Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
J Cancer Res Clin Oncol ; 149(9): 5677-5685, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36539535

ABSTRACT

PURPOSE: The genetic characteristics and mismatch repair (MMR) status of the primary tumor and corresponding metastases in colorectal cancer (CRC) are generally considered to be highly concordant. This implies that either the primary or metastatic tumor can be used for testing gene mutation and MMR status. However, whether this is also true for CRC and their ovarian metastases is currently unknown. Ovarian metastases generally show a poorer response to systemic therapy compared to other metastatic sites. Differences in biomarker status between primary CRC and ovarian metastases could possibly explain this difference in therapy response. METHODS: The study cohort was selected from CRC patients treated in two Dutch hospitals. Eligible patients with CRC and ovarian metastasis who were surgically treated between 2011 and 2018 were included. CRC and corresponding ovarian metastatic tissues were paired. Gene mutation status was established using next-generation sequencing, while the MMR status was established using either immunohistochemistry or microsatellite instability analysis. RESULTS: Matched samples of CRC and ovarian metastasis from 26 patients were available for analysis. A biomarker concordance of 100% was detected. CONCLUSION: Complete biomarker concordance was found between MMR proficient CRC and their matching ovarian metastasis. Biomarker testing of MMR proficient CRC tissue appears to be sufficient, and additional testing of metastatic ovarian tissue is not necessary. Differences in therapy response between ovarian metastases and other metastases from CRC are thus unlikely to be caused by differences in the genetic status.


Subject(s)
Colorectal Neoplasms , Ovarian Neoplasms , Humans , Female , Cohort Studies , Colorectal Neoplasms/pathology , Mutation , Biomarkers , Ovarian Neoplasms/genetics , DNA Mismatch Repair/genetics , Microsatellite Instability
2.
Virchows Arch ; 474(6): 673-680, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30888490

ABSTRACT

Next-generation sequencing (NGS) panel analysis on DNA from formalin-fixed paraffin-embedded (FFPE) tissue is increasingly used to also identify actionable copy number gains (gene amplifications) in addition to sequence variants. While guidelines for the reporting of sequence variants are available, guidance with respect to reporting copy number gains from gene-panel NGS data is limited. Here, we report on Dutch consensus recommendations obtained in the context of the national Predictive Analysis for THerapy (PATH) project, which aims to optimize and harmonize routine diagnostics in molecular pathology. We briefly discuss two common approaches to detect gene copy number gains from NGS data, i.e., the relative coverage and B-allele frequencies. In addition, we provide recommendations for reporting gene copy gains for clinical purposes. In addition to general QC metrics associated with NGS in routine diagnostics, it is recommended to include clinically relevant quantitative parameters of copy number gains in the clinical report, such as (i) relative coverage and estimated copy numbers in neoplastic cells, (ii) statistical scores to show significance (e.g., z-scores), and (iii) the sensitivity of the assay and restrictions of NGS-based detection of copy number gains. Collectively, this information can guide clinical and analytical decisions such as the reliable detection of high-level gene amplifications and the requirement for additional in situ assays in case of borderline results or limited sensitivity.


Subject(s)
DNA Copy Number Variations/physiology , Gene Dosage/genetics , Genetic Testing , High-Throughput Nucleotide Sequencing , High-Throughput Nucleotide Sequencing/methods , Humans , Mutation/genetics , Pathology, Molecular/methods , Sequence Analysis, DNA/methods
3.
Sci Rep ; 4: 5260, 2014 Jun 11.
Article in English | MEDLINE | ID: mdl-24919120

ABSTRACT

Drug resistance is a major issue in the treatment of glioblastoma. Almost all glioblastomas are intrinsically resistant to chemotherapeutic temozolomide (TMZ) or develop resistance during treatment. The interaction networks of microRNAs (miRNAs) and mRNAs likely regulate most biological processes and can be employed to better understand complex processes including drug resistance in cancer. In this study, we examined if integrative miRNA/mRNA network analysis using the web-service tool mirConnX could be used to identify drug resistance factors in glioblastoma. We used TMZ-resistant glioblastoma cells and their integrated miRNA/mRNA networks to identify TMZ-sensitizing factors. TMZ resistance was previously induced in glioblastoma cell lines U87, Hs683, and LNZ308. miRNA/mRNA expression profiling of these cells and integration of the profiles using mirConnX resulted in the identification of plant homeodomain (PHD)-like finger 6 (PHF6) as a potential TMZ-sensitizing factor in resistant glioblastoma cells. Analysis of PHF6 expression showed significant upregulation in glioblastoma as compared to normal tissue. Interference with PHF6 expression in three TMZ-resistant subclones significantly enhanced TMZ-induced cell kill in two of these cell lines. Altogether, these results demonstrate that mirConnX is a feasible and useful tool to investigate miRNA/mRNA interactions in TMZ-resistant cells and has potential to identify drug resistance factors in glioblastoma.


Subject(s)
Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm/genetics , Gene Regulatory Networks/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , MicroRNAs/genetics , RNA, Messenger/genetics , Carrier Proteins/genetics , Cell Line, Tumor , Dacarbazine/pharmacology , Humans , Repressor Proteins , Temozolomide , Up-Regulation/genetics
4.
Oncotarget ; 5(2): 363-74, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24495907

ABSTRACT

Glioblastoma is the most common malignant primary brain tumor. Temozolomide (TMZ) is the standard chemotherapeutic agent for this disease. However, intrinsic and acquired TMZ-resistance represents a major obstacle for this therapy. In order to identify factors involved in TMZ-resistance, we engineered different TMZ-resistant glioblastoma cell lines. Gene expression analysis demonstrated that EFEMP1, an extracellular matrix protein, is associated with TMZ-resistant phenotype. Silencing of EFEMP1 in glioblastoma cells resulted in decreased cell survival following TMZ treatment, whereas overexpression caused TMZ-resistance. EFEMP1 acts via multiple signaling pathways, including γ-secretase-mediated activation of the Notch pathway. We show that inhibition of γ-secretase by RO4929097 causes at least partial sensitization of glioblastoma cells to temozolomide in vitro and in vivo. In addition, we show that EFEMP1 expression levels correlate with survival in TMZ-treated glioblastoma patients. Altogether our results suggest EFEMP1 as a potential therapeutic target to overcome TMZ-resistance in glioblastoma.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Extracellular Matrix Proteins/metabolism , Glioblastoma/drug therapy , Receptors, Notch/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzazepines/administration & dosage , Benzazepines/pharmacology , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Dacarbazine/administration & dosage , Dacarbazine/pharmacology , Drug Resistance, Neoplasm , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/genetics , Female , Gene Expression , Glioblastoma/genetics , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Receptors, Notch/genetics , Signal Transduction , Temozolomide , Transfection
5.
Neuro Oncol ; 16(9): 1263-73, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24510240

ABSTRACT

BACKGROUND: Genetic and epigenetic profiling of glioblastomas has provided a comprehensive list of altered cancer genes of which only O(6)-methylguanine-methyltransferase (MGMT) methylation is used thus far as a predictive marker in a clinical setting. We investigated the prognostic significance of genetic and epigenetic alterations in glioblastoma patients. METHODS: We screened 98 human glioblastoma samples for genetic and epigenetic alterations in 10 genes and chromosomal loci by PCR and multiplex ligation-dependent probe amplification (MLPA). We tested the association between these genetic and epigenetic alterations and glioblastoma patient survival. Subsequently, we developed a 2-gene survival predictor. RESULTS: Multivariate analyses revealed that mutations in isocitrate dehydrogenase 1 (IDH1), promoter methylation of MGMT, irradiation dosage, and Karnofsky Performance Status (KFS) were independent prognostic factors. A 2-gene predictor for glioblastoma survival was generated. Based on the genetic and epigenetic status of IDH1 and MGMT, glioblastoma patients were stratified into 3 clinically different genotypes: glioblastoma patients with IDH1mt/MGMTmet had the longest survival, followed by patients with IDH1mt/MGMTunmet or IDH1wt/MGMTmet, and patients with IDH1wt/MGMTunmet had the shortest survival. This 2-gene predictor was an independent prognostic factor and performed significantly better in predicting survival than either IDH1 mutations or MGMT methylation alone. The predictor was validated in 3 external datasets. DISCUSSION: The combination of IDH1 mutations and MGMT methylation outperforms either IDH1 mutations or MGMT methylation alone in predicting survival of glioblastoma patients. This information will help to increase our understanding of glioblastoma biology, and it may be helpful for baseline comparisons in future clinical trials.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/mortality , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Glioblastoma/genetics , Glioblastoma/mortality , Isocitrate Dehydrogenase/genetics , Mutation , Tumor Suppressor Proteins/genetics , Adult , Aged , Aged, 80 and over , DNA Methylation , Epigenesis, Genetic , Female , Humans , Male , Middle Aged , Survival Analysis
6.
Anal Chem ; 85(21): 10205-10, 2013 Nov 05.
Article in English | MEDLINE | ID: mdl-24131224

ABSTRACT

Reporters secreted into the conditioned medium of cells in culture or into blood in vivo have shown to be useful tools for simple and noninvasive monitoring of biological processes in real-time. Here, we characterize the naturally secreted Vargula luciferase as a secreted blood reporter and show that this reporter can be multiplexed with the secreted Gaussia luciferase and alkaline phosphatase for simultaneous monitoring of three different cellular processes in the same biological system. We applied this system to monitor the response of three different subsets of glioma cells to a clinically relevant chemotherapeutic agent in the same well in culture or animal in vivo. This system could be extended to any field to detect multiple processes in the same biological system and is amenable for high-throughput screening to find drugs that affect multiple cellular populations/phenomena simultaneously.


Subject(s)
Blood , Luciferases/blood , Animals , Cell Line, Tumor , Humans , Mice , Mice, Nude
7.
Mod Pathol ; 26(7): 922-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23429602

ABSTRACT

The histopathological diagnosis of diffuse gliomas often lacks the precision that is needed for tailored treatment of individual patients. Assessment of the molecular aberrations will probably allow more robust and prognostically relevant classification of these tumors. Markers that have gained a lot of interest in this respect are co-deletion of complete chromosome arms 1p and 19q, (hyper)methylation of the MGMT promoter and IDH1 mutations. The aim of this study was to assess the prognostic significance of complete 1p/19q co-deletion, MGMT promoter methylation and IDH1 mutations in patients suffering from diffuse gliomas. The presence of these molecular aberrations was investigated in a series of 561 diffuse astrocytic and oligodendroglial tumors (low grade n=110, anaplastic n=118 and glioblastoma n=333) and correlated with age at diagnosis and overall survival. Complete 1p/19q co-deletion, MGMT promoter methylation and/or IDH1 mutation generally signified a better prognosis for patients with a diffuse glioma including glioblastoma. However, in all 10 patients with a histopathological diagnosis of glioblastoma included in this study complete 1p/19q co-deletion was not associated with improved survival. Furthermore, in glioblastoma patients >50 years of age the favorable prognostic significance of IDH1 mutation and MGMT promoter methylation was absent. In conclusion, molecular diagnostics is a powerful tool to obtain prognostically relevant information for glioma patients. However, for individual patients the molecular information should be interpreted with caution and weighed in the context of parameters such as age and histopathological diagnosis.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Chromosome Deletion , Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 1 , Glioma/genetics , Adult , Brain Neoplasms/classification , Brain Neoplasms/mortality , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 19/genetics , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Female , Gene Deletion , Glioma/classification , Glioma/mortality , Humans , Isocitrate Dehydrogenase/genetics , Kaplan-Meier Estimate , Male , Middle Aged , Mutation , Pathology, Molecular , Prognosis , Promoter Regions, Genetic , Tumor Suppressor Proteins/genetics
8.
PLoS One ; 7(7): e41298, 2012.
Article in English | MEDLINE | ID: mdl-22844452

ABSTRACT

BACKGROUND: To investigate the dynamics of inter- and intratumoral molecular alterations during tumor progression in recurrent gliomas. METHODOLOGY/PRINCIPAL FINDINGS: To address intertumoral heterogeneity we investigated non-microdissected tumor tissue of 106 gliomas representing 51 recurrent tumors. To address intratumoral heterogeneity a set of 16 gliomas representing 7 tumor pairs with at least one recurrence, and 4 single mixed gliomas were investigated by microdissection of distinct oligodendroglial and astrocytic tumor components. All tumors and tumor components were analyzed for allelic loss of 1p/19q (LOH 1p/19q), for TP53- mutations and for R132 mutations in the IDH1 gene. The investigation of non-microdissected tumor tissue revealed clonality in 75% (38/51). Aberrant molecular alterations upon recurrence were detected in 25% (13/51). 64% (9/14) of these were novel and associated with tumor progression. Loss of previously detected alterations was observed in 36% (5/14). One tumor pair (1/14; 7%) was significant for both. Intratumoral clonality was detected in 57% (4/7) of the microdissected tumor pairs and in 75% (3/4) of single microdissected tumors. 43% (3/7) of tumor pairs and one single tumor (25%) revealed intratumoral heterogeneity. While intratumoral heterogeneity affected both the TP53- mutational status and the LOH1p/19q status, all tumors with intratumoral heterogeneity shared the R132 IDH1- mutation as a common feature in both their microdissected components. CONCLUSIONS/SIGNIFICANCE: The majority of recurrent gliomas are of monoclonal origin. However, the detection of divertive tumor cell clones in morphological distinct tumor components sharing IDH1- mutations as early event may provide insight into the tumorigenesis of true mixed gliomas.


Subject(s)
Astrocytoma/genetics , Astrocytoma/pathology , Isocitrate Dehydrogenase/genetics , Mutation , Oligodendroglioma/genetics , Oligodendroglioma/pathology , Adolescent , Adult , Child , Clone Cells/metabolism , Clone Cells/pathology , Codon/genetics , Disease Progression , Female , Humans , Loss of Heterozygosity/genetics , Male , Middle Aged , Recurrence , Tumor Suppressor Protein p53/genetics , Young Adult
9.
J Pathol ; 223(5): 626-34, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21341272

ABSTRACT

Currently available compounds that interfere with VEGF-A signalling effectively inhibit angiogenesis in gliomas, but influence diffuse infiltrative growth to a much lesser extent. Development of a functional tumour vascular bed not only involves VEGF-A but also requires platelet-derived growth factor receptor-ß (PDGFRß), which induces maturation of tumour blood vessels. Therefore, we tested whether combined inhibition of VEGFR and PDGFRß increases therapeutic benefit in the orthotopic glioma xenograft models E98 and E473, both displaying the diffuse infiltrative growth that is characteristically observed in most human gliomas. We used bevacizumab and vandetanib as VEGF(R) inhibitors, and sunitinib to additionally target PDGFRß. We show that combination therapy of sunitinib and vandetanib does not improve therapeutic efficacy compared to treatment with sunitinib, vandetanib or bevacizumab alone. Furthermore, all compounds induced reduction of vessel leakage in compact E98 tumour areas, resulting in decreased detectability of these mostly infiltrative xenografts in Gd-DTPA-enhanced MRI scans. These data show that inhibition of VEGF signalling cannot be optimized by additional PDGFR inhibition and support the concept that diffuse infiltrative areas in gliomas are resistant to anti-angiogenic therapy.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Glioma/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab , Blood-Brain Barrier/drug effects , Brain Neoplasms/blood supply , Glioma/blood supply , Humans , Indoles/administration & dosage , Indoles/pharmacology , Indoles/therapeutic use , Magnetic Resonance Imaging/methods , Mice , Mice, Nude , Neovascularization, Pathologic/drug therapy , Piperidines/administration & dosage , Piperidines/pharmacology , Piperidines/therapeutic use , Pyrroles/administration & dosage , Pyrroles/pharmacology , Pyrroles/therapeutic use , Quinazolines/administration & dosage , Quinazolines/pharmacology , Quinazolines/therapeutic use , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Signal Transduction/drug effects , Sunitinib , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays
10.
Pediatr Blood Cancer ; 56(1): 35-8, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20848662

ABSTRACT

BACKGROUND: In cancers like osteosarcoma with a 5-year overall survival of 50-60%, archived histological specimens can be a useful source of biological material. However, this material generally has been decalcified and formalin-fixed for many years. In our study, we investigated whether DNA obtained from these tissues can be used for reliable single nucleotide polymorphism (SNP) genotyping. PROCEDURE: We studied two SNPs in the drug transporter MDR1 using Taqman® SNP genotyping assays. Genotypes of the germ line DNA derived from freshly isolated DNA of 20 surviving patients with osteosarcoma were compared with genotypes obtained from archived material from decalcified formalin-fixed, paraffin-embedded (FFPE) blocks of the same patients. RESULTS: Decalcified FFPE-derived DNA yielded smaller PCR fragments compared to DNA extracted from peripheral blood cells, with a reliable size of ∼200 bp. However, we were able to evaluate each SNP in 19 of 20 cases included in this study. All successfully genotyped samples showed 100% concordance between genotypes obtained from DNA of FFPE tissue and the genotypes obtained from DNA of blood from the same patients. CONCLUSIONS: In conclusion, we have demonstrated that decalcified FFPE tissue can be used for genetic polymorphism analysis using Taqman® allelic discrimination assays. This forms a unique opportunity to combine new insights in genetic research with historical patient cohorts.


Subject(s)
DNA Mutational Analysis/methods , Osteosarcoma/genetics , DNA, Neoplasm/genetics , Genotype , Humans , Methods , Osteosarcoma/pathology , Paraffin Embedding , Polymorphism, Single Nucleotide , Tissue Fixation
11.
J Neurooncol ; 103(2): 221-30, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20820870

ABSTRACT

Despite similar morphological aspects, anaplastic oligodendroglial tumors (AOTs) form a heterogeneous clinical subgroup of gliomas. The chromosome arms 1p/19q codeletion has been shown to be a relevant biomarker in AOTs and to be perfectly exclusive from EGFR amplification in gliomas. To identify new genomic regions associated with prognosis, 60 AOTs from the EORTC trial 26951 were analyzed retrospectively using BAC-array-based comparative genomic hybridization. The data were processed using a binary tree method. Thirty-three BACs with prognostic value were identified distinguishing four genomic subgroups of AOTs with different prognosis (p < 0.0001). Type I tumors (25%) were characterized by: (1) an EGFR amplification, (2) a poor prognosis, (3) a higher rate of necrosis, and (4) an older age of patients. Type II tumors (21.7%) had: (1) loss of prognostic BACs located on 1p tightly associated with 19q deletion, (2) a longer survival, (3) an oligodendroglioma phenotype, and (4) a frontal location in brain. Type III AOTs (11.7%) exhibited: (1) a deletion of prognostic BACs located on 21q, and (2) a short survival. Finally, type IV tumors (41.7%) had different genomic patterns and prognosis than type I, II and III AOTs. Multivariate analysis showed that genomic type provides additional prognostic data to clinical, imaging and pathological features. Similar results were obtained in the cohort of 45 centrally reviewed-validated cases of AOTs. Whole genome analysis appears useful to screen the numerous genomic abnormalities observed in AOTs and to propose new biomarkers particularly in the non-1p/19q codeleted AOTs.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Chromosome Aberrations , Oligodendroglioma/genetics , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols , Biomarkers, Tumor/analysis , Brain Neoplasms/drug therapy , Brain Neoplasms/radiotherapy , Chromosomes, Artificial, Bacterial , Combined Modality Therapy , Comparative Genomic Hybridization , Humans , In Situ Hybridization, Fluorescence , Kaplan-Meier Estimate , Lomustine/administration & dosage , Male , Middle Aged , Multicenter Studies as Topic , Oligodendroglioma/drug therapy , Oligodendroglioma/radiotherapy , Procarbazine/administration & dosage , Prognosis , Proportional Hazards Models , Radiotherapy , Retrospective Studies , Treatment Outcome , Vincristine/administration & dosage , Young Adult
12.
Brain Pathol ; 21(3): 308-20, 2011 May.
Article in English | MEDLINE | ID: mdl-21029244

ABSTRACT

Up till now, typing and grading of diffuse gliomas is based on histopathological features. However, more objective tools are needed to improve reliable assessment of their biological behavior. We evaluated 331 diffuse gliomas for copy number changes involving 1p, 19q, CDKN2A, PTEN and EGFR(vIII) by Multiplex Ligation-dependent Probe Amplification (MLPA®, Amsterdam, The Netherlands). Specifically based on the co-occurrence of these aberrations we built a model for the timing of the different events and their exact nature (hemi- → homozygous loss; low-level gain → (high-copy) amplification) in the course of molecular progression. The mutation status of IDH1 and TP53 was also evaluated and shown to correlate with the level of molecular progression. The relevance of the proposed model was confirmed by analysis of 36 sets of gliomas and their 39 recurrence(s) whereas survival analysis for anaplastic gliomas confirmed the actual prognostic relevance of detecting molecular malignancy. Moreover, based on our results, molecular diagnostic analysis of 1p/19q can be further improved as different aberrations were identified, some of them being indicative for advanced molecular malignancy rather than for favorable tumor behavior. In conclusion, identification of molecular malignancy as proposed will aid in establishing a risk profile for individual patients and thereby in therapeutic decision making.


Subject(s)
Brain Neoplasms/genetics , Chromosomes, Human, Pair 19/genetics , Chromosomes, Human, Pair 1/genetics , DNA Copy Number Variations/genetics , Glioma/genetics , Neoplasm Proteins/genetics , Brain Neoplasms/classification , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Progression , ErbB Receptors/genetics , Glioma/classification , Humans , Models, Biological , Nucleic Acid Amplification Techniques , PTEN Phosphohydrolase/genetics , Pathology, Molecular/methods , Time Factors
13.
J Pathol ; 222(4): 324-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20976706

ABSTRACT

Recently, a new mechanism for activation of B-RAF was identified resulting from a tandem duplication, generating a fusion protein with constitutive BRAF activity and thereby activating the MAPK pathway. Different fusion variants involving BRAF and KIAA1549 were demonstrated, present in 80% of pilocytic astrocytomas in children. As the KIAA1549-BRAF fusion gene is detected at a much lower frequency in diffuse low-grade astrocytomas and survival was much longer than expected in the patients with a 'non-pilocytic' astrocytoma carrying the fusion gene, identification of this fusion gene can be of diagnostic and prognostic value. In the near future, interference with the (fusion gene causing) activation of the MAPK signalling cascade may open new therapeutic avenues for children with pilocytic astrocytomas, as a first line of defence against tumour growth or in situations where the tumour has become refractory to other therapeutic modalities.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , MAP Kinase Signaling System/genetics , Oncogene Fusion/physiology , Proto-Oncogene Proteins B-raf/genetics , Astrocytoma/diagnosis , Astrocytoma/therapy , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Child , Humans , MAP Kinase Signaling System/physiology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/physiology , Prognosis
14.
Acta Neuropathol ; 120(5): 567-84, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20714900

ABSTRACT

Modern neuropathology serves a key function in the multidisciplinary management of brain tumor patients. Owing to the recent advancements in molecular neurooncology, the neuropathological assessment of brain tumors is no longer restricted to provide information on a tumor's histological type and malignancy grade, but may be complemented by a growing number of molecular tests for clinically relevant tissue-based biomarkers. This article provides an overview and critical appraisal of the types of genetic and epigenetic aberrations that have gained significance in the molecular diagnostics of gliomas, namely deletions of chromosome arms 1p and 19q, promoter hypermethylation of the O6-methylguanine-methyl-transferase (MGMT) gene, and the mutation status of the IDH1 and IDH2 genes. In addition, the frequent oncogenic aberration of BRAF in pilocytic astrocytomas may serve as a novel diagnostic marker and therapeutic target. Finally, this review will summarize recent mechanistic insights into the molecular alterations underlying treatment resistance in malignant gliomas and outline the potential of genome-wide profiling approaches for increasing our repertoire of clinically useful glioma markers.


Subject(s)
Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Epigenomics/methods , Glioma/diagnosis , Glioma/genetics , Pathology, Molecular/methods , Humans , Molecular Biology/methods , Pathology, Molecular/trends
15.
Acta Neurol Belg ; 109(3): 238-42, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19902821

ABSTRACT

Glioblastoma (GBM) is the most malignant primary brain tumour in adults. Since 2005 surgery followed by radiotherapy with concomitant Temozolomide (TMZ) is the standard care for patients with a GBM. Despite these improved treatment strategies, survival of GBM-patients remains poor; and there are very few patients who survive for a long time. Also there is no standard therapeutic strategy after six cycles of TMZ, and further treatment is at the physician's discretion. We report a case of a young patient with a glioblastoma who, not only showed dramatic clinical and radiological improvement after TMZ treatment but who now also (under continued TMZ therapy) survives over 6 years, with complete remission clinically and radiologically. Up till now there are no studies describing TMZ treatment in GBM patients for as long as 6 years.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Dacarbazine/analogs & derivatives , Glioblastoma/drug therapy , Adult , Brain Neoplasms/pathology , Dacarbazine/therapeutic use , Female , Glioblastoma/pathology , Humans , Magnetic Resonance Imaging , Survivors , Temozolomide , Treatment Outcome
16.
J Clin Oncol ; 27(35): 5881-6, 2009 Dec 10.
Article in English | MEDLINE | ID: mdl-19901104

ABSTRACT

PURPOSE: O6-methylguanine-methyltransferase (MGMT) promoter methylation has been shown to predict survival of patients with glioblastomas if temozolomide is added to radiotherapy (RT). It is unknown if MGMT promoter methylation is also predictive to outcome to RT followed by adjuvant procarbazine, lomustine, and vincristine (PCV) chemotherapy in patients with anaplastic oligodendroglial tumors (AOT). PATIENTS AND METHODS: In the European Organisation for the Research and Treatment of Cancer study 26951, 368 patients with AOT were randomly assigned to either RT alone or to RT followed by adjuvant PCV. From 165 patients of this study, formalin-fixed, paraffin-embedded tumor tissue was available for MGMT promoter methylation analysis. This was investigated with methylation specific multiplex ligation-dependent probe amplification. RESULTS: In 152 cases, an MGMT result was obtained, in 121 (80%) cases MGMT promoter methylation was observed. Methylation strongly correlated with combined loss of chromosome 1p and 19q loss (P = .00043). In multivariate analysis, MGMT promoter methylation, 1p/19q codeletion, tumor necrosis, and extent of resection were independent prognostic factors. The prognostic significance of MGMT promoter methylation was equally strong in the RT arm and the RT/PCV arm for both progression-free survival and overall survival. In tumors diagnosed at central pathology review as glioblastoma, no prognostic effect of MGMT promoter methylation was observed. CONCLUSION: In this study, on patients with AOT MGMT promoter methylation was of prognostic significance and did not have predictive significance for outcome to adjuvant PCV chemotherapy. The biologic effect of MGMT promoter methylation or pathogenetic features associated with MGMT promoter methylation may be different for AOT compared with glioblastoma.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Astrocytoma/drug therapy , Brain Neoplasms/drug therapy , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Oligodendroglioma/drug therapy , Promoter Regions, Genetic , Tumor Suppressor Proteins/genetics , Astrocytoma/genetics , Astrocytoma/mortality , Astrocytoma/pathology , Astrocytoma/radiotherapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Chemotherapy, Adjuvant , Chromosome Deletion , Chromosomes, Human, Pair 1 , Chromosomes, Human, Pair 19 , CpG Islands , Disease-Free Survival , Europe/epidemiology , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lomustine/administration & dosage , Middle Aged , Oligodendroglioma/genetics , Oligodendroglioma/pathology , Oligodendroglioma/radiotherapy , Polymerase Chain Reaction/methods , Procarbazine/administration & dosage , Proportional Hazards Models , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome , Vincristine/administration & dosage
17.
Brain Pathol ; 19(4): 661-71, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19744038

ABSTRACT

Epidermal growth factor receptor (EGFR) is commonly affected in cancer, generally in the form of an increase in DNA copy number and/or as mutation variants [e.g., EGFR variant III (EGFRvIII), an in-frame deletion of exons 2-7]. While detection of EGFR aberrations can be expected to be relevant for glioma patients, such analysis has not yet been implemented in a routine setting, also because feasible and robust assays were lacking. We evaluated multiplex ligation-dependent probe amplification (MLPA) for detection of EGFR amplification and EGFRvIII in DNA of a spectrum of 216 diffuse gliomas. EGFRvIII detection was verified at the protein level by immunohistochemistry and at the RNA level using the conventionally used endpoint RT-PCR as well as a newly developed quantitative RT-PCR. Compared to these techniques, the DNA-based MLPA assay for EGFR/EGFRvIII analysis tested showed 100% sensitivity and specificity. We conclude that MLPA is a robust assay for detection of EGFR/EGFRvIII aberrations. While the exact diagnostic, prognostic and predictive value of such EGFR testing remains to be seen, MLPA has great potential as it can reliably and relatively easily be performed on routinely processed (formalin-fixed, paraffin-embedded) tumor tissue in combination with testing for other relevant glioma markers.


Subject(s)
ErbB Receptors/genetics , Glioma/diagnosis , Glioma/genetics , Molecular Diagnostic Techniques/methods , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Gene Amplification , Gene Dosage , Humans , Immunohistochemistry/methods , Mutation , Predictive Value of Tests , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction/methods , Sensitivity and Specificity
18.
Acta Neuropathol ; 118(4): 469-74, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19554337

ABSTRACT

Somatic mutations in the IDH1 gene encoding cytosolic NADP+-dependent isocitrate dehydrogenase have been shown in the majority of astrocytomas, oligodendrogliomas and oligoastrocytomas of WHO grades II and III. IDH2 encoding mitochondrial NADP+-dependent isocitrate dehydrogenase is also mutated in these tumors, albeit at much lower frequencies. Preliminary data suggest an importance of IDH1 mutation for prognosis showing that patients with anaplastic astrocytomas, oligodendrogliomas and oligoastrocytomas harboring IDH1 mutations seem to fare much better than patients without this mutation in their tumors. To determine mutation types and their frequencies, we examined 1,010 diffuse gliomas. We detected 716 IDH1 mutations and 31 IDH2 mutations. We found 165 IDH1 (72.7%) and 2 IDH2 mutations (0.9%) in 227 diffuse astrocytomas WHO grade II, 146 IDH1 (64.0%) and 2 IDH2 mutations (0.9%) in 228 anaplastic astrocytomas WHO grade III, 105 IDH1 (82.0%) and 6 IDH2 mutations (4.7%) in 128 oligodendrogliomas WHO grade II, 121 IDH1 (69.5%) and 9 IDH2 mutations (5.2%) in 174 anaplastic oligodendrogliomas WHO grade III, 62 IDH1 (81.6%) and 1 IDH2 mutations (1.3%) in 76 oligoastrocytomas WHO grade II and 117 IDH1 (66.1%) and 11 IDH2 mutations (6.2%) in 177 anaplastic oligoastrocytomas WHO grade III. We report on an inverse association of IDH1 and IDH2 mutations in these gliomas and a non-random distribution of the mutation types within the tumor entities. IDH1 mutations of the R132C type are strongly associated with astrocytoma, while IDH2 mutations predominantly occur in oligodendroglial tumors. In addition, patients with anaplastic glioma harboring IDH1 mutations were on average 6 years younger than those without these alterations.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Adult , Age Factors , Brain/pathology , Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Cell Differentiation , DNA Mutational Analysis , Female , Glioma/enzymology , Glioma/pathology , Humans , Male , Middle Aged , Mutation , Prognosis , Tumor Cells, Cultured
19.
Neuropathology ; 28(4): 440-3, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18312547

ABSTRACT

The chromosome (chr) 1p deletion is a favorable biomarker in oligodendroglial tumors and is even more powerful a marker when combined with chr 19q loss. As a result, the 1p deletion is taken into account more and more in clinical trials and the management of patients. However, the laboratory technique implemented for detection of this biomarker has been a topic of debate. To illustrate the usefulness of evaluating multiple loci, we here report two anaplastic oligodendrogliomas that were investigated using fluorescent in situ hybridization (FISH) and bacterial artificial chromosome (BAC)-array-based comparative genomic hybridization (aCGH). Indeed, segmental analysis using FISH, limited to chr 1p36 was unable to discriminate between complete and partial deletions of chrs 1p. However, complete and partial deletions of 1p are reported to have distinct clinical outcomes. Our results illustrate that aCGH (or other multiple loci technologies) provide complementary information to single locus technologies such as FISH because multiple loci technologies can evaluate the extent of the chr 1p deletion.


Subject(s)
Brain Neoplasms/genetics , Chromosome Deletion , Chromosomes, Human, Pair 1/genetics , Nucleic Acid Hybridization/methods , Oligodendroglioma/genetics , Biomarkers, Tumor/genetics , Chromosomes, Artificial, Bacterial , Humans , In Situ Hybridization, Fluorescence , Middle Aged
20.
Brain Pathol ; 18(3): 423-33, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18371177

ABSTRACT

Most human gliomas are characterized by diffuse infiltrative growth in the brain parenchyma. Partly because of this characteristic growth pattern, gliomas are notorious for their poor response to current therapies. Many animal models for human gliomas, however, do not display this diffuse infiltrative growth pattern. Furthermore, there is a need for glioma models that represent adequate genocopies of different subsets of human gliomas (e.g., oligodendrogliomas). Here, we assessed the intracerebral growth patterns and copy number changes [using multiplex ligation-dependent probe amplification (MLPA)/comparative genomic hybridization (CGH)] of 15 human glioma lines in nude mice. Most xenografts present with compact growing lesions intracerebrally. Only the E98 and, to a lesser degree, E106 xenograft lines (propagated through subcutaneous growth) consistently produced intracerebral tumors, displaying diffuse infiltrative growth in the brain parenchyma. In contrast, four xenograft lines (E434, E468, E473 and E478), established by direct intracerebral inoculation of human glioma cells and serially propagated intracerebrally, consistently showed extensive diffuse infiltration throughout the brain. After several passages, the neoplastic cells still carry typical chromosomal aberrations [(-1p/-19q in oligodendroglioma, +7/-10 in glioblastoma multiforme (GBM)]. Especially these latter four models and the E98 line thus represent adequate geno- and phenocopies of human gliomas and form an attractive platform to investigate different therapeutic approaches in a preclinical setting.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Disease Models, Animal , Glioma/genetics , Glioma/pathology , Animals , Cell Line, Tumor , Female , Genotype , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Nude , Microscopy, Electron, Transmission , Phenotype , RNA, Messenger/analysis , Vascular Endothelial Growth Factor A/biosynthesis , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL