Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 123
Filter
1.
Ann Hematol ; 101(3): 607-616, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34982196

ABSTRACT

Primary central nervous system non-Hodgkin lymphomas (PCNS-NHLs) are extranodal B-cell lymphomas with poor prognosis. The role of high-dose therapy (HDT) followed by autologous blood stem cell transplantation (ASCT) as first-line therapy is still not clear. We retrospectively collected long-term follow up data of 61 consecutive patients with PCNS-NHL at the University Hospital Düsseldorf from January 2004 to December 2016. Thirty-six patients were treated with conventional chemoimmunotherapy (cCIT) only (CT-group). Seventeen patients received an induction cCIT followed by HDT and ASCT. In the CT-group, the overall response rate (ORR) was 61% (CR 47%, PR 14%), and there were 8% treatment-related deaths (TRD). Progression-free survival (PFS) was 31.8 months, and overall survival (OS) was 57.3 months. In the HDT-group, the ORR was 88% (59% CR, 29% PR), and there were 6% TRD. Median PFS and OS were not reached at 5 years. The 5-year PFS and OS were 64.7%. After a median follow up of 71 months, 10 patients (59%) were still alive in CR/PR following HDT and ASCT, one patient was treated for progressive disease (PD), and 7 had died (41%, 6 PD, 1 TRD). All patients achieving CR prior to HDT achieved durable CR. In the CT-group, 8 patients (22%) were alive in CR/PR after a median follow-up of 100 months. Twenty-eight patients died (78%, 24 PD, 2 TRD, 2 deaths in remission). In the univariate analysis, the HDT-group patients had significantly better PFS (not reached vs 31.8 months, p = 0.004) and OS (not reached vs 57.3 months, p = 0.021). The multivariate analysis showed HDT was not predictive for survival. Treatment with HDT + ASCT is feasible and offers the chance for long-term survival with low treatment-related mortality in younger patients. In this analysis, ORR, PFS and OS were better with HDT than with conventional cCIT alone. This result was not confirmed in the multivariate analysis, and further studies need to be done to examine the role of HDT in PCNSL.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Central Nervous System Neoplasms/therapy , Hematopoietic Stem Cell Transplantation , Lymphoma, Non-Hodgkin/therapy , Aged , Female , Humans , Male , Middle Aged , Progression-Free Survival , Retrospective Studies , Transplantation, Autologous
2.
Biomed Pharmacother ; 144: 112278, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34628166

ABSTRACT

The utility of patient-derived tumor cell lines as experimental models for glioblastoma has been challenged by limited representation of the in vivo tumor biology and low clinical translatability. Here, we report on longitudinal epigenetic and transcriptional profiling of seven glioblastoma spheroid cell line models cultured over an extended period. Molecular profiles were associated with drug response data obtained for 231 clinically used drugs. We show that the glioblastoma spheroid models remained molecularly stable and displayed reproducible drug responses over prolonged culture times of 30 in vitro passages. Integration of gene expression and drug response data identified predictive gene signatures linked to sensitivity to specific drugs, indicating the potential of gene expression-based prediction of glioblastoma therapy response. Our data thus empowers glioblastoma spheroid disease modeling as a useful preclinical assay that may uncover novel therapeutic vulnerabilities and associated molecular alterations.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Brain Neoplasms/drug therapy , Cell Proliferation/drug effects , Genomic Instability , Glioma/drug therapy , Transcriptome , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , DNA Mutational Analysis , Drug Screening Assays, Antitumor , Gene Expression Profiling , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Humans , Mutation , Reproducibility of Results , Spheroids, Cellular , Time Factors
3.
Neuropathol Appl Neurobiol ; 47(1): 108-126, 2021 02.
Article in English | MEDLINE | ID: mdl-32696543

ABSTRACT

AIMS: We aimed to reclassify a population-based cohort of 529 adult glioma patients to evaluate the prognostic impact of the 2016 World Health Organization (WHO) central nervous system tumour classification. Moreover, we evaluated the feasibility of gene panel next-generation sequencing (NGS) in daily diagnostics of 225 prospective glioma patients. METHODS: The retrospective cohort was reclassified according to WHO 2016 criteria by immunohistochemistry for IDH-R132H, fluorescence in situ hybridization for 1p/19q-codeletion and gene panel NGS. All tumours of the prospective cohort were subjected to NGS analysis up-front. RESULTS: The entire population-based cohort was successfully reclassified according to WHO 2016 criteria. NGS results were obtained for 98% of the prospective patients. Survival analyses in the population-based cohort confirmed three major prognostic subgroups, that is, isocitrate dehydrogenase (IDH)-mutant and 1p/19q-codeleted oligodendrogliomas, IDH-mutant astrocytomas and IDH-wildtype glioblastomas. The distinction between WHO grade II and III was prognostic in patients with IDH-mutant astrocytoma. The survival of patients with IDH-wildtype diffuse astrocytomas carrying TERT promoter mutation and/or EGFR amplification overlapped with the poor survival of IDH-wildtype glioblastoma patients. CONCLUSIONS: Gene panel NGS proved feasible in daily diagnostics. In addition, our study confirms the prognostic role of glioma classification according to WHO 2016 in a large population-based cohort. Molecular features of glioblastoma in IDH-wildtype diffuse glioma were linked to poor survival corresponding to IDH-wildtype glioblastoma patients. The distinction between WHO grade II and III retained prognostic significance in patients with IDH-mutant diffuse astrocytic gliomas.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/diagnosis , Glioma/pathology , High-Throughput Nucleotide Sequencing , Adolescent , Adult , Aged , Aged, 80 and over , Astrocytoma/diagnosis , Astrocytoma/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Female , Glioblastoma/diagnosis , Glioblastoma/genetics , Glioma/diagnosis , Glioma/genetics , Humans , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , Mutation/genetics , Prognosis , Telomerase/genetics , Young Adult
5.
Pathologe ; 40(Suppl 1): 9-17, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31025086

ABSTRACT

BACKGROUND: Diffuse astrocytic and oligodendroglial gliomas are the most common neuroepithelial tumors. Their classification is based on the integration of histological and molecular findings according to the classification of tumors of the central nervous system published by the World Health Organization (WHO) in 2016. OBJECTIVES: This review describes the different entities and variants of diffuse gliomas and summarizes the current diagnostic criteria for these tumors. MATERIALS AND METHODS: Based on the 2016 WHO classification and selected other publications, the histomolecular diagnostics of diffuse gliomas is presented and illustrated. RESULTS: Diffuse gliomas are divided into isocitrate dehydrogenase (IDH)-mutant or IDH-wildtype gliomas by detection of mutations in the IDH1 or IDH2 genes. Among the IDH-mutant gliomas, oligodendroglial tumors are characterized by combined losses of chromosome arms 1p and 19q. Loss of nuclear expression of the ATRX protein is a marker of IDH- mutant astrocytic gliomas. Glioblastoma, IDH-wildtype, is the most common diffuse glioma. Diffuse and anaplastic astrocytic gliomas without IDH mutation should be further evaluated for molecular features of glioblastoma, IDH-wildtype. Diffuse gliomas in the thalamus, brainstem, or spinal cord carrying a histone 3 (H3)-K27M mutation are classified as diffuse midline gliomas, H3-K27M-mutant. By determining the IDH and 1p/19q status, oligoastrocytomas can be stratified into either astrocytic or oligodendroglial gliomas. Gliomatosis cerebri is no longer regarded as a distinct glioma entity. CONCLUSIONS: Diffuse gliomas can today be classified accurately and reproducibly by means of histological, immunohistochemical, and molecular analyses.


Subject(s)
Astrocytes/pathology , Brain Neoplasms/diagnosis , Glioma/diagnosis , Oligodendroglioma/pathology , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Glioma/enzymology , Glioma/genetics , Glioma/pathology , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutation , X-linked Nuclear Protein/metabolism
6.
Pathologe ; 40(2): 131-139, 2019 Mar.
Article in German | MEDLINE | ID: mdl-30790013

ABSTRACT

BACKGROUND: Diffuse astrocytic and oligodendroglial gliomas are the most common neuroepithelial tumors. Their classification is based on the integration of histological and molecular findings according to the classification of tumors of the central nervous system published by the World Health Organization (WHO) in 2016. OBJECTIVES: This review describes the different entities and variants of diffuse gliomas and summarizes the current diagnostic criteria for these tumors. MATERIALS AND METHODS: Based on the 2016 WHO classification and selected other publications, the histomolecular diagnostics of diffuse gliomas is presented and illustrated. RESULTS: Diffuse gliomas are divided into isocitrate dehydrogenase (IDH)-mutant or IDH-wildtype gliomas by detection of mutations in the IDH1 or IDH2 genes. Among the IDH-mutant gliomas, oligodendroglial tumors are characterized by combined losses of chromosome arms 1p and 19q. Loss of nuclear expression of the ATRX protein is a marker of IDH- mutant astrocytic gliomas. Glioblastoma, IDH-wildtype, is the most common diffuse glioma. Diffuse and anaplastic astrocytic gliomas without IDH mutation should be further evaluated for molecular features of glioblastoma, IDH-wildtype. Diffuse gliomas in the thalamus, brainstem, or spinal cord carrying a histone 3 (H3)-K27M mutation are classified as diffuse midline gliomas, H3-K27M-mutant. By determining the IDH and 1p/19q status, oligoastrocytomas can be stratified into either astrocytic or oligodendroglial gliomas. Gliomatosis cerebri is no longer regarded as a distinct glioma entity. CONCLUSIONS: Diffuse gliomas can today be classified accurately and reproducibly by means of histological, immunohistochemical, and molecular analyses.


Subject(s)
Brain Neoplasms , Glioma , Oligodendroglioma , Humans , Isocitrate Dehydrogenase , Mutation , X-linked Nuclear Protein
7.
Neuropathol Appl Neurobiol ; 45(5): 441-458, 2019 08.
Article in English | MEDLINE | ID: mdl-30548945

ABSTRACT

AIMS: Aberrant expression of microRNAs (miRNAs) is frequent in various cancers including gliomas. We aimed to characterize the role of miR-16-5p as a candidate tumour suppressor miRNA in gliomas. METHODS: Real-time PCR-based approaches were used for miRNA and mRNA expression profiling of glioma and non-neoplastic brain tissues as well as glioma cell lines. Protein levels were determined by Western blotting. In vitro analyses were performed following overexpression of miR-16-5p, trichostatin A (TSA) treatment, and siRNA-mediated knock-down of HDAC3 in glioma cells. Effects of miR-16-5p on glioma cell viability, apoptosis and response to irradiation and temozolomide (TMZ) were assessed. RESULTS: Expression of miR-16-5p was reduced relative to control brain tissue in isocitrate dehydrogenase (IDH)-mutant astrocytomas of World Health Organization (WHO) grades II, III and IV, and a subset of IDH-wildtype glioblastomas WHO grade IV. MiR-16-5p expression was lower in IDH-mutant than in IDH-wildtype gliomas, and down-regulated in IDH-wildtype glioma lines. MiR-16-5p overexpression reduced expression of important cell cycle and apoptosis regulators in glioma cells, including CDK6, CDC25A, CCND3, CCNE1, WEE1, CHEK1, BCL2 and MCL1. In line, CDK6, WEE1, CHEK1, BCL2 and MCL1 transcript levels were increased in WHO grade III or IV gliomas. TSA treatment and HDAC3 knockdown in glioma cells induced miR-16-5p up-regulation and reduced expression of its targets. Moreover, miR-16-5p overexpression inhibited proliferation and induced apoptosis in various glioma cell lines and increased sensitivity of A172 glioma cells to irradiation and TMZ. CONCLUSION: Reduced expression of miR-16-5p contributes to glioma cell proliferation, survival and resistance to cytotoxic therapy.


Subject(s)
Brain Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , MicroRNAs/genetics , Apoptosis/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Down-Regulation , Glioma/pathology , Humans
8.
Ann Oncol ; 29(6): 1423-1430, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29648580

ABSTRACT

Background: The addition of bevacizumab to temozolomide-based chemoradiotherapy (TMZ/RT â†’ TMZ) did not prolong overall survival (OS) in patients with newly diagnosed glioblastoma in phase III trials. Elderly and frail patients are underrepresented in clinical trials, but early reports suggested preferential benefit in this population. Patients and methods: ARTE was a 2 : 1 randomized, multi-center, open-label, non-comparative phase II trial of hypofractionated RT (40 Gy in 15 fractions) with bevacizumab (10 mg/kg×14 days) (arm A, N = 50) or without bevacizumab (arm B, N = 25) in patients with newly diagnosed glioblastoma aged ≥65 years. The primary objective was to obtain evidence for prolongation of median OS by the addition of bevacizumab to RT. Response was assessed by RANO criteria. Quality of life (QoL) was monitored by the EORTC QLQ-C30/BN20 modules. Exploratory studies included molecular subtyping by 450k whole methylome and gene expression analyses. Results: Median PFS was longer in arm A than in arm B (7.6 and 4.8 months, P = 0.003), but OS was similar (12.1 and 12.2 months, P = 0.77). Clinical deterioration was delayed and more patients came off steroids in arm A. Prolonged PFS in arm A was confined to tumors with the receptor tyrosine kinase (RTK) I methylation subtype (HR 0.25, P = 0.014) and proneural gene expression (HR 0.29, P = 0.025). In a Cox model of OS controlling for established prognostic factors, associations with more favorable outcome were identified for age <70 years (HR 0.52, P = 0.018) and Karnofsky performance score 90%-100% (HR 0.51, P = 0.026). Including molecular subtypes into that model identified an association of the RTK II gene methylation subtype with inferior OS (HR 1.73, P = 0.076). Conclusion: Efficacy outcomes and exploratory analyses of ARTE do not support the hypothesis that the addition of bevacizumab to RT generally prolongs survival in elderly glioblastoma patients. Molecular biomarkers may identify patients with preferential benefit from bevacizumab. Clinical trial registration number: NCT01443676.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Bevacizumab/therapeutic use , Chemoradiotherapy/mortality , Glioblastoma/drug therapy , Glioblastoma/radiotherapy , Quality of Life , Radiation Dose Hypofractionation , Aged , Aged, 80 and over , Female , Follow-Up Studies , Glioblastoma/pathology , Humans , Male , Prognosis , Survival Rate
9.
Virchows Arch ; 472(4): 557-565, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29374318

ABSTRACT

The simultaneous detection of multiple somatic mutations in the context of molecular diagnostics of cancer is frequently performed by means of amplicon-based targeted next-generation sequencing (NGS). However, only few studies are available comparing multicenter testing of different NGS platforms and gene panels. Therefore, seven partner sites of the German Cancer Consortium (DKTK) performed a multicenter interlaboratory trial for targeted NGS using the same formalin-fixed, paraffin-embedded (FFPE) specimen of molecularly pre-characterized tumors (n = 15; each n = 5 cases of Breast, Lung, and Colon carcinoma) and a colorectal cancer cell line DNA dilution series. Detailed information regarding pre-characterized mutations was not disclosed to the partners. Commercially available and custom-designed cancer gene panels were used for library preparation and subsequent sequencing on several devices of two NGS different platforms. For every case, centrally extracted DNA and FFPE tissue sections for local processing were delivered to each partner site to be sequenced with the commercial gene panel and local bioinformatics. For cancer-specific panel-based sequencing, only centrally extracted DNA was analyzed at seven sequencing sites. Subsequently, local data were compiled and bioinformatics was performed centrally. We were able to demonstrate that all pre-characterized mutations were re-identified correctly, irrespective of NGS platform or gene panel used. However, locally processed FFPE tissue sections disclosed that the DNA extraction method can affect the detection of mutations with a trend in favor of magnetic bead-based DNA extraction methods. In conclusion, targeted NGS is a very robust method for simultaneous detection of various mutations in FFPE tissue specimens if certain pre-analytical conditions are carefully considered.


Subject(s)
Biomarkers, Tumor/genetics , DNA, Neoplasm/analysis , High-Throughput Nucleotide Sequencing/methods , Neoplasms/genetics , Humans , Pathology, Molecular/methods , Pathology, Molecular/standards , Reproducibility of Results , Translational Research, Biomedical/methods
10.
Nat Commun ; 7: 12329, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27470974

ABSTRACT

Diffuse invasion of the surrounding brain parenchyma is a major obstacle in the treatment of gliomas with various therapeutics, including anti-angiogenic agents. Here we identify the epi-/genetic and microenvironmental downregulation of ephrinB2 as a crucial step that promotes tumour invasion by abrogation of repulsive signals. We demonstrate that ephrinB2 is downregulated in human gliomas as a consequence of promoter hypermethylation and gene deletion. Consistently, genetic deletion of ephrinB2 in a murine high-grade glioma model increases invasion. Importantly, ephrinB2 gene silencing is complemented by a hypoxia-induced transcriptional repression. Mechanistically, hypoxia-inducible factor (HIF)-1α induces the EMT repressor ZEB2, which directly downregulates ephrinB2 through promoter binding to enhance tumour invasiveness. This mechanism is activated following anti-angiogenic treatment of gliomas and is efficiently blocked by disrupting ZEB2 activity. Taken together, our results identify ZEB2 as an attractive therapeutic target to inhibit tumour invasion and counteract tumour resistance mechanisms induced by anti-angiogenic treatment strategies.


Subject(s)
Drug Resistance, Neoplasm , Ephrin-B2/genetics , Glioma/genetics , Glioma/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Zinc Finger E-box Binding Homeobox 2/metabolism , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Cell Hypoxia/genetics , Down-Regulation/genetics , Drug Resistance, Neoplasm/genetics , Ephrin-B2/metabolism , Gene Expression Regulation, Neoplastic , Glioma/blood supply , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Up-Regulation/genetics , Xenograft Model Antitumor Assays , Zinc Finger E-box Binding Homeobox 2/genetics
11.
Oncogene ; 35(24): 3163-77, 2016 06 16.
Article in English | MEDLINE | ID: mdl-26549022

ABSTRACT

Glioblastoma is the most aggressive primary brain tumor in adults. Although the rapid recurrence of glioblastomas after treatment is a major clinical challenge, the relationships between tumor growth and intracerebral spread remain poorly understood. We have identified the cofilin phosphatase chronophin (gene name: pyridoxal phosphatase, PDXP) as a glial tumor modifier. Monoallelic PDXP loss was frequent in four independent human astrocytic tumor cohorts and increased with tumor grade. We found that aberrant PDXP promoter methylation can be a mechanism leading to further chronophin downregulation in glioblastomas, which correlated with shorter glioblastoma patient survival. Moreover, we observed an inverse association between chronophin protein expression and cofilin phosphorylation levels in glioma tissue samples. Chronophin-deficient glioblastoma cells showed elevated cofilin phosphorylation, an increase in polymerized actin, a higher directionality of cell migration, and elevated in vitro invasiveness. Tumor growth of chronophin-depleted glioblastoma cells xenografted into the immunodeficient mouse brain was strongly impaired. Our study suggests a mechanism whereby the genetic and epigenetic alterations of PDXP resulting in altered chronophin expression may regulate the interplay between glioma cell proliferation and invasion.


Subject(s)
Brain Neoplasms/enzymology , Brain Neoplasms/pathology , Glioblastoma/enzymology , Glioblastoma/pathology , Phosphoprotein Phosphatases/metabolism , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Proliferation/physiology , DNA Methylation , Female , Glioblastoma/genetics , Heterografts , Humans , Mice , Mice, Inbred NOD , Neoplasm Invasiveness , Phosphoprotein Phosphatases/genetics , Promoter Regions, Genetic
12.
Nervenarzt ; 86(6): 672, 674-6, 678-80, passim, 2015 Jun.
Article in German | MEDLINE | ID: mdl-25989737

ABSTRACT

The diagnostic subdivision of gliomas is traditionally based on histological features as defined by the World Health Organization (WHO) classification of tumors of the central nervous system. In recent years molecular studies have identified a number of genetic and epigenetic markers that could contribute to an improved tumor classification and better prediction of response to therapy and prognosis in the individual patient. The most important molecular tests with differential diagnostic relevance in patients with astrocytic and oligodendroglial tumors include the detection of genetic mutations in the isocitrate dehydrogenase 1 (IDH1), IDH2, alpha thalassemia/mental retardation syndrome X-linked (ATRX), histone H3.3 (H3F3A) and v-raf murine sarcoma viral oncogene homolog B (BRAF) genes as well as the demonstration of codeletions of chromosomal arms 1p and 19q. Important predictive markers that have been linked to the response to alkylating chemotherapy are O-6-methylguanine-DNA methyltransferase (MGMT) promoter methylation in glioblastoma patients and 1p/19q codel status in anaplastic glioma patients. Oncogenic c11orf95/RELA fusion gene formation is characteristic for a subgroup of patients with supratentorial ependymoma. In addition to diagnostic testing of individual genes, novel microarray and next generation sequencing (NGS) techniques show promising perspectives in glioma diagnostics. The assessment of DNA methylation profiles using DNA methylation arrays representing 450,000 CpG dinucleotides distributed throughout the human genome (450 k array test) now allows the robust molecular classification of gliomas into clinically relevant entities and variants. Moreover, glioma-associated gene panel NGS promises the timely parallel sequencing of relevant diagnostic and predictive marker genes in a single test. It will now be a major task to integrate these novel results and techniques into the conventional histological procedures in the up-coming revision of the WHO classification.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Genetic Testing/methods , Glioma/diagnosis , Glioma/genetics , Brain Neoplasms/classification , DNA Mutational Analysis/methods , Gene Expression Profiling/methods , Genetic Markers/genetics , Genetic Predisposition to Disease/genetics , Glioma/classification , Humans , Molecular Diagnostic Techniques/methods , Polymorphism, Single Nucleotide/genetics
13.
Ann Oncol ; 24(12): 3117-23, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24130262

ABSTRACT

BACKGROUND: This prospective multicenter study assessed the prognostic influence of the extent of resection when compared with biopsy only in a contemporary patient population with newly diagnosed glioblastoma. PATIENTS AND METHODS: Histology, O(6)-methylguanine-DNA methyltransferase (MGMT) promoter methylation status, and clinical data were centrally analyzed. Survival analyses were carried out with the Kaplan-Meier method. Prognostic factors were assessed with proportional hazard models. RESULTS: Of 345 patients, 273 underwent open tumor resection and 72 biopsies; 125 patients had gross total resections (GTRs) and 148, incomplete resections. Surgery-related morbidity was lower after biopsy (1.4% versus 12.1%, P = 0.007). 64.3% of patients received radiotherapy and chemotherapy (RT plus CT), 20.0% RT alone, 4.3% CT alone, and 11.3% best supportive care as an initial treatment. Patients ≤60 years with a Karnofsky performance score (KPS) of ≥90 were more likely to receive RT plus CT (P < 0.01). Median overall survival (OS) (progression free survival; PFS) ranged from 33.2 months (15 months) for patients with MGMT-methylated tumors after GTR and RT plus CT to 3.0 months (2.4 months) for biopsied patients receiving supportive care only. Favorable prognostic factors in multivariate analyses for OS were age ≤60 years [hazard ratio (HR) = 0.52; P < 0.001], preoperative KPS of ≥80 (HR = 0.55; P < 0.001), GTR (HR = 0.60; P = 0.003), MGMT promoter methylation (HR = 0.44; P < 0.001), and RT plus CT (HR = 0.18, P < 0.001); patients undergoing incomplete resection did not better than those receiving biopsy only (HR = 0.85; P = 0.31). CONCLUSIONS: The value of incomplete resection remains questionable. If GTR cannot be safely achieved, biopsy only might be used as an alternative surgical strategy.


Subject(s)
Brain Neoplasms/surgery , Glioblastoma/surgery , Adult , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Chemoradiotherapy , Disease-Free Survival , Female , Glioblastoma/mortality , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Prognosis , Proportional Hazards Models , Prospective Studies , Treatment Outcome , Young Adult
14.
Br J Neurosurg ; 27(6): 772-8, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23662801

ABSTRACT

The prognostic role of O(6)-methylguanine-DNA methyltransferase (MGMT) promoter methylation in glioblastoma patients treated with carmustine (BCNU) wafer implantation is unclear. Here, we report on a retrospective study of 47 patients with either newly diagnosed (30 patients) or recurrent (17 patients) glioblastoma (WHO grade IV) treated with BCNU (bis-chloroethylnitrosourea) wafers. Thirteen of the newly diagnosed patients received local BCNU and irradiation only (first-line BCNU), while 17 patients additionally received concomitant and adjuvant temozolomide (TMZ) radiochemotherapy (first-line BCNU + TMZ). Of the 17 patients treated for recurrent glioblastoma (second-line BCNU), 16 had received radiotherapy with concomitant and adjuvant TMZ as an initial treatment. Median overall survival (OS) did not significantly differ between 19 patients with MGMT promoter methylated tumors when compared to 28 patients with unmethylated tumors (18.9 vs 15.0 months; p = 0.1054). In the first-line BCNU + TMZ group, MGMT promoter methylation was associated with longer OS (21.0 vs 11.1 months, p = 0.0127), while no significant survival differences were detected in the other two subgroups. Progression-free survival did not significantly differ between patients with and without MGMT promoter methylated tumors in the entire patient cohort or any of the three subgroups. The first-line BCNU + TMZ group showed no significant difference in OS when compared to the first-line BCNU group (18.9 vs 14.7 months), but tended to have more therapy-related adverse effects (53% vs 24%, p = 0.105). In summary, MGMT promoter methylation showed a non-significant trend toward longer survival in our patient cohort. The combination of TMZ radiochemotherapy with local delivery of BCNU did not provide a significant survival benefit compared to local BCNU alone, but was associated with a higher rate of adverse effects. Owing to the small number of patients investigated, however, these findings would need to be corroborated in larger patient cohorts.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Carmustine/therapeutic use , Glioblastoma/drug therapy , Glioblastoma/genetics , O(6)-Methylguanine-DNA Methyltransferase/genetics , Adult , Aged , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carmustine/administration & dosage , Carmustine/adverse effects , Chemoradiotherapy/methods , Combined Modality Therapy , DNA Methylation , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Disease-Free Survival , Female , Humans , Karnofsky Performance Status , Male , Middle Aged , Prognosis , Promoter Regions, Genetic/genetics , Retrospective Studies , Survival Analysis , Temozolomide
15.
Oncogene ; 32(39): 4712-20, 2013 Sep 26.
Article in English | MEDLINE | ID: mdl-23108408

ABSTRACT

Meningiomas are frequent, mostly benign intracranial or spinal tumors. A small subset of meningiomas is characterized by histological features of atypia or anaplasia that are associated with more aggressive biological behavior resulting in increased morbidity and mortality. Infiltration into the adjacent brain tissue is a major factor linked to higher recurrence rates. The molecular mechanisms of progression, including brain invasion are still poorly understood. We have studied the role of micro-RNA 145 (miR-145) in meningiomas and detected significantly reduced miR-145 expression in atypical and anaplastic tumors as compared with benign meningiomas. Overexpression of miR-145 in IOMM-Lee meningioma cells resulted in reduced proliferation, increased sensitivity to apoptosis, reduced anchorage-independent growth and reduction of orthotopic tumor growth in nude mice as compared with control cells. Moreover, meningioma cells with high miR-145 levels had impaired migratory and invasive potential in vitro and in vivo. PCR-array studies of miR145-overexpressing cells suggested that collagen type V alpha (COL5A1) expression is downregulated by miR-145 overexpression. Accordingly, COL5A1 expression was significantly upregulated in atypical and anaplastic meningiomas. Collectively, our data indicate an important anti-migratory and anti-proliferative function of miR-145 in meningiomas.


Subject(s)
Meningeal Neoplasms/metabolism , Meningioma/metabolism , MicroRNAs/physiology , Neoplasm Invasiveness/genetics , RNA, Neoplasm/physiology , Animals , Cell Adhesion , Cell Differentiation , Cell Division , Cell Movement , Collagen Type V/biosynthesis , Collagen Type V/genetics , Down-Regulation , Humans , Meningeal Neoplasms/genetics , Meningeal Neoplasms/pathology , Meningioma/genetics , Meningioma/pathology , Mice , Mice, Nude , MicroRNAs/biosynthesis , MicroRNAs/genetics , Neoplasm Grading , Neoplasm Invasiveness/physiopathology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Transplantation , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Tumor Stem Cell Assay
16.
Acta Neurochir (Wien) ; 155(3): 429-35, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23254891

ABSTRACT

BACKGROUND: To compare survival and hematological toxicity rates between two postoperative therapy regimens in patients with primary glioblastoma (GBM), namely temozolomide (TMZ) concomitant to radiation, followed by adjuvant TMZ, versus adjuvant TMZ after radiation only. PATIENTS AND METHODS: A total of 191 patients with primary GBM were postoperatively treated with either radiation and concomitant TMZ, followed by adjuvant TMZ (Stupp protocol) (n = 154), or radiation followed by adjuvant TMZ (n = 37). The incidence of hematological adverse effects (AE) was recorded for all patients. From both treatment groups, 26 patients were matched according to age, Karnofsky performance scale (KPS) score, and O6-methylguanine-DNA-methyltransferase (MGMT) promoter methylation. RESULTS: Hematological AEs were mild in both unmatched groups, but were significantly more frequent in the concomitant plus adjuvant TMZ group (p < 0.001). Matched-pair analysis confirmed significantly more frequent hematological AEs in the concomitant and adjuvant group compared to the sequential (adjuvant) TMZ group (p = 0,012). Patients treated with concomitant plus adjuvant TMZ showed significantly longer progression-free survival (PFS) (10.6 versus 6.6 months; p = 0.014), but no prolonged overall survival (OS) (16.9 vs. 15.6 months; p = 0.717) compared to patients who received the sequential treatment regimen. CONCLUSION: In this retrospective study, the OS in patients with primary GBM treated with sequential TMZ following radiation appeared to be similar to that in patients treated with concomitant plus adjuvant TMZ. Given the significantly higher risk of hematological AE for concomitant treatment, the role of concomitant plus adjuvant TMZ use compared to sequential administration of TMZ, especially for patients with MGMT-unmethylated tumors, should be further evaluated.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/toxicity , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Chemoradiotherapy, Adjuvant , Chemoradiotherapy , Dacarbazine/analogs & derivatives , Glioblastoma/mortality , Glioblastoma/therapy , Adult , Aged , Aged, 80 and over , Combined Modality Therapy , Dacarbazine/administration & dosage , Dacarbazine/toxicity , Female , Humans , Male , Matched-Pair Analysis , Middle Aged , Prognosis , Retrospective Studies , Survival Rate , Temozolomide
17.
Oncogene ; 31(27): 3235-43, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22056879

ABSTRACT

The concept of cancer stem-like cells (CSCs) has gained considerable attention in various solid tumors including glioblastoma, the most common primary brain tumor. This sub-population of tumor cells has been intensively investigated and their role in therapy resistance as well as tumor recurrence has been demonstrated. In that respect, development of therapeutic strategies that target CSCs (and possibly also the tumor bulk) appears a promising approach in patients suffering from primary brain tumors. In the present study, we utilized RNA interference (RNAi) to screen the complete human kinome and phosphatome (682 and 180 targets, respectively) in order to identify genes and pathways relevant for the survival of brain CSCs and thereby potential therapeutical targets for glioblastoma. We report of 46 putative candidates including known survival-related kinases and phosphatases. Interestingly, a number of genes identified are involved in metabolism, especially glycolysis, such as PDK1 and PKM2 and, most prominently PFKFB4. In vitro studies confirmed an essential role of PFKFB4 in the maintenance of brain CSCs. Furthermore, high PFKFB4 expression was associated with shorter survival of primary glioblastoma patients. Our findings support the importance of the glycolytic pathway in the maintenance of malignant glioma cells and brain CSCs and imply tumor metabolism as a promising therapeutic target in glioblastoma.


Subject(s)
Glioma/genetics , Glioma/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phosphofructokinase-2/deficiency , Phosphofructokinase-2/genetics , RNA Interference , Adenosine Triphosphate/biosynthesis , Brain Neoplasms/diagnosis , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Death/genetics , Cell Survival/genetics , Gene Expression Regulation, Neoplastic/genetics , Glioma/diagnosis , Glioma/metabolism , Glycolysis/genetics , Humans , Isoenzymes/deficiency , Isoenzymes/genetics , Lactic Acid/biosynthesis , Lentivirus/genetics , Prognosis , RNA, Small Interfering/genetics
18.
Neuropathol Appl Neurobiol ; 38(2): 201-12, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21722156

ABSTRACT

AIMS: The molecular mechanisms underlying the infiltrative growth of glioblastomas, the most common primary tumours of the central nervous system in adults, are still poorly understood. We aimed to identify and functionally validate novel glioma invasion-associated candidate genes. METHODS: Microarray-based expression analysis was applied to identify differentially expressed genes in microdissected infiltrating glioma cells in vivo. Promising candidate genes were selected by the invasion-associated gene ontology terms cell adhesion, endocytosis, extracellular matrix and cell migration and validated in vitro by invasion assays and in situ by immunohistochemistry. RESULTS: We identified 180 up-regulated and 61 down-regulated genes (fold change: ≥ 2; P < 0.01) in the infiltration zone relative to more central cell-rich tumour areas of malignant astrocytic gliomas (n = 11). Twenty-seven of these genes matched to invasion-related gene ontology terms. From these, we confirmed the genes encoding cadherin-11 (CDH11), proprotein convertase subtilisin/kexin type 6 (PCSK6) and SH3-domain GRB2-like 3 (SH3GL3) as novel glioma invasion-associated candidate genes, with knockdown of PCSK6 and SH3GL3 inhibiting glioma cell invasion, while inhibition of CDH11 promoted glioma cell invasion in vitro. Immunohistochemistry on glioblastoma tissue sections revealed expression of CDH11 and PCSK6 protein in glioma cells of more central, cell-rich tumour areas, with only weak or absent CDH11 immunoreactivity but consistent PCSK6 staining in infiltrating glioma cells. CONCLUSION: Using molecular profiling of microdissected primary tumour tissue specimens followed by functional in vitro analysis, we identified and validated CDH11, PCSK6 and SH3GL3 as novel glioma invasion-associated candidate genes that likely contribute to the invasive phenotype of malignant gliomas.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Brain Neoplasms/genetics , Cadherins/genetics , Glioma/genetics , Neoplasm Invasiveness/genetics , Proprotein Convertases/genetics , Serine Endopeptidases/genetics , Adolescent , Adult , Brain Neoplasms/metabolism , Cell Movement/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Humans , Male , Middle Aged , Young Adult
19.
Oncogene ; 31(29): 3409-18, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22158042

ABSTRACT

Deletions of chromosomal arms 1p and 19q are frequent in oligodendroglial tumours and linked to radio- and chemotherapy response as well as longer survival. The molecular mechanisms underlying this clinically important association are as yet unknown. Here, we studied the peroxiredoxin 1 (PRDX1) gene at 1p34.1 for promoter methylation and expression in primary gliomas and investigated its role in radio- and chemosensitivity of glioma cells in vitro. In total, we screened primary glioma tissues from 93 patients for methylation of the 5'-CpG island of PRDX1 by sodium bisulfite sequencing. PRDX1 mRNA and protein expression levels were determined in subsets of the tumours by quantitative PCR and western blot analysis, respectively. PRDX1 hypermethylation and reduced expression were frequently detected in oligodendroglial tumours and secondary glioblastomas, but not in primary glioblastomas. In oligodendroglial tumours, both PRDX1 hypermethylation and reduced mRNA expression were significantly associated with 1p/19q-deletion. Stable knockdown of PRDX1 by lentiviral transduction of short-hairpin (sh)RNA constructs significantly increased apoptosis and reduced cell viability of Hs683 glioma cells exposed to ionizing irradiation or temozolomide in vitro. Taken together, our findings indicate that epigenetic silencing of PRDX1 is frequent in 1p/19q-deleted oligodendroglial tumours and likely contributes to radio- and chemosensitivity of these tumours.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 1/genetics , Glioma/pathology , Oligodendroglia/metabolism , Peroxiredoxins/genetics , Promoter Regions, Genetic/genetics , Radiation Tolerance/genetics , Adult , Aged , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/radiation effects , Cell Line, Tumor , CpG Islands/genetics , DNA Methylation/drug effects , DNA Methylation/radiation effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Down-Regulation/genetics , Female , Gene Knockdown Techniques , Gene Silencing , Glioma/drug therapy , Glioma/genetics , Glioma/radiotherapy , Humans , Isocitrate Dehydrogenase/genetics , Male , Middle Aged , Mutation , Oligodendroglia/drug effects , Oligodendroglia/pathology , Oligodendroglia/radiation effects , Peroxiredoxins/deficiency , Promoter Regions, Genetic/drug effects , Promoter Regions, Genetic/radiation effects , Temozolomide , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...