulloblastoma/therapy , Medulloblastoma/drug therapy , Retrospective Studies , Prospective Studies , Cerebellar Neoplasms/therapy , Cerebellar Neoplasms/drug therapy , Germ-Line Mutation , Tumor Suppressor Protein p53/genetics
8.
Blood Cells Mol Dis ; 48(2): 128-31, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22178060

ABSTRACT

More than 90% of Fanconi anemia (FA) patients experience progressive bone marrow failure during life with a median onset at 8 years of age. As matched sibling donor transplantation as preferred treatment is not available for the majority of patients, several synthetic androgens have been used as short-term treatment options for the marrow failure in FA patients for more than 50 years. Here, we retrospectively collected data on eight FA patients who received danazol for the off-label treatment of their marrow failure at a starting dose of approximately 5mg/kg body weight/die. The hematological parameters at the initiation of treatment were hemoglobin (Hb) <8 g/dL and/or thrombocytes <30,000/µl. In 7 out of 8 FA patients, the values for both parameters rose on average >50% over the starting counts within 6 months and remained stable for up to 3 years despite careful reduction of the danazol dose per kg body weight. In 4 patients with a follow-up of 3 years, the platelets finally reached an average of 68,000/µL or 2.8 times over the starting values, while the Hb remained stable >11 g/dL. Danazol was reduced to 54% of the starting dose or 2.6 mg/kg/die. One FA-A patient with an unusually severe phenotype did not response with her PB counts to either danazol or oxymethalone within 6 months. None of the patients developed severe or unacceptable side-effects from the danazol treatment that led to the discontinuation of therapy. This initial description suggests that danazol might be an effective and well-tolerated treatment option for delaying the progressive marrow failure in FA patients for at least 3 years and longer.


Subject(s)
Bone Marrow/pathology , Danazol/therapeutic use , Estrogen Antagonists/therapeutic use , Fanconi Anemia/drug therapy , Adolescent , Adult , Blood Cell Count , Child , Child, Preschool , Fanconi Anemia/blood , Fanconi Anemia/genetics , Female , Humans , Male , Mutation , Young Adult
9.
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
10.
Pediatr Blood Cancer ; 58(4): 539-44, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21922643

ABSTRACT

PURPOSE: Overall survival is poor in children with primary unresectable hepatocellular carcinoma. Sorafenib has been shown to significantly improve progression-free survival in adult hepatocellular carcinoma (HCC) patients. We evaluated the experience of PLADO (cisplatin 80 mg/m(2) /day, doxorubicin 2 × 30 mg/m(2) /day) in combination with sorafenib in pediatric HCC patients. PATIENTS AND METHODS: Clinical data of 12 patients (7-16 years), 7 with unresectable tumor, were retrospectively assessed. RESULTS: In total 6/12 (50%) patients are in complete remission after a median follow-up of 20 months (4 with PLADO/sorafenib/resection, 2 with liver transplantation after local relapse). Of the seven patients with unresectable tumor, PLADO/sorafenib resulted in partial response (PR) in four, stable disease (SD) in two, and progression in one. Three are alive in CR after complete resection after 12 (alternative therapy after two cycles PLADO/sorafenib), 12 and 18 months (six cycles PLADO/sorafenib), respectively. All four patients with elevated alpha-fetoprotein levels had a marked drop after two cycles. Of the five patients with primary complete tumor resection one is alive disease-free at 27 months. Four had local or metastatic relapses (13, 7, 12, and 13 months), two of whom were rescued by liver transplantation (CR after 25 and 32 months). The main toxicity attributable to sorafenib was a hand-foot skin reaction (HFSR) in seven patients. CONCLUSION: Sorafenib in combination with PLADO may be a promising approach in pediatric HCC; HFSR was the most important toxicity. Data based on prospective studies are needed to evaluate pharmacokinetics, resectability rates, and survival in pediatric HCC treated with sorafenib.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Benzenesulfonates/administration & dosage , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/therapy , Liver Transplantation , Protein Kinase Inhibitors/administration & dosage , Pyridines/administration & dosage , Adolescent , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzenesulfonates/adverse effects , Carcinoma, Hepatocellular/mortality , Child , Cisplatin/administration & dosage , Cisplatin/adverse effects , Disease-Free Survival , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Female , Follow-Up Studies , Humans , Liver Neoplasms/mortality , Male , Niacinamide/analogs & derivatives , Phenylurea Compounds , Prospective Studies , Protein Kinase Inhibitors/adverse effects , Pyridines/adverse effects , Retrospective Studies , Sorafenib , Survival Rate , Transplantation, Homologous
11.
J Clin Invest ; 118(5): 1739-49, 2008 May.
Article in English | MEDLINE | ID: mdl-18398503

ABSTRACT

The molecular pathogenesis of pediatric astrocytomas is still poorly understood. To further understand the genetic abnormalities associated with these tumors, we performed a genome-wide analysis of DNA copy number aberrations in pediatric low-grade astrocytomas by using array-based comparative genomic hybridization. Duplication of the BRAF protooncogene was the most frequent genomic aberration, and tumors with BRAF duplication showed significantly increased mRNA levels of BRAF and a downstream target, CCND1, as compared with tumors without duplication. Furthermore, denaturing HPLC showed that activating BRAF mutations were detected in some of the tumors without BRAF duplication. Similarly, a marked proportion of low-grade astrocytomas from adult patients also had BRAF duplication. Both the stable silencing of BRAF through shRNA lentiviral transduction and pharmacological inhibition of MEK1/2, the immediate downstream phosphorylation target of BRAF, blocked the proliferation and arrested the growth of cultured tumor cells derived from low-grade gliomas. Our findings implicate aberrant activation of the MAPK pathway due to gene duplication or mutation of BRAF as a molecular mechanism of pathogenesis in low-grade astrocytomas and suggest inhibition of the MAPK pathway as a potential treatment.


Subject(s)
Astrocytoma/enzymology , Astrocytoma/genetics , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Gene Duplication , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Astrocytoma/pathology , Brain Neoplasms/pathology , Cell Cycle/physiology , Child , Chromosome Aberrations , Cyclin D , Cyclins/genetics , Cyclins/metabolism , Enzyme Activation , Enzyme Inhibitors/metabolism , Female , Humans , Male , Microarray Analysis , Mitogen-Activated Protein Kinases/genetics , Mutation , Nucleic Acid Hybridization/methods , Proto-Oncogene Proteins B-raf/genetics
12.
Acta Neuropathol ; 121(6): 763-74, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21424530

ABSTRACT

Activation of the MAPK signaling pathway has been shown to be a unifying molecular feature in pilocytic astrocytoma (PA). Genetically, tandem duplications at chromosome 7q34 resulting in KIAA1549-BRAF fusion genes constitute the most common mechanism identified to date. To elucidate alternative mechanisms of aberrant MAPK activation in PA, we screened 125 primary tumors for RAF fusion genes and mutations in KRAS, NRAS, HRAS, PTPN11, BRAF and RAF1. Using microarray-based comparative genomic hybridization (aCGH), we identified in three cases an interstitial deletion of ~2.5 Mb as a novel recurrent mechanism forming BRAF gene fusions with FAM131B, a currently uncharacterized gene on chromosome 7q34. This deletion removes the BRAF N-terminal inhibitory domains, giving a constitutively active BRAF kinase. Functional characterization of the novel FAM131B-BRAF fusion demonstrated constitutive MEK phosphorylation potential and transforming activity in vitro. In addition, our study confirmed previously reported BRAF and RAF1 fusion variants in 72% (90/125) of PA. Mutations in BRAF (8/125), KRAS (2/125) and NF1 (4/125) and the rare RAF1 gene fusions (2/125) were mutually exclusive with BRAF rearrangements, with the exception of two cases in our series that concomitantly harbored more than one hit in the MAPK pathway. In summary, our findings further underline the fundamental role of RAF kinase fusion products as a tumor-specific marker and an ideally suited drug target for PA.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , MAP Kinase Signaling System/genetics , Mutation/genetics , Oncogene Proteins, Fusion/genetics , Proto-Oncogene Proteins B-raf/genetics , Animals , Child, Preschool , Chromosomes, Human, Pair 7/genetics , Comparative Genomic Hybridization , Female , Follow-Up Studies , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , Infant , Male , Mice , NIH 3T3 Cells , Oligonucleotide Array Sequence Analysis , Phosphorylation/genetics , Sequence Deletion , Statistics, Nonparametric , Transfection/methods
13.
Leukemia ; 34(1): 151-166, 2020 01.
Article in English | MEDLINE | ID: mdl-31431735

ABSTRACT

We used hybrid capture-targeted next-generation sequencing of circulating cell-free DNA (ccfDNA) of pediatric Hodgkin lymphoma (PHL) patients to determine pathogenic mechanisms and assess the clinical utility of this method. Hodgkin-Reed/Sternberg (HRS) cell-derived single nucleotide variants, insertions/deletions, translocations and VH-DH-JH rearrangements were detected in pretherapy ccfDNA of 72 of 96 patients. Number of variants per patient ranged from 1 to 21 with allele frequencies from 0.6 to 42%. Nine translocation breakpoints were detected. Genes involved in JAK/STAT, NFkB and PI3K signaling and antigen presentation were most frequently affected. SOCS1 variants, mainly deletions, were found in most circulating tumor (ct) DNAs, and seven of the nine translocation breakpoints involved SOCS1. Analysis of VH-DH-JH rearrangements revealed an origin of PHL HRS cells from partially selected germinal center B cells. Amounts of pretherapy ctDNA were correlated with metabolic tumor volumes. Furthermore, in all ccfDNA samples of 43 patients with early response assessment quantitative qPET < 3, indicative of a favorable clinical course, ctDNA was not detectable. In contrast, in five of six patients with qPET > 3, indicative of an unfavorable clinical course, ctDNA remained detectable. ccfDNA analysis of PHL is thus a suitable approach to determine pathogenic mechanisms and monitor therapy response.


Subject(s)
Circulating Tumor DNA/genetics , Hodgkin Disease/genetics , Adolescent , Child , Child, Preschool , Female , Genotype , Humans , Male
14.
Nucleic Acids Res ; 35(7): e51, 2007.
Article in English | MEDLINE | ID: mdl-17344319

ABSTRACT

Existing microarray-based approaches for screening of DNA methylation are hampered by a number of shortcomings, such as the introduction of bias by DNA copy-number imbalances in the test genome and negligence of tissue-specific methylation patterns. We developed a method designated array-based profiling of reference-independent methylation status (aPRIMES) that allows the detection of direct methylation status rather than relative methylation. Array-PRIMES is based on the differential restriction and competitive hybridization of methylated and unmethylated DNA by methylation-specific and methylation-sensitive restriction enzymes, respectively. We demonstrate the accuracy of aPRIMES in detecting the methylation status of CpG islands for different states of methylation. Application of aPRIMES to the DNA from desmoplastic medulloblastomas of monozygotic twins showed strikingly similar methylation profiles. Additional analysis of 18 sporadic medulloblastomas revealed an overall correlation between highly methylated tumors and poor clinical outcome and identified ZIC2 as a frequently methylated gene in pediatric medulloblastoma.


Subject(s)
Cerebellar Neoplasms/genetics , DNA Methylation , Medulloblastoma/genetics , Nuclear Proteins/genetics , Oligonucleotide Array Sequence Analysis/methods , Promoter Regions, Genetic , Transcription Factors/genetics , Cerebellar Neoplasms/metabolism , Child, Preschool , Down-Regulation , Epigenesis, Genetic , Gene Silencing , Genomics/methods , Humans , Medulloblastoma/metabolism , RNA, Messenger/metabolism , Twins, Monozygotic
15.
Leuk Lymphoma ; 60(1): 78-84, 2019 01.
Article in English | MEDLINE | ID: mdl-29718744

ABSTRACT

Osteonecrosis (ON) is a common and debilitating side effect of anti-leukemic treatment in children with acute lymphoblastic leukemia (ALL). However, the impact of leukemia itself on ON development remains elusive. We analyzed 76 children enrolled in the ongoing OPAL trial, who had magnetic resonance imaging (MRI) studies at diagnosis. MRI screening revealed 14 osteonecrotic lesions (5 × hips, 9 × knees) of any grade (I-III) in 7 (9.2%) patients. Six months on, the number of ON per patient increased (1 patient), remained constant (2), and decreased (2). The severity increased from grade I to II in two patients, remained constant (1), completely resolved (2), and decreased from grade III to osteoedema (1). No differences between adolescents initially presenting with/without ON were observed concerning age, pubertal stage, body mass index, leukemia characteristics, and clinical presentation. In MRI screening, a remarkable number of adolescents with ALL present with ON at diagnosis. The course of these ON remains highly unpredictable.


Subject(s)
Bone and Bones/pathology , Osteonecrosis/diagnosis , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Adolescent , Bone and Bones/diagnostic imaging , Child , Disease Progression , Female , Humans , Incidence , Magnetic Resonance Imaging , Male , Osteonecrosis/epidemiology , Osteonecrosis/etiology , Severity of Illness Index
16.
Eur J Cancer ; 81: 1-8, 2017 08.
Article in English | MEDLINE | ID: mdl-28586748

ABSTRACT

BACKGROUND: Temozolomide (TMZ) is widely used in high-grade glioma (HGG). There is a major concern of treatment-induced secondary haematological malignancies (SHMs). Due to the poor overall survival of HGG patients, the true incidence is yet elusive. Thus, the aim of this study was to determine the risk of SHMs following TMZ in paediatric HGG. METHODS: We analysed 487 patients from the HIT-HGG database of the German-speaking Society of Pediatric Oncology and Hematology with follow up beyond 1 year. RESULTS: The incidence of SHM was 7.7 ± 3.2% at 10 years. No SHM occurred in 194 patients after first-line TMZ therapy, but four out of 131 patients treated with TMZ for relapse following first-line multiagent chemotherapy experienced SHM (20% at 10 years; p = 0.041). SHMs occurred in two out of 162 patients who underwent multiagent chemotherapy without TMZ (4.1% at 10 years). Gender, patient age and acute haematological toxicity during treatment did not affect the incidence of SHMs. CONCLUSION: Data of our cohort do not indicate an increased risk of SHM following TMZ treatment when compared to previous chemotherapy regimen. However, if TMZ is administered as a second-line treatment following conventional chemotherapy regimen, the risk might be disproportionately increasing.


Subject(s)
Antineoplastic Agents, Alkylating/adverse effects , Dacarbazine/analogs & derivatives , Glioma/drug therapy , Hematologic Neoplasms/chemically induced , Adolescent , Austria/epidemiology , Child , Child, Preschool , Dacarbazine/adverse effects , Female , Follow-Up Studies , Germany/epidemiology , Hematologic Neoplasms/epidemiology , Humans , Incidence , Infant , Kaplan-Meier Estimate , Male , Switzerland/epidemiology , Temozolomide
17.
Pathol Oncol Res ; 10(1): 17-21, 2004.
Article in English | MEDLINE | ID: mdl-15029256

ABSTRACT

Methylation of promoter regions of CpG-rich sites is an important mechanism for silencing of tumor suppressor genes (TSG). To evaluate the role of tumor suppressor genes caspase-8 (CASP8), TIMP-3, E-cadherin (CDH1), p16INK4A, and MGMT in medulloblastoma tumorigenesis, 51 medulloblastomas (46 primary tumor specimens, 5 cell lines) were screened for methylation of promoter linked CpG-islands. For CASP8, we examined the 5' UTR region that has been shown to be associated with expression of CASP8. As detected by methylation specific PCR, methylation rate was low for TIMP-3 (3% of tumor samples; 1/5 cell lines), for MGMT (0% of tumor samples; 1/5 cell lines), for p16INK4A (2% of tumor samples; 2/5 cell lines) and for CDH1 (8% of tumor samples; 1/4 cell lines). CASP8, however, was methylated in 90% of tumor samples and 4/5 cell lines examined. Screening other tumor entities for CASP8 methylation, we found a similarly high level in 6 neuroblastoma cell lines in contrast to 5 osteosarcoma-, 4 Ewing's sarcoma- and 6 non-embryonic tumor cell lines without any increased promoter methylation. From our results we conclude that methylation of the CASP8 5' UTR region may play a role in inactivation of CASP8 in neural crest tumors.


Subject(s)
Brain Neoplasms/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/physiology , Medulloblastoma/genetics , Promoter Regions, Genetic/genetics , Adolescent , Brain Neoplasms/metabolism , Brain Neoplasms/secondary , Cadherins/genetics , Caspase 8 , Caspases/genetics , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p16/genetics , Female , Humans , Infant , Male , Medulloblastoma/metabolism , Medulloblastoma/secondary , O(6)-Methylguanine-DNA Methyltransferase/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics
18.
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
19.
Cancer Cell ; 22(4): 425-37, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23079654

ABSTRACT

Glioblastoma (GBM) is a brain tumor that carries a dismal prognosis and displays considerable heterogeneity. We have recently identified recurrent H3F3A mutations affecting two critical amino acids (K27 and G34) of histone H3.3 in one-third of pediatric GBM. Here, we show that each H3F3A mutation defines an epigenetic subgroup of GBM with a distinct global methylation pattern, and that they are mutually exclusive with IDH1 mutations, which characterize a third mutation-defined subgroup. Three further epigenetic subgroups were enriched for hallmark genetic events of adult GBM and/or established transcriptomic signatures. We also demonstrate that the two H3F3A mutations give rise to GBMs in separate anatomic compartments, with differential regulation of transcription factors OLIG1, OLIG2, and FOXG1, possibly reflecting different cellular origins.


Subject(s)
Brain Neoplasms/genetics , Epigenesis, Genetic , Glioblastoma/genetics , Histones/genetics , Isocitrate Dehydrogenase/genetics , Mutation , Adult , Brain Neoplasms/pathology , Child , DNA Methylation , Glioblastoma/pathology , Humans , Receptor, Platelet-Derived Growth Factor alpha/genetics , Transcriptome
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