Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
PLoS One ; 14(10): e0222717, 2019.
Article in English | MEDLINE | ID: mdl-31603915

ABSTRACT

INTRODUCTION: The subventricular zone (SVZ) in the brain is associated with gliomagenesis and resistance to treatment in glioblastoma. In this study, we investigate the prognostic role and biological characteristics of subventricular zone (SVZ) involvement in glioblastoma. METHODS: We analyzed T1-weighted, gadolinium-enhanced MR images of a retrospective cohort of 647 primary glioblastoma patients diagnosed between 2005-2013, and performed a multivariable Cox regression analysis to adjust the prognostic effect of SVZ involvement for clinical patient- and tumor-related factors. Protein expression patterns of a.o. markers of neural stem cellness (CD133 and GFAP-δ) and (epithelial-) mesenchymal transition (NF-κB, C/EBP-ß and STAT3) were determined with immunohistochemistry on tissue microarrays containing 220 of the tumors. Molecular classification and mRNA expression-based gene set enrichment analyses, miRNA expression and SNP copy number analyses were performed on fresh frozen tissue obtained from 76 tumors. Confirmatory analyses were performed on glioblastoma TCGA/TCIA data. RESULTS: Involvement of the SVZ was a significant adverse prognostic factor in glioblastoma, independent of age, KPS, surgery type and postoperative treatment. Tumor volume and postoperative complications did not explain this prognostic effect. SVZ contact was associated with increased nuclear expression of the (epithelial-) mesenchymal transition markers C/EBP-ß and phospho-STAT3. SVZ contact was not associated with molecular subtype, distinct gene expression patterns, or markers of stem cellness. Our main findings were confirmed in a cohort of 229 TCGA/TCIA glioblastomas. CONCLUSION: In conclusion, involvement of the SVZ is an independent prognostic factor in glioblastoma, and associates with increased expression of key markers of (epithelial-) mesenchymal transformation, but does not correlate with stem cellness, molecular subtype, or specific (mi)RNA expression patterns.


Subject(s)
Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , CCAAT-Enhancer-Binding Protein-beta/genetics , Glioblastoma/genetics , Lateral Ventricles/metabolism , STAT3 Transcription Factor/genetics , AC133 Antigen/genetics , AC133 Antigen/metabolism , Aged , Biomarkers, Tumor/metabolism , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/mortality , Brain Neoplasms/surgery , CCAAT-Enhancer-Binding Protein-beta/metabolism , DNA Copy Number Variations , Epithelial-Mesenchymal Transition , Female , Gene Expression , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glioblastoma/diagnostic imaging , Glioblastoma/mortality , Glioblastoma/surgery , Humans , Lateral Ventricles/diagnostic imaging , Lateral Ventricles/pathology , Lateral Ventricles/surgery , Magnetic Resonance Imaging , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Polymorphism, Single Nucleotide , Prognosis , Proportional Hazards Models , Retrospective Studies , STAT3 Transcription Factor/metabolism , Tumor Burden
2.
Neurooncol Adv ; 1(1): vdz025, 2019.
Article in English | MEDLINE | ID: mdl-32642660

ABSTRACT

BACKGROUND: The antiepileptic drug valproic acid (VPA) inhibits histone deacetylase in glioblastoma cells in vitro, which influences several oncogenic pathways and decreases glioma cell proliferation. The clinical relevance of these observations remains unclear, as VPA does not seem to affect glioblastoma patient survival. In this study, we analyzed whether the in vitro effects of VPA treatment on histone acetylation are also observed in tumor tissues of glioblastoma patients. METHODS: The in vitro effects of VPA treatment on histone acetylation were assessed with immunofluorescence and western blotting. On tissue microarrays and in fresh-frozen glioblastoma tissues we investigated the histone acetylation patterns of patients who were either treated with VPA or did not receive antiepileptic drugs at the time of their surgery. We also performed mRNA expression-based and gene set enrichment analyses on these tissues. RESULTS: VPA increased the expression levels of acetylated histones H3 and H4 in vitro, in agreement with previous reports. In tumor samples obtained from glioblastoma patients, however, VPA treatment affected neither gene (set) expression nor histone acetylation. CONCLUSIONS: The in vitro effects of VPA on histone acetylation status in glioblastoma cells could not be confirmed in clinical tumor samples of glioblastoma patients using antiepileptic doses of VPA, which reflects the lack of effect of VPA on the clinical outcome of glioblastoma patients.

3.
Int J Pharm ; 537(1-2): 1-8, 2018 Feb 15.
Article in English | MEDLINE | ID: mdl-29225099

ABSTRACT

With the progression towards personalised and age-appropriate medicines, the production of drug loaded liposomes at the point of care would be highly desirable. In particular, liposomal solubilisation agents that can be produced rapidly and easily would provide a new option in personalised medicines. Such a process could also be used as a rapid tool for the formulation and pre-clinical screening of low soluble drugs. Within this paper, we outline a novel easy-to-use production method for point of use production of liposome solubilised drugs. Our results demonstrate that pre-formed multilamellar liposomes, stored in a fresh or frozen format, can be bilayer loaded with low solubility drugs using a simple bath sonication process. Sonication is undertaken in a sealed vial allowing the contents to remain sterile. Liposomes around 100 nm were prepared and these liposomes were able to increase the amount of drug dissolved by up to 10 fold. These liposomal solubilisation agents were stable in terms of size and drug solubilisation for up to 8 days when stored in the fridge making them an easy to use and robust small-scale tool for drug solubilisation.


Subject(s)
Liposomes/chemistry , Pharmaceutical Preparations/chemistry , Chemistry, Pharmaceutical/methods , Particle Size , Phospholipids/chemistry , Point-of-Care Systems , Solubility , Sonication/methods
4.
Talanta ; 174: 780-788, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28738654

ABSTRACT

An analytical procedure to measure the whole-body uptake of pharmaceuticals in zebrafish has been developed using state-of-the-art methodologies. A sample preparation procedure for 9 pharmaceuticals displaying a variety in physicochemical properties was developed using 10-day old zebrafish (TG898). For an efficient homogenization of the samples and subsequent recovery of the compounds of interest, different amounts of organic solvents in combination with acidic modifiers were added to zebrafish samples. Samples were subsequently processed using a powerful bath sonicator and centrifuged. Supernatant was then removed and evaporated in a vacuum oven before being reconstituted in a mobile phase-like solvent. Samples were analyzed using ultra-high performance liquid chromatography (UHPLC) on an Acquity BEH C18 column (100 × 2.1mm, dp=1.7µm) coupled to a Waters Xevo TQ-S mass spectrometer. For this purpose, a generic gradient was run, wherein the percentage of acetonitrile was varied from 3% to 82% in 10.5min at a flow rate of 0.41mL/min. Linearity of the method was demonstrated for all compounds (R2 > 0.997) in a practically relevant concentration range. Matrix effects were between 81% and 106%, except for amitriptyline (51%). Using this method, it was demonstrated that a sample pretreatment using 1:2 (v/v) water:methanol in combination with 0.1% formic acid resulted in acceptable recoveries between 74% and 100% for all compounds. Together with the obtained lower limits of quantification of the analytical method (between 0.005 and 1.5ng/mL), this allowed the use of a single zebrafish to study the whole-body uptake of a particular drug, after incubating zebrafish at the maximum tolerated concentration for this drug.


Subject(s)
Chromatography, High Pressure Liquid/methods , Limit of Detection , Pharmaceutical Preparations/metabolism , Tandem Mass Spectrometry/methods , Zebrafish/metabolism , Animals , Biological Transport , Molecular Weight , Pharmaceutical Preparations/chemistry
5.
Neuro Oncol ; 19(1): 66-77, 2017 01.
Article in English | MEDLINE | ID: mdl-27370398

ABSTRACT

BACKGROUND: Patients with glioblastoma (GBM) have an overall median survival of 15 months despite multimodal therapy. These catastrophic survival rates are to be correlated to systematic relapses that might arise from remaining glioblastoma stem cells (GSCs) left behind after surgery. In this line, it has recently been demonstrated that GSCs are able to escape the tumor mass and preferentially colonize the adult subventricular zone (SVZ). At a distance from the initial tumor site, these GSCs might therefore represent a high-quality model of clinical resilience to therapy and cancer relapses as they specifically retain tumor-initiating abilities. METHOD: While relying on recent findings that have validated the existence of GSCs in the human SVZ, we questioned the role of the SVZ niche as a potential GSC reservoir involved in therapeutic failure. RESULTS: Our results demonstrate that (i) GSCs located in the SVZ are specifically resistant to radiation in vivo, (ii) these cells display enhanced mesenchymal roots that are known to be associated with cancer radioresistance, (iii) these mesenchymal traits are specifically upregulated by CXCL12 (stromal cell-derived factor-1) both in vitro and in the SVZ environment, (iv) the amount of SVZ-released CXCL12 mediates GBM resistance to radiation in vitro, and (v) interferes with the CXCL12/CXCR4 signalling system, allowing weakening of the tumor mesenchymal roots and radiosensitizing SVZ-nested GBM cells. CONCLUSION: Together, these data provide evidence on how the adult SVZ environment, through the release of CXCL12, supports GBM therapeutic failure and potential tumor relapse.


Subject(s)
Brain Neoplasms/pathology , Chemokine CXCL12/metabolism , Cranial Irradiation/adverse effects , Glioblastoma/pathology , Lateral Ventricles/pathology , Neoplastic Stem Cells/pathology , Radiation Tolerance , Animals , Brain Neoplasms/metabolism , Brain Neoplasms/radiotherapy , Gamma Rays/adverse effects , Glioblastoma/metabolism , Glioblastoma/radiotherapy , Humans , Lateral Ventricles/metabolism , Lateral Ventricles/radiation effects , Mice , Mice, Nude , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/radiation effects , Signal Transduction/radiation effects , Tumor Cells, Cultured
6.
Int J Oncol ; 48(6): 2445-52, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27098015

ABSTRACT

Casein kinase II contributes to the growth and survival of malignant gliomas and attracts increasing attention as a therapeutic target in these tumors. Several reports have suggested that this strategy might be most relevant for specific subgroups of patients, namely Verhaak's classical and TP53 wild-type tumors. Using kinase assays and microarray genetic profiling in a series of 27 proprietary fresh frozen surgical glioma samples, we showed that constitutive CK2 kinase activation is not restricted to tumors that present increased copy numbers or mRNA expression of its catalytic or regulatory subunits, and can result from a functional activation by various cytokines from the glioma microenvironment. Using corresponding primary tumor and human astrocyte cell cultures as well as glioma cell lines, we confirmed that CK2 inhibition is selectively toxic to malignant glial tumors, without any restriction to tumor class or to TP53 status. We finally showed that while the contribution of CK2 to the constitutive NF-κB hyperactivation in malignant gliomas is at best moderate, a delayed activation of NF-κB may associate with the therapeutic resistance of glioma cells to CK2 inhibition.


Subject(s)
Brain Neoplasms/enzymology , Gene Expression Profiling/methods , Glioblastoma/enzymology , Tissue Array Analysis/methods , Apigenin/pharmacology , Brain Neoplasms/genetics , Casein Kinase II/antagonists & inhibitors , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Enzyme Activation , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Humans , Naphthyridines/pharmacology , Phenazines , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics
7.
Neuro Oncol ; 18(5): 700-6, 2016 05.
Article in English | MEDLINE | ID: mdl-26420896

ABSTRACT

BACKGROUND: Epileptogenic glioblastomas are thought to convey a favorable prognosis, either due to early diagnosis or potential antitumor effects of antiepileptic drugs. We investigated the relationship between survival and epilepsy at presentation, early diagnosis, and antiepileptic drug therapy in glioblastoma patients. METHODS: Multivariable Cox regression was applied to survival data of 647 consecutive patients diagnosed with de novo glioblastoma between 2005 and 2013 in order to investigate the association between epilepsy and survival in glioblastoma patients. In addition, we quantified the association between survival and valproic acid (VPA) treatment. RESULTS: Epilepsy correlated positively with survival (HR: 0.75 (95% CI: 0.61-0.92), P < .01). This effect is independent of age, sex, performance status, type of surgery, adjuvant therapy, tumor location, and tumor volume, suggesting that this positive correlation cannot be attributed solely to early diagnosis. For patients who presented with epilepsy, the use of the antiepileptic drug VPA did not associate with survival when compared with patients who did not receive VPA treatment. CONCLUSION: Epilepsy is an independent prognostic factor for longer survival in glioblastoma patients. This prognostic effect is not solely explained by early diagnosis, and survival is not associated with VPA treatment.


Subject(s)
Brain Neoplasms/complications , Brain Neoplasms/mortality , Epilepsy/etiology , Glioblastoma/complications , Glioblastoma/mortality , Adult , Anticonvulsants/therapeutic use , Epilepsy/drug therapy , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Proportional Hazards Models , Tissue Array Analysis , Valproic Acid/therapeutic use
8.
Int J Cancer ; 137(7): 1630-42, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-25802036

ABSTRACT

Glioblastoma multiforme (GBM) is the most common primary brain tumor and is without exception lethal. GBMs modify the immune system, which contributes to the aggressive nature of the disease. Particularly, cells of the monocytic lineage, including monocytes, macrophages and microglia, are affected. We investigated the influence of GBM-derived extracellular vesicles (EVs) on the phenotype of monocytic cells. Proteomic profiling showed GBM EVs to be enriched with proteins functioning in extracellular matrix interaction and leukocyte migration. GBM EVs appeared to skew the differentiation of peripheral blood-derived monocytes to alternatively activated/M2-type macrophages. This was observed for EVs from an established cell line, as well as for EVs from primary cultures of GBM stem-like cells (GSCs). Unlike EVs of non-GBM origin, GBM EVs induced modified expression of cell surface proteins, modified cytokine secretion (e.g., an increase in vascular endothelial growth factor and IL-6) and increased phagocytic capacity of the macrophages. Most pronounced effects were observed upon incubation with EVs from mesenchymal GSCs. GSC EVs also affected primary human microglia, resulting in increased expression of Membrane type 1-matrix metalloproteinase, a marker for GBM microglia and functioning as tumor-supportive factor. In conclusion, GBM-derived EVs can modify cells of the monocytic lineage, which acquire characteristics that resemble the tumor-supportive phenotypes observed in patients.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Leukocytes, Mononuclear/pathology , Brain Neoplasms/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Exosomes/metabolism , Exosomes/pathology , Glioblastoma/metabolism , Humans , Leukocytes, Mononuclear/metabolism , Macrophages/metabolism , Macrophages/pathology , Microglia/metabolism , Microglia/pathology , Phenotype
9.
Int J Cancer ; 137(5): 1047-57, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-25620078

ABSTRACT

Osteopontin (OPN) is a secreted protein involved in most aspects of tumor progression and metastasis development. Elevated OPN expression has been reported in multiple types of cancer including glioblastoma (GBM), the highest grade and most aggressive brain tumor. GBMs contain a subpopulation of glioma-initiating cells (GICs) implicated in progression, therapeutic resistance and recurrence. We have previously demonstrated that OPN silencing inhibited GBM cell growth in vitro and in vivo. Moreover, activation of CD44 signaling upon OPN ligation has been recently implicated in the acquisition of a stem cell phenotype by GBM cells. The present study is aimed to explore OPN autocrine function using shRNA silencing strategy in GICs enriched from GBM cell lines and a human primary GBM grown in EGF and bFGF defined medium. The removal of these growth factors and addition of serum induced a significant loss of OPN expression in GICs. We showed that OPN-silenced GICs were unable to grow as spheres and this capacity was restored by exogenous OPN. Importantly, the expression of Sox2, Oct3/4 and Nanog, key stemness transcription factors, was significantly decreased in GICs upon OPN targeting. We identified Akt/mTOR/p70S6K as the main signaling pathway triggered following OPN-mediated EGFR activation in GICs. Finally, in an orthotopic xenograft mouse model, the tumorigenic potential of U87-MG sphere cells was completely abrogated upon OPN silencing. Our demonstration of endogenous OPN major regulatory effects on GICs stemness phenotype and tumorigenicity implies a greater role than anticipated for OPN in GBM pathogenesis from initiation and progression to probable recurrence.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Neoplastic Stem Cells/physiology , Osteopontin/antagonists & inhibitors , Animals , Autocrine Communication , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Gene Silencing , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , Neoplasm Transplantation , Osteopontin/metabolism , RNA, Small Interfering/metabolism , Spheroids, Cellular/metabolism
10.
Neuro Oncol ; 17(1): 81-94, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25085362

ABSTRACT

BACKGROUND: Patients with glioblastoma multiforme (GBM) have an overall median survival of 15 months. This catastrophic survival rate is the consequence of systematic relapses that could arise from remaining glioblastoma stem cells (GSCs) left behind after surgery. We previously demonstrated that GSCs are able to escape the tumor mass and specifically colonize the adult subventricular zones (SVZs) after transplantation. This specific localization, away from the initial injection site, therefore represents a high-quality model of a clinical obstacle to therapy and relapses because GSCs notably retain the ability to form secondary tumors. METHOD: In this work, we questioned the role of the CXCL12/CXCR4 signaling in the GSC-specific invasion of the SVZs. RESULTS: We demonstrated that both receptor and ligand are respectively expressed by different GBM cell populations and by the SVZ itself. In vitro migration bio-assays highlighted that human U87MG GSCs isolated from the SVZs (U87MG-SVZ) display stronger migratory abilities in response to recombinant CXCL12 and/or SVZ-conditioned medium (SVZ-CM) compared with cancer cells isolated from the tumor mass (U87MG-TM). Moreover, in vitro inhibition of the CXCR4 signaling significantly decreased the U87MG-SVZ cell migration in response to the SVZ-CM. Very interestingly, treating U87MG-xenografted mice with daily doses of AMD3100, a specific CXCR4 antagonist, prevented the specific invasion of the SVZ. Another in vivo experiment, using CXCR4-invalidated GBM cells, displayed similar results. CONCLUSION: Taken together, these data demonstrate the significant role of the CXCL12/CXCR4 signaling in this original model of brain cancer invasion.


Subject(s)
Brain Neoplasms/metabolism , Chemokine CXCL12/metabolism , Glioblastoma/metabolism , Lateral Ventricles/metabolism , Neoplasm Invasiveness/physiopathology , Neoplastic Stem Cells/metabolism , Receptors, CXCR4/metabolism , Animals , Benzylamines , Brain Neoplasms/pathology , Cell Line, Tumor , Cyclams , Disease Models, Animal , Female , Glioblastoma/pathology , Heterocyclic Compounds/pharmacology , Humans , Lateral Ventricles/drug effects , Lateral Ventricles/pathology , Mice , Mice, Nude , Receptors, CXCR4/antagonists & inhibitors , Signal Transduction
11.
Neuro Oncol ; 17(3): 392-406, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25155356

ABSTRACT

BACKGROUND: Glioblastomas remain ominous tumors that almost invariably escape treatment. Connexins are a family of transmembrane, gap junction-forming proteins, some members of which were reported to act as tumor suppressors and to modulate cellular metabolism in response to cytotoxic stress. METHODS: We analyzed the copy number and expression of the connexin (Cx)30 gene gap junction beta-6 (GJB6), as well as of its protein immunoreactivity in several public and proprietary repositories of glioblastomas, and their influence on patient survival. We evaluated the effect of the expression of this gap junction protein on the growth, DNA repair and energy metabolism, and treatment resistance of these tumors. RESULTS: The GJB6 gene was deleted in 25.8% of 751 analyzed tumors and mutated in 15.8% of 158 tumors. Cx30 immunoreactivity was absent in 28.9% of 145 tumors. Restoration of Cx30 expression in human glioblastoma cells reduced their growth in vitro and as xenografts in the striatum of immunodeficient mice. Cx30 immunoreactivity was, however, found to adversely affect survival in 2 independent retrospective cohorts of glioblastoma patients. Cx30 was found in clonogenic assays to protect glioblastoma cells against radiation-induced mortality and to decrease radiation-induced DNA damage. This radioprotection correlated with a heat shock protein 90-dependent mitochondrial translocation of Cx30 following radiation and an improved ATP production following this genotoxic stress. CONCLUSION: These results underline the complex relationship between potential tumor suppressors and treatment resistance in glioblastomas and single out GJB6/Cx30 as a potential biomarker and target for therapeutic intervention in these tumors.


Subject(s)
Brain Neoplasms/genetics , Connexins/genetics , Glioma/genetics , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/mortality , Brain Neoplasms/radiotherapy , Cell Line, Tumor , Cell Proliferation/genetics , Connexin 30 , Connexins/metabolism , Gene Deletion , Glioma/mortality , Glioma/radiotherapy , HSP90 Heat-Shock Proteins/metabolism , Heterografts/metabolism , Humans , Inhibitor of Apoptosis Proteins/metabolism , Kaplan-Meier Estimate , Mice , Mitochondria/metabolism , RNA, Messenger/metabolism , Survivin
12.
Cancers (Basel) ; 5(3): 1049-71, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-24202333

ABSTRACT

Glioblastoma multiforme (GBM, WHO grade IV) is the most common and lethal subtype of primary brain tumor with a median overall survival of 15 months from the time of diagnosis. The presence in GBM of a cancer population displaying neural stem cell (NSC) properties as well as tumor-initiating abilities and resistance to current therapies suggests that these glioblastoma-initiating cells (GICs) play a central role in tumor development and are closely related to NSCs. However, it is nowadays still unclear whether GICs derive from NSCs, neural progenitor cells or differentiated cells such as astrocytes or oligodendrocytes. On the other hand, NSCs are located in specific regions of the adult brain called neurogenic niches that have been shown to control critical stem cell properties, to nourish NSCs and to support their self-renewal. This "seed-and-soil" relationship has also been adapted to cancer stem cell research as GICs also require a specific micro-environment to maintain their "stem cell" properties. In this review, we will discuss the controversies surrounding the origin and the identification of GBM stem cells and highlight the micro-environment impact on their biology.

13.
Int J Oncol ; 41(2): 776-82, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22614258

ABSTRACT

Inhibitors of casein kinase 2 (CK2), a regulator of cell proliferation and mediator of the DNA damage response, are being evaluated in clinical trials for the treatment of cancers. Apigenin was capable of inhibiting the activation of CK2 following γ irradiation in LN18 and U87 malignant glioma cells. Apigenin and siRNA-mediated CK2 protein depletion further inhibited NF-κB activation and altered the Tyr68 phosphorylation of Chk2 kinase, a DNA damage response checkpoint kinase, following irradiation. However, CK2 inhibition did not decrease the ability of these glioma cells to repair double-strand DNA breaks, as assessed by COMET assays and γ-H2Ax staining. Likewise, apigenin and siRNA-induced depletion of CK2 failed to sensitize glioma cells to the cytotoxic effect of 2 to 10 G-rays of γ irradiation, as assessed by clonogenic assays. These results contrast with those found in other cancer types, and urge to prudence regarding the inclusion of malignant glioma patients in clinical trials that assess the radiosensitizing role of CK2 inhibitors in solid cancers.


Subject(s)
Apigenin/pharmacology , Casein Kinase II/antagonists & inhibitors , DNA Damage , Glioma/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Analysis of Variance , Casein Kinase II/genetics , Casein Kinase II/metabolism , Cell Line, Tumor/drug effects , Cell Line, Tumor/radiation effects , Cell Survival/radiation effects , Checkpoint Kinase 2 , DNA Repair/drug effects , Enzyme Activation/radiation effects , Gene Knockdown Techniques , Humans , NF-kappa B/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Interference , Radiation Tolerance/drug effects , Statistics, Nonparametric
14.
Clin Neuropathol ; 31(6): 430-4, 2012.
Article in English | MEDLINE | ID: mdl-22541786

ABSTRACT

BACKGROUND: Multiple choroid plexus papillomas (CPPs) are rare. Usually, they correspond to villous hypertrophy or metastasis occurring during cerebrospinal dissemination. Multiple CPPs have rarely been reported as synchronous tumors. CASE REPORT: Three synchronous CPPs were resected in a 59-year-old female 6 years after their first imaging description. Pathology showed mucus-producing CPP in all 3, 1 of the 3 presenting some signs of atypia. No p53 or hSNF5/INI1 mutation, or signs of polyoma viruses infection were found. CONCLUSION: Although no clear cause for the multifocality was found, the simultaneous presence of the three tumors and their benign histology suggest that they were synchronous and not metastatic. The issue of differentiating synchronous CPPs from metastatic CPP is discussed.


Subject(s)
Neoplasms, Multiple Primary/pathology , Papilloma, Choroid Plexus/pathology , Female , Humans , Hydrocephalus, Normal Pressure/complications , Hydrocephalus, Normal Pressure/surgery , Middle Aged , Neoplasms, Multiple Primary/complications , Papilloma, Choroid Plexus/complications , Ventriculoperitoneal Shunt
15.
Int J Cancer ; 129(3): 574-85, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-20886597

ABSTRACT

In patients with glioblastoma multiforme, recurrence is the rule despite continuous advances in surgery, radiotherapy and chemotherapy. Within these malignant gliomas, glioblastoma stem cells or initiating cells have been recently described, and they were shown to be specifically involved in experimental tumorigenesis. In this study, we show that some human glioblastoma cells injected into the striatum of immunodeficient nude mice exhibit a tropism for the subventricular zones. There and similarily to neurogenic stem cells, these subventricular glioblastoma cells were then able to migrate toward the olfactory bulbs. Finally, the glioblastoma cells isolated from the adult mouse subventricular zones and olfactory bulbs display high tumorigenicity when secondary injected in a new mouse brain. Together, these data suggest that neurogenic zones could be a reservoir for particular cancer-initiating cells.


Subject(s)
Brain Neoplasms/pathology , Cell Movement , Cerebral Ventricles/pathology , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Olfactory Bulb , Animals , Corpus Striatum , Female , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL
...