Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 72
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Transl Med ; 21(1): 764, 2023 10 28.
Article in English | MEDLINE | ID: mdl-37898750

ABSTRACT

BACKGROUND: Animal models are widely used to study pathological processes and drug (side) effects in a controlled environment. There is a wide variety of methods available for establishing animal models depending on the research question. Commonly used methods in tumor research include xenografting cells (established/commercially available or primary patient-derived) or whole tumor pieces either orthotopically or heterotopically and the more recent genetically engineered models-each type with their own advantages and disadvantages. The current systematic review aimed to investigate the meningioma model types used, perform a meta-analysis on tumor take rate (TTR), and perform critical appraisal of the included studies. The study also aimed to assess reproducibility, reliability, means of validation and verification of models, alongside pros and cons and uses of the model types. METHODS: We searched Medline, Embase, and Web of Science for all in vivo meningioma models. The primary outcome was tumor take rate. Meta-analysis was performed on tumor take rate followed by subgroup analyses on the number of cells and duration of incubation. The validity of the tumor models was assessed qualitatively. We performed critical appraisal of the methodological quality and quality of reporting for all included studies. RESULTS: We included 114 unique records (78 using established cell line models (ECLM), 21 using primary patient-derived tumor models (PTM), 10 using genetically engineered models (GEM), and 11 using uncategorized models). TTRs for ECLM were 94% (95% CI 92-96) for orthotopic and 95% (93-96) for heterotopic. PTM showed lower TTRs [orthotopic 53% (33-72) and heterotopic 82% (73-89)] and finally GEM revealed a TTR of 34% (26-43). CONCLUSION: This systematic review shows high consistent TTRs in established cell line models and varying TTRs in primary patient-derived models and genetically engineered models. However, we identified several issues regarding the quality of reporting and the methodological approach that reduce the validity, transparency, and reproducibility of studies and suggest a high risk of publication bias. Finally, each tumor model type has specific roles in research based on their advantages (and disadvantages). SYSTEMATIC REVIEW REGISTRATION: PROSPERO-ID CRD42022308833.


Subject(s)
Meningeal Neoplasms , Meningioma , Animals , Humans , Reproducibility of Results , Disease Models, Animal
2.
Acta Neuropathol ; 145(5): 667-680, 2023 05.
Article in English | MEDLINE | ID: mdl-36933012

ABSTRACT

Glioneuronal tumors are a heterogenous group of CNS neoplasms that can be challenging to accurately diagnose. Molecular methods are highly useful in classifying these tumors-distinguishing precise classes from their histological mimics and identifying previously unrecognized types of tumors. Using an unsupervised visualization approach of DNA methylation data, we identified a novel group of tumors (n = 20) that formed a cluster separate from all established CNS tumor types. Molecular analyses revealed ATRX alterations (in 16/16 cases by DNA sequencing and/or immunohistochemistry) as well as potentially targetable gene fusions involving receptor tyrosine-kinases (RTK; mostly NTRK1-3) in all of these tumors (16/16; 100%). In addition, copy number profiling showed homozygous deletions of CDKN2A/B in 55% of cases. Histological and immunohistochemical investigations revealed glioneuronal tumors with isomorphic, round and often condensed nuclei, perinuclear clearing, high mitotic activity and microvascular proliferation. Tumors were mainly located supratentorially (84%) and occurred in patients with a median age of 19 years. Survival data were limited (n = 18) but point towards a more aggressive biology as compared to other glioneuronal tumors (median progression-free survival 12.5 months). Given their molecular characteristics in addition to anaplastic features, we suggest the term glioneuronal tumor with ATRX alteration, kinase fusion and anaplastic features (GTAKA) to describe these tumors. In summary, our findings highlight a novel type of glioneuronal tumor driven by different RTK fusions accompanied by recurrent alterations in ATRX and homozygous deletions of CDKN2A/B. Targeted approaches such as NTRK inhibition might represent a therapeutic option for patients suffering from these tumors.


Subject(s)
Brain Neoplasms , Central Nervous System Neoplasms , Neoplasms, Neuroepithelial , Humans , Young Adult , Biomarkers, Tumor/genetics , Brain/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Gene Fusion , Neoplasms, Neuroepithelial/genetics , Neoplasms, Neuroepithelial/pathology , Receptor Protein-Tyrosine Kinases/genetics , X-linked Nuclear Protein/genetics
3.
Neuropathol Appl Neurobiol ; 48(1): e12767, 2022 02.
Article in English | MEDLINE | ID: mdl-34533233

ABSTRACT

The advent of checkpoint immunotherapy, particularly with programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) inhibitors, has provided ground-breaking results in several advanced cancers. Substantial efforts are being made to extend these promising therapies to other refractory cancers such as gliomas, especially glioblastoma, which represents the most frequent and malignant glioma and carries an exceptionally grim prognosis. Thus, there is a need for new therapeutic strategies with related biomarkers. Gliomas have a profoundly immunosuppressive tumour micro-environment and evade immunological destruction by several mechanisms, one being the expression of inhibitory immune checkpoint molecules such as PD-L1. PD-L1 is recognised as an important therapeutic target and its expression has been shown to hold prognostic value in different cancers. Several clinical trials have been launched and some already completed, but PD-1/PD-L1 inhibitors have yet to show convincing clinical efficacy in gliomas. Part of the explanation may reside in the vast molecular heterogeneity of gliomas and a complex interplay within the tumour micro-environment. In parallel, critical knowledge about PD-L1 expression is beginning to accumulate including knowledge on expression levels, testing methodology, co-expression with other checkpoint molecules and prognostic and predictive value. This article reviews these aspects and points out areas where biomarker research is needed to develop more successful checkpoint-related therapeutic strategies in gliomas.


Subject(s)
B7-H1 Antigen , Glioma , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Glioma/therapy , Humans , Prognosis , Programmed Cell Death 1 Receptor/metabolism , Tumor Microenvironment
4.
Neuropathol Appl Neurobiol ; 48(3): e12773, 2022 04.
Article in English | MEDLINE | ID: mdl-34799864

ABSTRACT

TERT promoter mutations have been associated with increased risk of recurrence in meningioma cohorts, thus a potential biomarker for aggressive phenotypes. A main purpose of refining tumour classification is better predictions on the patient level. We compiled data from previous published cohorts to investigate patient-level predictions of recurrence based on TERTp-mut status. Implementation of TERTp-mut into the WHO grading led to better patient prognostication by improved prediction of recurrence. Our results support implementation of TERTp-mut into diagnostics and classification of meningiomas.


Subject(s)
Meningeal Neoplasms , Meningioma , Telomerase , Humans , Meningeal Neoplasms/genetics , Meningeal Neoplasms/pathology , Meningioma/genetics , Meningioma/pathology , Mutation , Promoter Regions, Genetic/genetics , Telomerase/genetics , World Health Organization
5.
Neurosurg Rev ; 45(5): 3067-3081, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35984552

ABSTRACT

Treatment-refractory meningiomas have a dismal prognosis and limited treatment options. Meningiomas express high-densities of somatostatin receptors (SSTR), thus potentially susceptible to antitumorigenic effects of somatostatin analogues (SSA). Evidence for SSA in meningiomas is scarce, and it is unclear if published literature would either (1) support wider use of SSA, if (2) more evidence is desirable, or if (3) available evidence is sufficient to discard SSA. We addressed the need for more evidence with a systematic review and meta-analysis. We performed an individual patient data (IPD) meta-analysis. Main outcomes were toxicity, best radiological response, progression-free survival, and overall survival. We applied multivariable logistic regression models to estimate the effect of SSA on the probability of obtaining radiological disease control. The predictive performance was evaluated using area under the curve and Brier scores. We included 16 studies and compiled IPD from 8/9 of all previous cohorts. Quality of evidence was overall ranked "very low." Stable disease was reported in 58% of patients as best radiological response. Per 100 mg increase in total SSA dosage, the odds ratios for obtaining radiological disease control was 1.42 (1.11 to 1.81, P = 0.005) and 1.44 (1.00 to 2.08, P = 0.05) for patients treated with SSA as monodrug therapy vs SSA in combination with everolimus, respectively. Low quality of evidence impeded exact quantification of treatment efficacy, and the association between response and treatment may represent reverse causality. Yet, the SSA treatment was well tolerated, and beneficial effect cannot be disqualified. A prospective trial without bias from inconsistent study designs is warranted to assess SSA therapy for well-defined meningioma subgroups.


Subject(s)
Meningeal Neoplasms , Meningioma , Everolimus/therapeutic use , Humans , Meningeal Neoplasms/drug therapy , Meningioma/drug therapy , Prospective Studies , Receptors, Somatostatin/therapeutic use , Somatostatin/therapeutic use
6.
Acta Neurochir (Wien) ; 164(1): 229-238, 2022 01.
Article in English | MEDLINE | ID: mdl-34714434

ABSTRACT

INTRODUCTION: The extent of meningioma resection is the most fundamental risk factor for recurrence, and exact knowledge of extent of resection is necessary for prognostication and for planning of adjuvant treatment. Currently used classifications are the EANO-grading and the Simpson grading. The former comprises radiological imaging with contrast-enhanced MRI and differentiation between "gross total removal" and "subtotal removal," while the latter comprises a five-tiered differentiation of the surgeon's impression of the extent of resection. The extent of resection of tumors is usually defined via analyses of resection margins but has until now not been implemented for meningiomas. PET/MRI imaging with 68Ga-DOTATOC allows more sensitive and specific imaging than MRI following surgery of meningiomas. OBJECTIVE: To develop an objective grading system based on microscopic analyses of resection margins and sensitive radiological analyses to improve management of follow-up, adjuvant therapy, and prognostication of meningiomas. Based on the rationale of resection-margin analyses as gold standard and superior imaging performance of 68Ga DOTATOC PET, we propose "Copenhagen Grading" for meningiomas. RESULTS: Copenhagen Grading was described for six pilot patients with examples of positive and negative findings on histopathology and DOTATOC PET scanning. The grading could be traceably implemented and parameters of grading appeared complementary. Copenhagen Grading is prospectively implemented as a clinical standard at Rigshospitalet, Copenhagen. CONCLUSION: Copenhagen Grading provided a comprehensive, logical, and reproducible definition of the extent of resection. It offers promise to be the most sensitive and specific imaging modality available for meningiomas. Clinical and cost-efficacy remain to be established during prospective implementation.


Subject(s)
Meningeal Neoplasms , Meningioma , Humans , Meningeal Neoplasms/diagnostic imaging , Meningeal Neoplasms/surgery , Meningioma/diagnostic imaging , Meningioma/surgery , Neoplasm Grading , Neoplasm Recurrence, Local , Positron-Emission Tomography , Prospective Studies , Retrospective Studies
7.
Acta Neuropathol ; 142(1): 179-189, 2021 07.
Article in English | MEDLINE | ID: mdl-33876327

ABSTRACT

Glioblastoma IDH-wildtype presents with a wide histological spectrum. Some features are so distinctive that they are considered as separate histological variants or patterns for the purpose of classification. However, these usually lack defined (epi-)genetic alterations or profiles correlating with this histology. Here, we describe a molecular subtype with overlap to the unique histological pattern of glioblastoma with primitive neuronal component. Our cohort consists of 63 IDH-wildtype glioblastomas that harbor a characteristic DNA methylation profile. Median age at diagnosis was 59.5 years. Copy-number variations and genetic sequencing revealed frequent alterations in TP53, RB1 and PTEN, with fewer gains of chromosome 7 and homozygous CDKN2A/B deletions than usually described for IDH-wildtype glioblastoma. Gains of chromosome 1 were detected in more than half of the cases. A poorly differentiated phenotype with frequent absence of GFAP expression, high proliferation index and strong staining for p53 and TTF1 often caused misleading histological classification as carcinoma metastasis or primitive neuroectodermal tumor. Clinically, many patients presented with leptomeningeal dissemination and spinal metastasis. Outcome was poor with a median overall survival of only 12 months. Overall, we describe a new molecular subtype of IDH-wildtype glioblastoma with a distinct histological appearance and genetic signature.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA Methylation , Glioblastoma/genetics , Glioblastoma/pathology , Neuroectodermal Tumors, Primitive/genetics , Neuroectodermal Tumors, Primitive/pathology , PTEN Phosphohydrolase/genetics , Retinoblastoma Binding Proteins/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics , Chromosomes, Human, Pair 1/genetics , Chromosomes, Human, Pair 7/genetics , Cohort Studies , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Copy Number Variations , Female , Gene Deletion , Glial Fibrillary Acidic Protein/biosynthesis , Glial Fibrillary Acidic Protein/genetics , Humans , Male , Middle Aged
8.
BMC Cancer ; 20(1): 710, 2020 Jul 29.
Article in English | MEDLINE | ID: mdl-32727404

ABSTRACT

BACKGROUND: The prognosis of glioblastoma remains poor, related to its diffuse spread within the brain. There is an ongoing search for molecular regulators of this particularly invasive behavior. One approach is to look for actin regulating proteins that might be targeted by future anti-cancer therapy. The formin family of proteins orchestrates rearrangement of the actin cytoskeleton in multiple cellular processes. Recently, the formin proteins mDia1 and mDia2 were shown to be expressed in glioblastoma in vitro, and their function could be modified by small molecule agonists. This finding implies that the formins could be future therapeutic targets in glioblastoma. METHODS: In cell studies, we investigated the changes in expression of the 15 human formins in primary glioblastoma cells and commercially available glioblastoma cell lines during differentiation from spheroids to migrating cells using transcriptomic analysis and qRT-PCR. siRNA mediated knockdown of selected formins was performed to investigate whether their expression affects glioblastoma migration. Using immunohistochemistry, we studied the expression of two formins, FHOD1 and INF2, in tissue samples from 93 IDH-wildtype glioblastomas. Associated clinicopathological parameters and follow-up data were utilized to test whether formin expression correlates with survival or has prognostic value. RESULTS: We found that multiple formins were upregulated during migration. Knockdown of individual formins mDia1, mDia2, FHOD1 and INF2 significantly reduced migration in most studied cell lines. Among the studied formins, knockdown of INF2 generated the greatest reduction in motility in vitro. Using immunohistochemistry, we demonstrated expression of formin proteins FHOD1 and INF2 in glioblastoma tissues. Importantly, we found that moderate/high expression of INF2 was associated with significantly impaired prognosis. CONCLUSIONS: Formins FHOD1 and INF2 participate in glioblastoma cell migration. Moderate/high expression of INF2 in glioblastoma tissue is associated with worse outcome. Taken together, our in vitro and tissue studies suggest a pivotal role for INF2 in glioblastoma. When specific inhibiting compounds become available, INF2 could be a target in the search for novel glioblastoma therapies.


Subject(s)
Brain Neoplasms/metabolism , Cell Movement , Fetal Proteins/metabolism , Formins/metabolism , Glioblastoma/metabolism , Actins/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Fetal Proteins/genetics , Formins/genetics , Gene Knockdown Techniques , Glioblastoma/pathology , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Up-Regulation
9.
J Neurooncol ; 147(1): 1-14, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31960234

ABSTRACT

PURPOSE: Isocitrate dehydrogenase 1 (IDH1) mutations are associated with improved survival in gliomas. Depending on the IDH1 status, TERT promoter mutations affect prognosis. IDH1 mutations are associated with alpha-thalassemia/mental retardation syndrome X-linked (ATRX) mutations and alternative lengthening of telomeres (ALT), suggesting an interaction between IDH1 and telomeres. However, little is known how IDH1 mutations affect telomere maintenance. METHODS: We analyzed cell-specific telomere length (CS-TL) on a single cell level in 46 astrocytoma samples (WHO II-IV) by modified immune-quantitative fluorescence in situ hybridization, using endothelial cells as internal reference. In the same samples, we determined IDH1/TERT promoter mutation status and ATRX expression. The interaction of IDH1R132H mutation and CS-TL was studied in vitro using an IDH1R132H doxycycline-inducible glioma cell line system. RESULTS: Virtually all ALTpositive astrocytomas had normal TERT promoter and lacked ATRX expression. Further, all ALTpositive samples had IDH1R132H mutations, resulting in a significantly longer CS-TL of IDH1R132H gliomas, when compared to their wildtype counterparts. Conversely, TERT promotor mutations were associated with IDHwildtype, ATRX expression, lack of ALT and short CS-TL. ALT, TERT promoter mutations, and CS-TL remained without prognostic significance, when correcting for IDH1 status. In vitro, overexpression of IDHR132H in the glioma cell line LN319 resulted in downregulation of ATRX and rapid TERT-independent telomere lengthening consistent with ALT. CONCLUSION: ALT is the major telomere maintenance mechanism in IDHR132H mutated astrocytomas, while TERT promoter mutations were associated with IDHwildtype glioma. IDH1R132H downregulates ATRX expression in vitro resulting in ALT, which may contribute to the strong association of IDH1R132H mutations, ATRX loss, and ALT.


Subject(s)
Astrocytoma/genetics , Brain Neoplasms/genetics , Isocitrate Dehydrogenase/genetics , Telomerase/genetics , Telomere Homeostasis/genetics , X-linked Nuclear Protein/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Isocitrate Dehydrogenase/metabolism , Male , Middle Aged , Mutation , Single-Cell Analysis , Tumor Cells, Cultured , Young Adult
10.
Exp Cell Res ; 379(1): 73-82, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30922921

ABSTRACT

Glioblastoma multiforme (GBM) is the most common and malignant type of primary brain tumor and is characterized by its sudden onset and invasive growth into the brain parenchyma. The invasive tumor cells evade conventional treatments and are thought to be responsible for the ubiquitous tumor regrowth. Understanding the behavior of these invasive tumor cells and their response to therapeutic agents could help improve patient outcome. In this study, we present a GBM tumorsphere migration model with high biological complexity to study migrating GBM cells in a quantitative and qualitative manner. We demonstrated that the in vitro migration model could be used to investigate both inhibition and stimulation of cell migration with oxaliplatin and GBM-derived extracellular vesicles, respectively. The intercellular heterogeneity within the GBM tumorspheres was examined by immunofluorescent staining of nestin/vimentin and GFAP, which showed nestin and vimentin being highly expressed in the periphery of tumorspheres and GFAP mostly in cells in the tumorsphere core. We further showed that this phenotypic gradient was present in vivo after implanting dissociated GBM tumorspheres, with the cells migrating away from the tumor being nestin-positive and GFAP-negative. These results indicate that GBM tumorsphere migration models, such as the one presented here, could provide a more detailed insight into GBM cell biology and prove highly relevant as a pre-clinical platform for drug screening and assessing drug response in the treatment of GBM.


Subject(s)
Brain Neoplasms/pathology , Cell Movement/physiology , Glioblastoma/pathology , Animals , Brain Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/physiology , Evaluation Studies as Topic , Gene Expression Regulation, Neoplastic/physiology , Glioblastoma/metabolism , Humans , Mice , Mice, Nude , Nestin/metabolism , Vimentin/metabolism
11.
Surgeon ; 18(6): 344-348, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32044289

ABSTRACT

INTRODUCTION: Glioblastoma has a high mortality rate. Current treatment includes largest possible surgical resection of the tumour using neuronavigation and fluorescence to better identify tumour tissue. In recent years, sodium fluorescein has been reintroduced in neurosurgery as a fluorescence to increase the resection rate. In this study we aimed to measure the surgeons experience of using sodium fluorescein to locate and remove tumour tissue. Furthermore we describe a case of sodium fluorescein tissue distribution. MATERIAL AND METHODS: 13 patients with glioblastoma and seven patients with cerebral metastases undergoing surgical resection were included. Surgery was performed using microscope alternating between white light and the YELLOW 560 filter, which visualized sodium fluorescein. Surgeons graded its usability in terms of location and removal on a scale from one to four. The resection rate was determined by neuroradiologists. Tissue samples obtained during surgery were analysed in relation to fluorescence and dysmorphic cells. RESULTS: Surgeons reported high usability in terms of location and removal of tumours using sodium fluorescein with medians of four in all groups, except for sub-total resections which had a median of three. Surgical complications were minimal and both resection rate and survival rate was within international standards. Histological analysis showed a visual correlation between tumorous tissue and intensity of fluorescence. CONCLUSION: Sodium fluorescence is an effective and useful tool for surgeons during fluorescence-guided surgery for the resection of glioblastoma and cerebral metastases.


Subject(s)
Brain Neoplasms/surgery , Fluorescein , Fluorescent Dyes , Glioblastoma/surgery , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Cohort Studies , Female , Glioblastoma/mortality , Glioblastoma/pathology , Humans , Male , Middle Aged , Practice Patterns, Physicians'
12.
Acta Neuropathol ; 138(3): 497-504, 2019 09.
Article in English | MEDLINE | ID: mdl-31250151

ABSTRACT

Rosette-forming glioneuronal tumor (RGNT) is a rare brain neoplasm that primarily affects young adults. Although alterations affecting the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) signaling pathway have been associated with this low-grade entity, comprehensive molecular investigations of RGNT in larger series have not been performed to date, and an integrated view of their genetic and epigenetic profiles is still lacking. Here we describe a genome-wide DNA methylation and targeted sequencing-based characterization of a molecularly distinct class of tumors (n = 30), initially identified through genome-wide DNA methylation screening among a cohort of > 30,000 tumors, of which most were diagnosed histologically as RGNT. FGFR1 hotspot mutations were observed in all tumors analyzed, with co-occurrence of PIK3CA mutations in about two-thirds of the cases (63%). Additional loss-of-function mutations in the tumor suppressor gene NF1 were detected in a subset of cases (33%). Notably, in contrast to most other low-grade gliomas, these tumors often displayed co-occurrence of two or even all three of these mutations. Our data highlight that molecularly defined RGNTs are characterized by highly recurrent combined genetic alterations affecting both MAPK and PI3K signaling pathways. Thus, these two pathways appear to synergistically interact in the formation of RGNT, and offer potential therapeutic targets for this disease.


Subject(s)
Brain Neoplasms/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Glioma/genetics , Neurofibromin 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Adolescent , Adult , Aged , Child , DNA Methylation , Female , Humans , Male , Middle Aged , Mutation , Neurons/pathology , Retrospective Studies , Young Adult
13.
Acta Neurochir (Wien) ; 161(3): 555-565, 2019 03.
Article in English | MEDLINE | ID: mdl-30756241

ABSTRACT

BACKGROUND: This study aimed to investigate the incidence of residual tumour after resection of brain metastases using early postoperative magnetic resonance imaging (MRI) and the influence of residual tumour on overall patient survival. METHODS: Data from 72 consecutive adult patients undergoing surgery for cerebral metastases over an 18-month study period were retrospectively collected. Early postoperative MRI was used to determine the presence of postoperative residual tumour. Patients were divided into three groups according to the presence of tumour remnant on early postoperative MRI: "no residual tumour", "non-measurable residual tumour" and "measurable residual tumour". Survival analysis (mean estimate survival time) was performed using the Kaplan-Meier and log-rank (mantel cox) tests and compared between groups. Surgical reports were evaluated with regard to the surgeon statement about intraoperative extent of resection (EOR) and compared with the presence of tumour remnant found on the early postoperative MRI. RESULTS: Sixty-eight procedures were followed by early postoperative MRI. MRI verified the presence of "measurable residual tumour" following 15 procedures (22%). MRI confirmed complete resection in 57%. Gross total resection was described by the operating surgeon in 85% of the procedures. There was a significant difference in survival time after surgery between the group having no residual tumour on MRI and the group with measurable residual tumour (p = 0.025). This difference could not be explained by the differences in postoperative radiation therapy. The longest survival was found in patients with non-measurable and no residual tumour on early postoperative MRI, who also received postoperative radiotherapy. CONCLUSION: Residual tumour was seen on MRI after 22% of the procedures. The intraoperative assessment of EOR performed by the surgeon diverged from the early postoperative MRI in 40% of procedures. Correct assessment of residual tumour thus requires early postoperative MRI. Measurable residual tumour on early postoperative MRI was associated with shorter overall survival independent on postoperative radiotherapy.


Subject(s)
Brain Neoplasms/surgery , Magnetic Resonance Imaging/methods , Postoperative Complications/epidemiology , Adult , Aged , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/secondary , Female , Humans , Male , Middle Aged , Neoplasm, Residual , Neurosurgical Procedures/adverse effects , Neurosurgical Procedures/methods , Postoperative Complications/diagnostic imaging , Survival Analysis
14.
J Neurooncol ; 138(1): 49-53, 2018 May.
Article in English | MEDLINE | ID: mdl-29404978

ABSTRACT

Epileptic seizures are an important cause of morbidity in glioma patients. Substantial lines of evidence support the concept of the excitatory neurotransmitter glutamate being a crucial mediator of glioma-associated seizures. In gliomas, non-vesicular secretion of glutamate via the cystine-glutamate exchanger (SLC7A11, xCT) constitutes the main mechanism contributing to high extracellular glutamate concentrations. However, a convincing "proof-of-relevance" of this mechanism in patient material is lacking. A cohort of 229 consecutive patients with newly diagnosed glioma was analyzed with respect to presence, time course, and severity of epileptic seizures. 14 patients were excluded due to previous epileptic seizures, insufficient clinical data or insufficient tumor material. The maximal immunohistochemical expression of xCT was determined in 1-3 independent samples from central tumor areas of each tumor using tissue microarrays. In addition to histological grading of the tumors, isocitrate dehydrogenase 1 (IDH1) R132H mutational status was determined by immunohistochemistry. 215 consecutive glioma patients were included in the study (7.4% grade II, 7.0% grade III, 85.6% grade IV). High xCT expression was significantly associated with seizures at onset (p = 0.05) but not with development of seizures or with refractory seizures. Low-grade gliomas (WHO II/III) had lower xCT expression than glioblastoma (p = 0.001), and tumors without IDH1 R132H mutation tended to have higher xCT levels (p = 0.07). In a multivariate analysis, high xCT expression and WHO tumor grade but not IDH1 R132H mutation, were significantly associated with epileptic seizures at diagnosis (odds ratio 2.2, p = 0.02). Further, xCT expression did not correlate with survival (p = 0.27, log-rank test). Thus, high xCT expression is an independent marker for glioma-associated seizures at diagnosis especially in high-grade glioma, but is not associated with worse survival in our cohort.


Subject(s)
Amino Acid Transport System y+/metabolism , Brain Neoplasms/complications , Glioma/complications , Seizures/etiology , Seizures/metabolism , Aged , Amino Acid Transport System y+/genetics , Brain Neoplasms/genetics , Cohort Studies , Female , Gene Expression Regulation, Neoplastic/physiology , Glioma/genetics , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Male , Middle Aged , Multivariate Analysis , Mutation/genetics , Survival Analysis , Tissue Array Analysis
15.
Acta Oncol ; 56(12): 1776-1785, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28675067

ABSTRACT

INTRODUCTION: A pilot study of temozolomide (TMZ) given before radiotherapy (RT) for anaplastic astrocytoma (AA) and glioblastoma (GBM) resulted in prolonged survival compared to historical controls receiving RT alone. We therefore investigated neoadjuvant TMZ (NeoTMZ) in a randomized trial. During enrollment, concomitant and adjuvant radio-chemotherapy with TMZ became standard treatment. The trial was amended to include concurrent TMZ. PATIENTS AND METHODS: Patients, after surgery for GBM or AA, age ≤60 years and performance status (PS) 0-2, were randomized to either 2-3 cycles of TMZ, 200 mg/m2 days 1-5 every 28 days, followed by RT 60 Gy in 30 fractions or RT only. Patients without progressive disease after two TMZ cycles, received the third cycle. From March 2005, TMZ 75 mg/m2 was administered daily concomitant with RT. TMZ was recommended first-line treatment at progression. Primary endpoint was overall survival and secondary safety. RESULTS: The study closed prematurely after enrolling 144 patients, 103 with GBM and 41 with AA. Median age was 53 years (range 24-60) and 89 (62%) were male. PS was 0-1 for 133 (92%) patients, 53 (37%) had complete surgical resection and 18 (12%) biopsy. Ninety-two (64%) received TMZ concomitant with RT. Seventy-two (50%) were randomized to neoadjuvant treatment. For the overall study population survival was 20.3 months for RT and 17.7 months for NeoTMZ (p = .76), this not reaching the primary objective. For the preplanned subgroup analysis, we found that NeoTMZ AA patients had a median survival of 95.1 months compared to 35.2 months for RT (p = .022). For patients with GBM, no difference in survival was observed (p = .10). MGMT and IDH status affected outcome. CONCLUSIONS: No advantage of NeoTMZ was noted for the overall study population or subgroup of GBM, while NeoTMZ resulted in 5 years longer median survival for patients diagnosed as AA.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Astrocytoma/therapy , Brain Neoplasms/therapy , Chemoradiotherapy, Adjuvant/methods , Dacarbazine/analogs & derivatives , Glioblastoma/therapy , Neurosurgical Procedures , Radiotherapy, Adjuvant/methods , Adult , Astrocytoma/genetics , Brain Neoplasms/genetics , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/therapeutic use , Female , Glioblastoma/genetics , Humans , Isocitrate Dehydrogenase/genetics , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Mutation , Pilot Projects , Prognosis , Promoter Regions, Genetic , Survival Rate , Temozolomide , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/genetics , X-linked Nuclear Protein/genetics , Young Adult
16.
Acta Neuropathol ; 131(5): 775-91, 2016 May.
Article in English | MEDLINE | ID: mdl-26860727

ABSTRACT

Cell-based therapies are emerging as new promising treatments in stroke. However, their functional mechanism and therapeutic potential during early infarct maturation has so far received little attention. Here, we asked if cell-based delivery of the interleukin-1 receptor antagonist (IL-1Ra), a known neuroprotectant in stroke, can promote neuroprotection, by modulating the detrimental inflammatory response in the tissue at risk. We show by the use of IL-1Ra-overexpressing and IL-1Ra-deficient mice that IL-1Ra is neuroprotective in stroke. Characterization of the cellular and spatiotemporal production of IL-1Ra and IL-1α/ß identifies microglia, not infiltrating leukocytes, as the major sources of IL-1Ra after experimental stroke, and shows IL-1Ra and IL-1ß to be produced by segregated subsets of microglia with a small proportion of these cells co-expressing IL-1α. Reconstitution of whole body irradiated mice with IL-1Ra-producing bone marrow cells is associated with neuroprotection and recruitment of IL-1Ra-producing leukocytes after stroke. Neuroprotection is also achieved by therapeutic injection of IL-1Ra-producing bone marrow cells 30 min after stroke onset, additionally improving the functional outcome in two different stroke models. The IL-1Ra-producing bone marrow cells increase the number of IL-1Ra-producing microglia, reduce the availability of IL-1ß, and modulate mitogen-activated protein kinase (MAPK) signaling in the ischemic cortex. The importance of these results is underlined by demonstration of IL-1Ra-producing cells in the human cortex early after ischemic stroke. Taken together, our results attribute distinct neuroprotective or neurotoxic functions to segregated subsets of microglia and suggest that treatment strategies increasing the production of IL-1Ra by infiltrating leukocytes or microglia may also be neuroprotective if applied early after stroke onset in patients.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Neuroprotective Agents/therapeutic use , Stroke/pathology , Stroke/therapy , Animals , Brain/metabolism , Brain/pathology , Brain Infarction , Disease Models, Animal , Exploratory Behavior , Female , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Strength/genetics , Muscle Strength/physiology , Stroke/genetics , Time Factors
17.
Eur J Nucl Med Mol Imaging ; 43(10): 1824-36, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27102266

ABSTRACT

PURPOSE: The purpose of this study was to determine the ability of dual time-point (DTP) PET/CT with (18)F-FDG to discriminate between malignant and benign lymphadenopathies. The relationship between DTP FDG uptake and glucose metabolism/hypoxia markers in lymphadenopathies was also assessed. METHODS: Patients with suspected lymphoma or recently diagnosed treatment-naive lymphoma were prospectively enrolled for DTP FDG PET/CT (scans 60 min and 180 min after FDG administration). FDG-avid nodal lesions were segmented to yield volume and standardized uptake values (SUV), including SUVmax, SUVmean, cSUVmean (with partial volume correction), total lesion glycolysis (TLG) and cTLG (with partial volume correction). Expression of glucose transporter-1 (GLUT-1), hexokinase-II (HK-II), glucose-6-phosphatase (G6Pase) and hypoxia-inducible factor-1alpha (HIF-1alpha) were assessed with immunohistochemistry and enzyme activity was determined for HK and G6Pase. RESULTS: FDG uptake was assessed in 203 lesions (146 malignant and 57 benign). Besides volume, there were significant increases over time for all parameters, with generally higher levels in the malignant lesions. The retention index (RI) was not able to discriminate between malignant and benign lesions. Volume, SUVmax, TLG and cTLG for both scans were able to discriminate between the two groups statistically, but without complete separation. Glucose metabolism/hypoxia markers were assessed in 15 lesions. TLG and cTLG were correlated with GLUT-1 expression on the 60-min scan. RI-max and RI-mean and SUVmax, SUVmean and cSUVmean on the 60-min scan were significantly correlated with HK-II expression. CONCLUSION: RI was not able to discriminate between malignant and benign lesions, but some of the SUVs were able to discriminate on the 60-min and 180-min scans. Furthermore, FDG uptake was correlated with GLUT-1 and HK-II expression.


Subject(s)
Fluorodeoxyglucose F18/pharmacokinetics , Glucose-6-Phosphatase/metabolism , Hexokinase/metabolism , Lymphoma/diagnostic imaging , Lymphoma/metabolism , Positron Emission Tomography Computed Tomography/methods , Adolescent , Adult , Aged , Algorithms , Diagnosis, Differential , Female , Humans , Image Enhancement/methods , Image Interpretation, Computer-Assisted/methods , Male , Middle Aged , Pilot Projects , Reproducibility of Results , Sensitivity and Specificity , Tumor Burden , Young Adult
18.
J Neurooncol ; 127(2): 381-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26738845

ABSTRACT

High-grade gliomas have an aggressive clinical course and new clinical biomarkers and therapeutic targets are highly needed. WEE1 is a regulator of the G2 checkpoint in glioblastoma (GBM) cells. Inhibition of this kinase has, in experimental glioma studies, been suggested to enhance sensitivity to irradiation and temozolomide. However, expression level and prognostic potential of WEE1 protein in gliomas remain uninvestigated. In this study, glioma samples from 235 patients across all four WHO grades were analyzed by immunohistochemistry. Using image analysis, we calculated the area fraction of WEE1 positive nuclei. We found that WEE1 protein was localized in tumor cell nuclei and expressed in all glioma types and grades. Although WEE1 protein levels are higher in GBMs (mean 24.5%) relative to grade III (mean 14,0%, p < 0.05) and grade II (mean 6.8%, p < 0.001) gliomas, high WEE1 protein was associated with better survival in GBMs (p = 0.002). This was confirmed in multivariate analysis (HR 0.60, p = 0.003) even when adjusted for MGMT status (HR 0.60, p = 0.005). In conclusion, we report a nuclear expression of WEE1 protein in all glioma grades and types. The WEE1 positive nuclear area was correlated with malignancy grade but it was inversely associated with prognosis in GBM. Although WEE1 is a frequently occurring protein and has been proposed as a novel target in GBM, the role of WEE1 in glioma patient survival appears to be connected to the MGMT status and is more complex than previously anticipated.


Subject(s)
Biomarkers, Tumor/metabolism , Brain Neoplasms/metabolism , Cell Cycle Proteins/metabolism , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Glioma/metabolism , Nuclear Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/surgery , Follow-Up Studies , Glioma/pathology , Glioma/surgery , Humans , Immunoenzyme Techniques , Neoplasm Grading , Prognosis , Retrospective Studies , Survival Rate
19.
J Neurooncol ; 130(1): 53-62, 2016 10.
Article in English | MEDLINE | ID: mdl-27510953

ABSTRACT

Glioblastoma (GBM) is the most frequent and malignant brain tumor with an overall survival of only 14.6 months. Although these tumors are treated with surgery, radiation and chemotherapy, recurrence is inevitable. A critical population of tumor cells in terms of therapy, the so-called cancer stem cells (CSCs), has been identified in gliomas and many other cancers. These tumor cells have a stem cell-like phenotype and are suggested to be responsible for tumor growth, chemo- and radio-resistance as well as recurrence. However, functional evidence for migrating glioma cells having a stem cell-like phenotype is currently lacking. In the present study, the aim was to characterize the phenotype of migrating tumor cells using a novel migration assay based on serum-free stem cell medium and patient-derived spheroid cultures. The results showed pronounced migration of five different GBM spheroid cultures, but not of the commercial cell line U87MG. An in vitro limiting dilution assay showed preserved but reduced spheroid formation capacity of migrating cells. Orthotopic xenografting in mice showed preserved but reduced tumorigenic capacity. Profiling of mRNAs revealed no significant deregulation of 16 predefined CSC-related genes and the HOX-gene list in migrating cells compared to spheroids. Determination of GBM molecular subtypes revealed that subtypes of spheroids and migrating cells were identical. In conclusion, migrating tumor cells preserve expression of stem cell markers and functional CSC characteristics. Since CSCs are reported to be highly resistant to therapy, these results emphasize that the CSC phenotype should be taken into consideration in future treatment of GBMs.


Subject(s)
Brain Neoplasms/pathology , Cell Movement/physiology , Gene Expression Regulation, Neoplastic/physiology , Glioma/pathology , Neoplastic Stem Cells/physiology , Transplantation, Heterologous , AC133 Antigen/metabolism , Animals , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Movement/drug effects , Culture Media, Serum-Free/pharmacology , Glioma/mortality , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Kaplan-Meier Estimate , Mice , Microarray Analysis , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , RNA, Messenger/metabolism , SOXB1 Transcription Factors/metabolism , Spheroids, Cellular , Time Factors
20.
J Neurooncol ; 122(3): 517-27, 2015 May.
Article in English | MEDLINE | ID: mdl-25800004

ABSTRACT

The tyrosine kinase receptor c-Met has been suggested to be involved in crucial parts of glioma biology like tumor stemness, growth and invasion. The aim of this study was to investigate the prognostic value of c-Met in a population-based glioma patient cohort. Tissue samples from 238 patients with WHO grade I, II, III and IV tumors were analyzed using immunohistochemical staining and advanced image analysis. Strong c-Met expression was found in tumor cells, blood vessels, and peri-necrotic areas. At the subcellular level, c-Met was identified in the cytoplasm and in the cell membrane. Measurements of high c-Met intensity correlated with high WHO grade (p = 0.006) but no association with survival was observed in patients with WHO grade II (p = 0.09) or III (p = 0.17) tumors. High expression of c-Met was associated with shorter overall survival in patients with glioblastoma multiforme (p = 0.03). However the prognostic effect of c-Met in glioblastomas was time-dependent and only observed in patients who survived more than 8.5 months, and not within the first 8.5 months after diagnosis. This was significant in multivariate analysis (HR 1.99, 95 % CI 1.29-3.08, p = 0.002) adjusted for treatment and the clinical variables age (HR 1.01, 95 % CI 0.99-1.03, p = 0.30), performance status (HR 1.34, 95 % CI 1.17-1.53, p < 0.001), and tumor crossing midline (HR 1.28, 95 % CI 0.79-2.07, p = 0.29). In conclusion, this study showed that high levels of c-Met holds unfavorable prognostic value in glioblastomas.


Subject(s)
Brain Neoplasms/metabolism , Cephalosporins/metabolism , Glioblastoma/metabolism , Melphalan/analogs & derivatives , Brain Neoplasms/diagnosis , Cell Line, Tumor , Cohort Studies , Community Health Planning , Female , Glioblastoma/diagnosis , Humans , Kaplan-Meier Estimate , Magnetic Resonance Imaging , Male , Melphalan/metabolism , Middle Aged , Proportional Hazards Models , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL