Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Neuropathol Appl Neurobiol ; 48(7): e12847, 2022 12.
Article in English | MEDLINE | ID: mdl-35977725

ABSTRACT

AIMS: Anaplastic ganglioglioma is a rare tumour, and diagnosis has been based on histological criteria. The 5th edition of the World Health Organization Classification of Tumours of the Central Nervous System (CNS WHO) does not list anaplastic ganglioglioma as a distinct diagnosis due to lack of molecular data in previous publications. We retrospectively compiled a cohort of 54 histologically diagnosed anaplastic gangliogliomas to explore whether the molecular profiles of these tumours represent a separate type or resolve into other entities. METHODS: Samples were subjected to histological review, desoxyribonucleic acid (DNA) methylation profiling and next-generation sequencing. Morphological and molecular data were summarised to an integrated diagnosis. RESULTS: The majority of tumours designated as anaplastic gangliogliomas resolved into other CNS WHO diagnoses, most commonly pleomorphic xanthoastrocytoma (16/54), glioblastoma, isocitrate dehydrogenase protein (IDH) wild type and diffuse paediatric-type high-grade glioma, H3 wild type and IDH wild type (11 and 2/54), followed by low-grade glial or glioneuronal tumours including pilocytic astrocytoma, dysembryoplastic neuroepithelial tumour and diffuse leptomeningeal glioneuronal tumour (5/54), IDH mutant astrocytoma (4/54) and others (6/54). A subset of tumours (10/54) was not assignable to a CNS WHO diagnosis, and common molecular profiles pointing to a separate entity were not evident. CONCLUSIONS: In summary, we show that tumours histologically diagnosed as anaplastic ganglioglioma comprise a wide spectrum of CNS WHO tumour types with different prognostic and therapeutic implications. We therefore suggest assigning this designation with caution and recommend comprehensive molecular workup.


Subject(s)
Astrocytoma , Brain Neoplasms , Central Nervous System Neoplasms , Ganglioglioma , Glioma , Child , Humans , Ganglioglioma/pathology , Retrospective Studies , Glioma/pathology , Astrocytoma/pathology , Brain Neoplasms/genetics , Central Nervous System Neoplasms/pathology , Isocitrate Dehydrogenase
2.
Acta Neuropathol ; 136(2): 273-291, 2018 08.
Article in English | MEDLINE | ID: mdl-29564591

ABSTRACT

Tumors with histological features of pilocytic astrocytoma (PA), but with increased mitotic activity and additional high-grade features (particularly microvascular proliferation and palisading necrosis) have often been designated anaplastic pilocytic astrocytomas. The status of these tumors as a separate entity has not yet been conclusively demonstrated and molecular features have only been partially characterized. We performed DNA methylation profiling of 102 histologically defined anaplastic pilocytic astrocytomas. T-distributed stochastic neighbor-embedding (t-SNE) and hierarchical clustering analysis of these 102 cases against 158 reference cases from 12 glioma reference classes revealed that a subset of 83 of these tumors share a common DNA methylation profile that is distinct from the reference classes. These 83 tumors were thus denominated DNA methylation class anaplastic astrocytoma with piloid features (MC AAP). The 19 remaining tumors were distributed amongst the reference classes, with additional testing confirming the molecular diagnosis in most cases. Median age of patients with MC AAP was 41.5 years. The most frequent localization was the posterior fossa (74%). Deletions of CDKN2A/B (66/83, 80%), MAPK pathway gene alterations (49/65, 75%, most frequently affecting NF1, followed by BRAF and FGFR1) and mutations of ATRX or loss of ATRX expression (33/74, 45%) were the most common molecular alterations. All tumors were IDH1/2 wildtype. The MGMT promoter was methylated in 38/83 tumors (45%). Outcome analysis confirmed an unfavorable clinical course in comparison to PA, but better than IDH wildtype glioblastoma. In conclusion, we show that a subset of histologically defined anaplastic pilocytic astrocytomas forms a separate DNA methylation cluster, harbors recurrent alterations in MAPK pathway genes in combination with alterations of CDKN2A/B and ATRX, affects patients who are on average older than those diagnosed with PA and has an intermediate clinical outcome.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Isocitrate Dehydrogenase/genetics , Signal Transduction/genetics , Adolescent , Adult , Age Factors , Aged , Aged, 80 and over , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Methylation/genetics , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Female , Histones/genetics , Histones/metabolism , Humans , Infant , Kaplan-Meier Estimate , Male , Middle Aged , Mitogen-Activated Protein Kinase Kinases/genetics , Mutation/genetics , Retrospective Studies , Tumor Suppressor Proteins/metabolism , X-linked Nuclear Protein/genetics , Young Adult
3.
Int J Cancer ; 134(10): 2437-47, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24614983

ABSTRACT

The epidermal growth factor receptor vIII mutant (EGFRvIII) is found in ~50% of all EGFR-amplified glioblastomas and constitutes a tumor-specific therapeutic target. To assess molecular testing approaches and the prognostic role of EGFRvIII in patients treated according to current standards of care, we compared different EGFRvIII detection methods and correlated EGFRvIII status with outcome in a prospective patient cohort of the German Glioma Network. In total, 184 newly diagnosed glioblastoma patients were investigated for EGFR amplification and for expression of EGFR and EGFRvIII by immunohistochemistry. Further, the EGFRvIII status was additionally studied by multiplex ligation-dependent probe amplification (MLPA) analysis and reverse transcription-PCR (RT-PCR). Immunohistochemistry demonstrated EGFRvIII in 34 of 184 patients (18%). RT-PCR or MLPA analysis detected four additional EGFRvIII-positive patients. Overall, RT-PCR and immunohistochemistry were more sensitive for EGFRvIII detection than MLPA. EGFRvIII status was not associated with progression-free and overall survival. EGFRvIII also had no prognostic significance in the subgroup of patients who were free from progression after concomitant radiochemotherapy and thus would be eligible for the ongoing ACT IV EGFRvIII vaccination trial. Age, extent of resection and O6-methylguanine DNA methyltransferase (MGMT) promoter methylation status appeared to be less prognostic in EGFRvIII-positive patients. Thus, EGFRvIII positivity is not a major negative prognostic factor in glioblastoma patients treated according to current standards of care. Data from phase II EGFRvIII-targeted vaccination trials compare favorably with the present contemporary results, supporting the further exploration of EGVRvIII vaccination in newly diagnosed glioblastoma.


Subject(s)
Dacarbazine/analogs & derivatives , ErbB Receptors/genetics , Glioblastoma/therapy , Mutation , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/therapeutic use , Blotting, Western , Chemoradiotherapy , Dacarbazine/therapeutic use , Disease-Free Survival , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/surgery , Humans , Immunohistochemistry , Male , Middle Aged , Multivariate Analysis , Outcome Assessment, Health Care , Prognosis , Prospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Temozolomide , Young Adult
4.
Neuro Oncol ; 25(2): 290-302, 2023 02 14.
Article in English | MEDLINE | ID: mdl-35802605

ABSTRACT

BACKGROUND: Recent efforts have described the evolution of glioblastoma from initial diagnosis to post-treatment recurrence on a genomic and transcriptomic level. However, the evolution of the proteomic landscape is largely unknown. METHODS: Sequential window acquisition of all theoretical fragment ion spectra mass spectrometry (SWATH-MS) was used to characterize the quantitative proteomes of two independent cohorts of paired newly diagnosed and recurrent glioblastomas. Recurrence-associated proteins were validated using immunohistochemistry and further studied in human glioma cell lines, orthotopic xenograft models, and human organotypic brain slice cultures. External spatial transcriptomic, single-cell, and bulk RNA sequencing data were analyzed to gain mechanistic insights. RESULTS: Although overall proteomic changes were heterogeneous across patients, we identified BCAS1, INF2, and FBXO2 as consistently upregulated proteins at recurrence and validated these using immunohistochemistry. Knockout of FBXO2 in human glioma cells conferred a strong survival benefit in orthotopic xenograft mouse models and reduced invasive growth in organotypic brain slice cultures. In glioblastoma patient samples, FBXO2 expression was enriched in the tumor infiltration zone and FBXO2-positive cancer cells were associated with synaptic signaling processes. CONCLUSIONS: These findings demonstrate a potential role of FBXO2-dependent glioma-microenvironment interactions to promote tumor growth. Furthermore, the published datasets provide a valuable resource for further studies.


Subject(s)
Brain Neoplasms , F-Box Proteins , Glioblastoma , Glioma , Humans , Animals , Mice , Glioblastoma/pathology , Proteomics , Mice, Knockout , Glioma/pathology , Brain/pathology , Brain Neoplasms/pathology , Proteins , Tumor Microenvironment , Neoplasm Proteins , Nerve Tissue Proteins , Cell Cycle Proteins , F-Box Proteins/genetics
5.
Acta Neuropathol Commun ; 10(1): 5, 2022 01 10.
Article in English | MEDLINE | ID: mdl-35012690

ABSTRACT

Pleomorphic xanthoastrocytoma (PXA) in its classic manifestation exhibits distinct morphological features and is assigned to CNS WHO grade 2 or grade 3. Distinction from glioblastoma variants and lower grade glial and glioneuronal tumors is a common diagnostic challenge. We compared a morphologically defined set of PXA (histPXA) with an independent set, defined by DNA methylation analysis (mcPXA). HistPXA encompassed 144 tumors all subjected to DNA methylation array analysis. Sixty-two histPXA matched to the methylation class mcPXA. These were combined with the cases that showed the mcPXA signature but had received a histopathological diagnosis other than PXA. This cohort constituted a set of 220 mcPXA. Molecular and clinical parameters were analyzed in these groups. Morphological parameters were analyzed in a subset of tumors with FFPE tissue available. HistPXA revealed considerable heterogeneity in regard to methylation classes, with methylation classes glioblastoma and ganglioglioma being the most frequent mismatches. Similarly, the mcPXA cohort contained tumors of diverse histological diagnoses, with glioblastoma constituting the most frequent mismatch. Subsequent analyses demonstrated the presence of canonical pTERT mutations to be associated with unfavorable prognosis among mcPXA. Based on these data, we consider the tumor type PXA to be histologically more varied than previously assumed. Histological approach to diagnosis will predominantly identify cases with the established archetypical morphology. DNA methylation analysis includes additional tumors in the tumor class PXA that share similar DNA methylation profile but lack the typical morphology of a PXA. DNA methylation analysis also assist in separating other tumor types with morphologic overlap to PXA. Our data suggest the presence of canonical pTERT mutations as a robust indicator for poor prognosis in methylation class PXA.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Telomerase/genetics , Astrocytoma/mortality , Astrocytoma/pathology , Brain Neoplasms/mortality , Brain Neoplasms/pathology , DNA Methylation , Humans , Mutation , Prognosis , Survival Rate
6.
Acta Neuropathol ; 122(4): 495-510, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21863242

ABSTRACT

Glioblastoma (GBM) is a highly aggressive brain tumour, where patients respond poorly to radiotherapy and exhibit dismal survival outcomes. The mechanisms of radioresistance are not completely understood. However, cancer cells with an immature stem-like phenotype are hypothesised to play a role in radioresistance. Since the progenitor marker neuron-glial-2 (NG2) has been shown to regulate several aspects of GBM progression in experimental systems, we hypothesised that its expression would influence the survival of GBM patients. Quantification of NG2 expression in 74 GBM biopsies from newly diagnosed and untreated patients revealed that 50% express high NG2 levels on tumour cells and associated vessels, being associated with significantly shorter survival. This effect was independent of age at diagnosis, treatment received and hypermethylation of the O(6)-methylguanine methyltransferase (MGMT) DNA repair gene promoter. NG2 was frequently co-expressed with nestin and vimentin but rarely with CD133 and the NG2 positive tumour cells harboured genetic aberrations typical for GBM. 2D proteomics of 11 randomly selected biopsies revealed upregulation of an antioxidant, peroxiredoxin-1 (PRDX-1), in the shortest surviving patients. Expression of PRDX-1 was associated with significantly reduced products of oxidative stress. Furthermore, NG2 expressing GBM cells showed resistance to ionising radiation (IR), rapidly recognised DNA damage and effectuated cell cycle checkpoint signalling. PRDX-1 knockdown transiently slowed tumour growth rates and sensitised them to IR in vivo. Our data establish NG2 as an important prognostic factor for GBM patient survival, by mediating resistance to radiotherapy through induction of ROS scavenging enzymes and preferential DNA damage signalling.


Subject(s)
Antigens/biosynthesis , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/radiotherapy , DNA Damage/genetics , Glioblastoma/genetics , Glioblastoma/radiotherapy , Proteoglycans/biosynthesis , Stem Cells/metabolism , Aged , Antigens/genetics , Antigens/radiation effects , Biomarkers, Tumor/radiation effects , Brain Neoplasms/pathology , DNA Damage/radiation effects , Female , Glioblastoma/pathology , Humans , Male , Middle Aged , Predictive Value of Tests , Prognosis , Proteoglycans/genetics , Proteoglycans/radiation effects , Radiation Tolerance , Radiation, Ionizing , Stem Cells/pathology , Stem Cells/radiation effects , Survival Rate/trends
7.
Cancers (Basel) ; 13(4)2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33578810

ABSTRACT

BACKGROUND: Development of brain metastases in advanced melanoma patients is a frequent event that limits patients' quality of life and survival. Despite recent insights into melanoma genetics, systematic analyses of genetic alterations in melanoma brain metastasis formation are lacking. Moreover, whether brain metastases harbor distinct genetic alterations beyond those observed at different anatomic sites of the same patient remains unknown. EXPERIMENTAL DESIGN AND RESULTS: In our study, 54 intracranial and 18 corresponding extracranial melanoma metastases were analyzed for mutations using targeted next generation sequencing of 29 recurrently mutated driver genes in melanoma. In 11 of 16 paired samples, we detected nucleotide modifications in brain metastases that were absent in matched metastases at extracranial sites. Moreover, we identified novel genetic variants in ARID1A, ARID2, SMARCA4 and BAP1, genes that have not been linked to brain metastases before; albeit most frequent mutations were found in ARID1A, ARID2 and BRAF. Conclusion: Our data provide new insights into the genetic landscape of intracranial melanoma metastases supporting a branched evolution model of metastasis formation.

8.
Cancer Res ; 79(6): 1226-1238, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30674534

ABSTRACT

Inevitable tumor recurrence and a poor median survival are frustrating reminders of the inefficacy of our current standard of care for patients with newly diagnosed glioblastoma (GBM), which includes surgery followed by radiotherapy and chemotherapy with the DNA alkylating agent temozolomide. Because resistance to genotoxic damage is achieved mainly through execution of the DNA damage response (DDR) and DNA repair pathways, knowledge of the changes in DNA repair and cell-cycle gene expression that occur during tumor development might help identify new targets and improve treatment. Here, we performed a gene expression analysis targeting components of the DNA repair and cell-cycle machineries in cohorts of paired tumor samples (i.e., biopsies from the same patient obtained at the time of primary tumor operation and at recurrence) from patients treated with radiotherapy or radiotherapy plus temozolomide. We identified and validated a 27-gene signature that resulted in the classification of GBM specimens into three groups, two of which displayed inverse expression profiles. Each group contained primary and recurrent samples, and the tumor at relapse frequently displayed a gene expression profile different from that of the matched primary biopsy. Within the groups that exhibited opposing gene expression profiles, the expression pattern of the gene signature at relapse was linked to progression-free survival. We provide experimental evidence that our signature exposes group-specific vulnerabilities against genotoxicants and inhibitors of the cell cycle and DDR, with the prospect of personalized therapeutic strategies.Significance: These findings suggest that classification of GBM tumors based on a DNA repair and cell-cycle gene expression signature exposes vulnerabilities to standard-of-care therapies and offers the potential for personalized therapeutic strategies.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Cell Cycle Proteins/genetics , DNA Repair Enzymes/genetics , Glioblastoma/genetics , Neoplasm Recurrence, Local/genetics , Adult , Aged , Apoptosis , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cell Proliferation , Cohort Studies , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/radiotherapy , Prognosis , Survival Rate , Tumor Cells, Cultured
9.
Acta Neuropathol Commun ; 7(1): 163, 2019 10 28.
Article in English | MEDLINE | ID: mdl-31661039

ABSTRACT

In this multi-institutional study we compiled a retrospective cohort of 86 posterior fossa tumors having received the diagnosis of cerebellar glioblastoma (cGBM). All tumors were reviewed histologically and subjected to array-based methylation analysis followed by algorithm-based classification into distinct methylation classes (MCs). The single MC containing the largest proportion of 25 tumors diagnosed as cGBM was MC anaplastic astrocytoma with piloid features representing a recently-described molecular tumor entity not yet included in the WHO Classification of Tumours of the Central Nervous System (WHO classification). Twenty-nine tumors molecularly corresponded to either of 6 methylation subclasses subsumed in the MC family GBM IDH wildtype. Further we identified 6 tumors belonging to the MC diffuse midline glioma H3 K27 M mutant and 6 tumors allotted to the MC IDH mutant glioma subclass astrocytoma. Two tumors were classified as MC pilocytic astrocytoma of the posterior fossa, one as MC CNS high grade neuroepithelial tumor with BCOR alteration and one as MC control tissue, inflammatory tumor microenvironment. The methylation profiles of 16 tumors could not clearly be assigned to one distinct MC. In comparison to supratentorial localization, the MC GBM IDH wildtype subclass midline was overrepresented, whereas the MCs GBM IDH wildtype subclass mesenchymal and subclass RTK II were underrepresented in the cerebellum. Based on the integration of molecular and histological findings all tumors received an integrated diagnosis in line with the WHO classification 2016. In conclusion, cGBM does not represent a molecularly uniform tumor entity, but rather comprises different brain tumor entities with diverse prognosis and therapeutic options. Distinction of these molecular tumor classes requires molecular analysis. More than 30% of tumors diagnosed as cGBM belong to the recently described molecular entity of anaplastic astrocytoma with piloid features.


Subject(s)
Cerebellar Neoplasms/diagnosis , Cerebellar Neoplasms/metabolism , Glioblastoma/diagnosis , Glioblastoma/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Cerebellar Neoplasms/pathology , Child , Child, Preschool , Cyclin-Dependent Kinase Inhibitor p16/metabolism , ErbB Receptors/metabolism , Female , Glioblastoma/pathology , Humans , Infant , Infant, Newborn , Infratentorial Neoplasms/diagnosis , Infratentorial Neoplasms/metabolism , Infratentorial Neoplasms/pathology , Male , Methylation , Middle Aged , Retrospective Studies , Telomerase/metabolism , Young Adult
10.
Eur J Hum Genet ; 26(1): 137-142, 2018 01.
Article in English | MEDLINE | ID: mdl-29230040

ABSTRACT

The sonic hedgehog (SHH) signaling pathway has been shown to play important roles in embryogenesis, cell proliferation as well as in cell differentiation. It is aberrantly activated in various common cancers in adults, but also in pediatric neoplasms, such as rhabdomyosarcoma (RMS) and atypical teratoid/rhabdoid tumors (AT/RTs). Dysregulation and germline mutation in PATCHED1 (PTCH1), a receptor for SHH, is responsible for the Gorlin Syndrome, a familial cancer predisposing syndrome including RMS. Here, we report a newborn diagnosed with congenital embryonal RMS. Whole-exome sequencing (WES) identified the presence of two heterozygous germline mutations in two target genes of the SHH signaling pathway. The PTCH1 mutation p.(Gly38Glu) is inherited from the mother, whereas the PTCH2 p.(His622Tyr) mutation is transmitted from the father. Quantitative RT-PCR expression analysis of GLI and SMO, key players of the SHH pathway, showed significantly increase in the tumor tissue of the patient and also enrichment in the germline sample in comparison to the parents indicating activation of the SHH pathway in the patient. These findings demonstrate that SHH pathway activity seems to play a role in eRMS as evidenced by high expression levels of GLI1 RNA transcripts. We speculate that PTCH2 modulates tumorigenesis linked to the PTCH1 mutation and is likely associated with the congenital onset of the RMS observed in our patient.


Subject(s)
Germ-Line Mutation , Patched-1 Receptor/genetics , Patched-2 Receptor/genetics , Rhabdomyosarcoma, Embryonal/genetics , Humans , Infant, Newborn , Male , Rhabdomyosarcoma, Embryonal/pathology
11.
Eur J Hum Genet ; 26(3): 440-444, 2018 03.
Article in English | MEDLINE | ID: mdl-29302048

ABSTRACT

Constitutional mismatch repair deficiency (CMMRD) is an autosomal recessively inherited childhood cancer susceptibility syndrome caused by biallelic germline mutations in one of the mismatch repair (MMR) genes. The spectrum of CMMRD-associated tumours is very broad and many CMMRD patients additionally display signposting non-neoplastic features, most frequently café-au-lait macules and other pigmentation alterations. We report on a 13-month-old girl suspected of having CMMRD due to a desmoplastic medulloblastoma and a striking skin pigmentation that included multiple café-au-lait macules, hypopigmented areas and Mongolian spots. Whole-exome sequencing revealed homozygosity for MSH2 variant p.(Leu92Val) and MSH6 variant p.(Val809del), both variants of uncertain significance (VUS). Immunohistochemical analysis of the tumour tissue showed expression of all four MMR proteins and gMSI testing was negative. However, functional assays demonstrated that the cells of the patient displayed methylation tolerance and ex vivo microsatellite instability, which unequivocally confirmed the diagnosis of CMMRD. Taken together, the results render the MSH2 variant unlikely to be responsible for the phenotype, while they are compatible with MSH6-associated CMMRD. This case illustrates the diagnostic strategy of confirming CMMRD syndrome in patients with VUS.


Subject(s)
Cerebellar Neoplasms/genetics , DNA-Binding Proteins/genetics , Genetic Testing/methods , Medulloblastoma/genetics , MutS Homolog 2 Protein/genetics , Phenotype , Cell Line, Tumor , Cells, Cultured , Cerebellar Neoplasms/pathology , DNA-Binding Proteins/metabolism , Diagnosis, Differential , Female , Homozygote , Humans , Infant , Medulloblastoma/pathology , MutS Homolog 2 Protein/metabolism
12.
Cancer Cell ; 34(1): 163-177.e7, 2018 07 09.
Article in English | MEDLINE | ID: mdl-29990498

ABSTRACT

We explored the clinical and pathological impact of epidermal growth factor receptor (EGFR) extracellular domain missense mutations. Retrospective assessment of 260 de novo glioblastoma patients revealed a significant reduction in overall survival of patients having tumors with EGFR mutations at alanine 289 (EGFRA289D/T/V). Quantitative multi-parametric magnetic resonance imaging analyses indicated increased tumor invasion for EGFRA289D/T/V mutants, corroborated in mice bearing intracranial tumors expressing EGFRA289V and dependent on ERK-mediated expression of matrix metalloproteinase-1. EGFRA289V tumor growth was attenuated with an antibody against a cryptic epitope, based on in silico simulation. The findings of this study indicate a highly invasive phenotype associated with the EGFRA289V mutation in glioblastoma, postulating EGFRA289V as a molecular marker for responsiveness to therapy with EGFR-targeting antibodies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Brain Neoplasms/genetics , ErbB Receptors/genetics , Glioblastoma/genetics , Magnetic Resonance Imaging , Mutation, Missense , Adolescent , Adult , Aged , Animals , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Child , Child, Preschool , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Genetic Predisposition to Disease , Glioblastoma/diagnostic imaging , Glioblastoma/metabolism , Humans , Image Interpretation, Computer-Assisted , Infant , Infant, Newborn , Machine Learning , Male , Matrix Metalloproteinase 1/metabolism , Mice, Nude , Middle Aged , Neoplasm Invasiveness , Phenotype , Phosphorylation , Predictive Value of Tests , Protein Domains , Retrospective Studies , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Young Adult
13.
World Neurosurg ; 108: 118-127, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28866060

ABSTRACT

OBJECTIVE: To compare fluorescence intensity of tumor specimens, as measured by a fluorescence-guided surgery microscope and a spectrometer, to evaluate tumor infiltration of dura mater around meningiomas with help of these 2 different 5-aminolevulinic acid (5-ALA)-based fluorescence tools, and to correlate fluorescence intensity with histopathologic data. MATERIAL AND METHODS: In a clinical series, meningiomas were resected by 5-ALA fluorescence-guided surgery. Fluorescence intensity was semiquantitatively rated by the surgeon at predefined points. Biopsies were harvested and fluorescence intensity measured by a spectrometer and histopathologically analyzed. Sampling was realized at the level of the dura in a centrifugal direction. RESULTS: A total of 104 biopsies (n = 13 tumors) were analyzed. Specificity and sensitivity of the microscope were 0.96 and 0.53 and of the spectrometer 0.95 and 0.93, respectively. Fluorescence intensity as measured by the spectrometer was correlated to histologically confirmed tumor burden. In a centrifugal direction, tumor burden and fluorescence intensity continuously decreased (along the dural tail). Below a threshold value of 639 arbitrary units no tumor was histologically detectable. CONCLUSIONS: At the level of the dura the spectrometer was highly sensitive for detection of meningioma cells. The surgical microscope showed false negative results and missed residual tumor cells in more than one half of the cases. The complementary use of both fluorescence tools may improve resection quality.


Subject(s)
Dura Mater/diagnostic imaging , Meningeal Neoplasms/diagnostic imaging , Meningeal Neoplasms/surgery , Meningioma/diagnostic imaging , Meningioma/surgery , Microscopy, Fluorescence , Spectrometry, Fluorescence , Aminolevulinic Acid , Calibration , Dura Mater/pathology , Dura Mater/surgery , Fluorescent Dyes , Humans , Magnetic Resonance Imaging , Meningeal Neoplasms/pathology , Meningioma/pathology , Microsurgery , Neurosurgical Procedures , Sensitivity and Specificity , Surgery, Computer-Assisted , Tumor Burden
14.
Oncotarget ; 7(50): 82028-82045, 2016 Dec 13.
Article in English | MEDLINE | ID: mdl-27880937

ABSTRACT

The outcome of patients with anaplastic gliomas varies considerably depending on single molecular markers, such as mutations of the isocitrate dehydrogenase (IDH) genes, as well as molecular classifications based on epigenetic or genetic profiles. Remarkably, 98% of the RNA within a cell is not translated into proteins. Of those, especially microRNAs (miRNAs) have been shown not only to have a major influence on physiologic processes but also to be deregulated and prognostic in malignancies.To find novel survival markers and treatment options we performed unbiased DNA methylation screens that revealed 12 putative miRNA promoter regions with differential DNA methylation in anaplastic gliomas. Methylation of these candidate regions was validated in different independent patient cohorts revealing a set of miRNA promoter regions with prognostic relevance across data sets. Of those, miR-155 promoter methylation and miR-155 expression were negatively correlated and especially the methylation showed superior correlation with patient survival compared to established biomarkers.Functional examinations in malignant glioma cells further cemented the relevance of miR-155 for tumor cell viability with transient and stable modifications indicating an onco-miRNA activity. MiR-155 also conferred resistance towards alkylating temozolomide and radiotherapy as consequence of nuclear factor (NF)κB activation.Preconditioning glioma cells with an NFκB inhibitor reduced therapy resistance of miR-155 overexpressing cells. These cells resembled tumors with a low methylation of the miR-155 promoter and thus mir-155 or NFκB inhibition may provide treatment options with a special focus on patients with IDH wild type tumors.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , DNA Methylation , Epigenesis, Genetic , Glioma/genetics , MicroRNAs/genetics , Antineoplastic Agents, Alkylating/pharmacology , Biomarkers, Tumor/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Cell Line, Tumor , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Glioma/mortality , Glioma/pathology , Glioma/therapy , Humans , Kaplan-Meier Estimate , MicroRNAs/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Phenotype , Prognosis , Promoter Regions, Genetic , Proportional Hazards Models , RNA Interference , Radiation Tolerance/genetics , Temozolomide , Time Factors , Transcription, Genetic , Transfection
15.
Neuro Oncol ; 16(12): 1630-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25028501

ABSTRACT

BACKGROUND: Molecular biomarkers including isocitrate dehydrogenase 1 or 2 (IDH1/2) mutation, 1p/19q codeletion, and O(6)-methylguanine-DNA-methyltransferase (MGMT) promoter methylation may improve prognostication and guide treatment decisions for patients with World Health Organization (WHO) anaplastic gliomas. At present, each marker is individually tested by distinct assays. Illumina Infinium HumanMethylation450 BeadChip arrays (HM450) enable the determination of large-scale methylation profiles and genome-wide DNA copy number changes. Algorithms have been developed to detect the glioma CpG island methylator phenotype (G-CIMP) associated with IDH1/2 mutation, 1p/19q codeletion, and MGMT promoter methylation using a single assay. METHODS: Here, we retrospectively investigated the diagnostic and prognostic performance of these algorithms in comparison to individual marker testing and patient outcome in the biomarker cohort (n = 115 patients) of the NOA-04 trial. RESULTS: Concordance for IDH and 1p/19q status was very high: In 92% of samples, the HM450 and reference data agreed. In discordant samples, survival analysis by Kaplan-Meier and Cox regression analyses suggested a more accurate assessment of biological phenotype by the HM450 analysis. The HM450-derived MGMT-STP27 model to calculate MGMT promoter methylation probability revealed this aberration in a significantly higher fraction of samples than conventional methylation-specific PCR, with 87 of 91 G-CIMP tumors predicted as MGMT promoter-methylated. Pyrosequencing of discordant samples confirmed the HM450 assessment in 14 of 17 cases. CONCLUSIONS: G-CIMP and 1p/19q codeletion are reliably detectable by HM450 analysis and are associated with prognosis in the NOA-04 trial. For MGMT, HM450 suggests promoter methylation in the vast majority of G-CIMP tumors, which is supported by pyrosequencing.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , DNA Methylation , Glioma/diagnosis , Glioma/genetics , Adult , Aged , Algorithms , Biomarkers , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 19/genetics , CpG Islands , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Female , Gene Deletion , Humans , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , Phenotype , Promoter Regions, Genetic , Retrospective Studies , Tumor Suppressor Proteins/genetics , Young Adult
16.
Int J Cancer ; 119(4): 792-800, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16550607

ABSTRACT

Loss of heterozygosity (LOH) on chromosomal arms 1p and 19q is the most common genetic alteration in oligodendroglial tumors and associated with response to radio- and chemotherapy as well as favorable prognosis. Using microsatellite analysis, we previously identified the chromosomal regions 1p36.22-p36.31 and 19q13.3, as candidate tumor suppressor gene regions being commonly deleted in these tumors. To identify genes within these regions that are downregulated in oligodendroglial tumors with LOH 1p/19q, we performed cDNA microarray-based RNA expression profiling of 35 gliomas with known allelic status on 1p and 19q, including 7 oligodendrogliomas and 8 diffuse astrocytomas of World Health Organization (WHO) grade II, as well as 14 anaplastic oligodendrogliomas and 6 anaplastic oligoastrocytomas of WHO grade III. The microarrays used for expression profiling carried approximately 7,000 gene-specific cDNAs, with complete coverage of the genes located in 1p36.13-p36.31 and 19q13.2-q13.33. Microarray analysis identified 8 genes from these regions (MGC4399, SRM, ICMT, RPL18, FTL, ZIN, FLJ10781 and DBP), which all showed significantly lower expression in 1p/19q-deleted gliomas when compared to gliomas without 1p/19q losses. Quantitative real-time reverse transcription-PCR analyses were performed for the MGC4399, ICMT and RPL18 genes and confirmed the microarray findings. In addition, we found that the cytosolic phospholipase A2 (PLA2G4C) gene at 19q13.3 demonstrated significantly lower expression in anaplastic oligodendrogliomas (WHO grade III) when compared to well-differentiated oligodendrogliomas (WHO grade II). Taken together, our study provides a set of interesting novel candidate genes that may play important roles in the pathogenesis of oligodendroglial tumors.


Subject(s)
Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 1/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Oligodendroglioma/genetics , Oligonucleotide Array Sequence Analysis , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Aged , Child , DNA, Complementary/genetics , Down-Regulation , Female , Humans , Male , Middle Aged , Transcription, Genetic/genetics , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL