Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
BMC Cancer ; 17(1): 178, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28270132

ABSTRACT

BACKGROUND: Glioblastoma is the most frequent and most malignant brain tumor with the patients having a median survival of only 14.6 months. Although glioblastoma patients are treated with surgery, radiation and chemotherapy recurrence is inevitable. A stem-like population of radio- and chemoresistant brain tumor-initiating cells combined with the invasive properties of the tumors is believed to be critical for treatment resistance. In the present study, the aim was to investigate the effect of a novel therapeutic strategy using the lysosomotropic detergent siramesine on glioblastomas. METHODS: Standard glioma cell lines and patient-derived spheroids cultures with tumor-initiating stem-like cells were used to investigate effects of siramesine on proliferation and cell death. Responsible mechanisms were investigated by inhibitors of caspases and cathepsins. Effects of siramesine on migrating tumor cells were investigated by a flat surface migration assay and by implanting spheroids into organotypic rat brain slice cultures followed by confocal time-lapse imaging. Finally the effect of siramesine was investigated in an orthotopic mouse glioblastoma model. Results obtained in vitro and in vivo were confirmed by immunohistochemical staining of histological sections of spheroids, spheroids in brain slice cultures and tumors in mice brains. RESULTS: The results showed that siramesine killed standard glioma cell lines in vitro, and loss of acridine orange staining suggested a compromised lysosomal membrane. Co-treatment of the cell lines with inhibitors of caspases and cathepsins suggested differential involvement in cell death. Siramesine caused tumor cell death and reduced secondary spheroid formation of patient-derived spheroid cultures. In the flat surface migration model siramesine caused tumor cell death and inhibited tumor cell migration. This could not be reproduced in the organotypic three dimensional spheroid-brain slice culture model or in the mice xenograft model. CONCLUSIONS: In conclusion the in vitro results obtained with tumor cells and spheroids suggest a potential of lysosomal destabilizing drugs in killing glioblastoma cells, but siramesine was without effect in the organotypic spheroid-brain slice culture model and the in vivo xenograft model.


Subject(s)
Glioblastoma/drug therapy , Indoles/administration & dosage , Lysosomes/drug effects , Neoplasm Recurrence, Local/drug therapy , Spiro Compounds/administration & dosage , Animals , Cell Death , Cell Line, Tumor , Cell Movement/drug effects , Disease Models, Animal , Glioblastoma/pathology , Humans , Lysosomes/pathology , Mice , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Spheroids, Cellular/drug effects , Xenograft Model Antitumor Assays
2.
J Neurooncol ; 130(3): 439-448, 2016 12.
Article in English | MEDLINE | ID: mdl-27619981

ABSTRACT

Tissue inhibitor of metalloproteinases-1 (TIMP-1) has been associated with poor prognosis and resistance towards chemotherapy in several cancer forms. In a previous study we found an association between a low TIMP-1 tumor immunoreactivity and increased survival for glioblastoma patients, when compared to moderate and high TIMP-1 tumor immunoreactivity. The aim of the present study was to further evaluate TIMP-1 as a biomarker in gliomas by studying TIMP-1 gene copy numbers by fluorescence in situ hybridization (FISH) on 33 glioblastoma biopsies and by measuring levels of TIMP-1 in plasma obtained pre-operatively from 43 patients (31 gliomas including 21 glioblastomas) by enzyme-linked immunosorbent assay (ELISA). The results showed TIMP-1 gene copy numbers per cell ranging from 1 to 5 and the TIMP-1/CEN-X ratio ranging between 0.7 and 1.09, suggesting neither amplification nor loss of the TIMP-1 gene. The TIMP-1 protein levels measured in plasma were not significantly higher than TIMP-1 levels measured in healthy subjects. No correlation was identified between TIMP-1 tumor cell immunoreactivities and the TIMP-1 gene copy numbers or the plasma TIMP-1 levels. In conclusion, high immunohistochemical TIMP-1 protein levels in glioblastomas were not caused by TIMP-1 gene amplification and TIMP-1 in plasma was low and not directly related to tumor TIMP-1 immunoreactivity. The study suggests that TIMP-1 immunohistochemistry is the method of choice for future clinical studies evaluating TIMP-1 as a biomarker in glioblastomas.


Subject(s)
Brain Neoplasms/blood , Brain Neoplasms/genetics , Glioblastoma/blood , Glioblastoma/genetics , Tissue Inhibitor of Metalloproteinase-1/blood , Tissue Inhibitor of Metalloproteinase-1/genetics , Adult , Aged , Aged, 80 and over , Cohort Studies , DNA Copy Number Variations , Denmark , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Male , Middle Aged , Tissue Array Analysis
3.
J Neurooncol ; 128(3): 395-404, 2016 07.
Article in English | MEDLINE | ID: mdl-27063952

ABSTRACT

Glioblastomas always recur despite surgery, radiotherapy and chemotherapy. A key player in the therapeutic resistance may be immature tumor cells with stem-like properties (TSCs) escaping conventional treatment. A group of promising molecular targets are microRNAs (miRs). miRs are small non-coding RNAs exerting post-transcriptional regulation of gene expression. In this study we aimed to identify over-expressed TSC-related miRs potentially amenable for therapeutic targeting. We used non-differentiated glioblastoma spheroid cultures (GSCs) containing TSCs and compared these to xenografts using a NanoString nCounter platform. This revealed 19 over-expressed miRs in the non-differentiated GSCs. Additionally, non-differentiated GSCs were compared to neural stem cells (NSCs) using a microarray platform. This revealed four significantly over-expressed miRs in the non-differentiated GSCs in comparison to the NSCs. The three most over-expressed miRs in the non-differentiated GSCs compared to xenografts were miR-126, -137 and -128. KEGG pathway analysis suggested the main biological function of these over-expressed miRs to be cell-cycle arrest and diminished proliferation. To functionally validate the profiling results suggesting association of these miRs with stem-like properties, experimental over-expression of miR-128 was performed. A consecutive limiting dilution assay confirmed a significantly elevated spheroid formation in the miR-128 over-expressing cells. This may provide potential therapeutic targets for anti-miRs to identify novel treatment options for GBM patients.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Animals , Brain Neoplasms/pathology , Cell Line, Tumor , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Heterografts , Humans , Male , Microarray Analysis , Neoplasm Transplantation , Neural Stem Cells/metabolism , Rats, Nude , Spheroids, Cellular/transplantation
4.
J Neurooncol ; 126(1): 47-55, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26428358

ABSTRACT

Over-expressed microRNAs (miRs) are promising new targets in glioblastoma (GBM) therapy. Inhibition of over-expressed miRs has been shown to diminish GBM proliferation, invasion and angiogenesis, indicating a significant therapeutic potential. However, the methods utilized for miR inhibition have had low translational potential. In clinical trials convection-enhanced delivery (CED) has been applied for local delivery of compounds in the brain. The aim of this study was to determine if safe and efficient miR inhibition was possible by CED of an anti-miR. We used a highly invasive GBM orthotopic xenograft model and targeted a well-validated miR, let-7a, with a 2'-O-methoxyethyl anti-miR with a combined phosphodiester/phosphorothioate backbone to establish an initial proof of concept. In vitro, anti-let-7a was delivered unassisted to the patient-derived T87 glioblastoma spheroid culture. In vivo, anti-let-7a or saline were administered by CED into orthotopic T87-derived tumors. After 1 month of infusion, tumors were removed and tumor mRNA levels of the target-gene High-mobility group AT-hook 2 (HMGA2) were determined. In vitro, 5 days inhibition was superior to 1 day at de-repressing the let-7a target HMGA2 and the inhibition was stable for 24 h. In vivo, anti-miR integrity was preserved in the pumps and no animals showed signs of severe adverse effects attributable to the anti-miR treatment. HMGA2 tumor level was significantly de-repressed in the anti-miR treated animals. The results showed-as an initial proof of concept-that miRs can be efficiently inhibited using CED delivery of anti-miR. The next step is to apply CED for anti-miR delivery focusing on key oncogenic miRs.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/therapy , Glioblastoma/therapy , MicroRNAs/metabolism , Animals , Antineoplastic Agents/administration & dosage , Brain Neoplasms/metabolism , Cell Line, Tumor , Convection , Drug Delivery Systems , Glioblastoma/metabolism , Glioma/pathology , HMGA2 Protein/genetics , HMGA2 Protein/metabolism , Humans , Mice , Mice, Nude , MicroRNAs/genetics , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/therapeutic use , RNA, Messenger/metabolism , Statistics, Nonparametric , Transfection , Xenograft Model Antitumor Assays
5.
Int J Clin Exp Pathol ; 7(7): 3739-51, 2014.
Article in English | MEDLINE | ID: mdl-25120750

ABSTRACT

Cancer stem cell-related (CSC) markers have been suggested to have promising potentials as novel types of prognostic and predictive markers in gliomas. However no single CSC-related marker is currently used in clinical decisions. The aim of this study was to investigate the prognostic value of CD133 and nestin separately and in combination using a novel quantitative approach in a well-characterized population-based cohort of glioma patients. The expression of CD133 and nestin was measured by systematic random sampling in stained paraffin sections from 239 glioma patients diagnosed between 2005 and 2009. We found that the expression of CD133 did not correlate with WHO grade, and there was no association with overall survival (OS). The level of nestin correlated positively with WHO grade. In patients with WHO grade II tumors, a high level of nestin was associated with short progression-free survival (PFS) in multivariate analysis. High levels of co-localization were associated with poor PFS in patients with WHO grade II tumors, but not with OS. We conclude that CD133 was not an independent prognostic factor, but a high level of nestin was associated with poor PFS in patients with WHO grade II tumors. The combination of double-immunofluorescence and automated analysis seems to be a feasible and reproducible approach for investigation of the prognostic potential of biomarkers.


Subject(s)
Antigens, CD/biosynthesis , Biomarkers, Tumor/analysis , Brain Neoplasms/pathology , Glioma/pathology , Glycoproteins/biosynthesis , Nestin/biosynthesis , AC133 Antigen , Aged , Antigens, CD/analysis , Brain Neoplasms/metabolism , Brain Neoplasms/mortality , Female , Fluorescent Antibody Technique , Glioma/metabolism , Glioma/mortality , Glycoproteins/analysis , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplastic Stem Cells/pathology , Nestin/analysis , Peptides/analysis , Prognosis
6.
Int J Clin Exp Pathol ; 6(7): 1294-305, 2013.
Article in English | MEDLINE | ID: mdl-23826410

ABSTRACT

Targeting of lysosomes is a novel therapeutic anti-cancer strategy for killing the otherwise apoptosis-resistant cancer cells. Such strategies are urgently needed for treatment of brain tumors, especially the glioblastoma, which is the most frequent and most malignant type. The aim of the present study was to investigate the presence of lysosomes in astrocytic brain tumors focussing also on the therapy resistant tumor stem cells. Expression of the lysosomal marker LAMP-1 (lysosomal-associated membrane protein-1) was investigated by immunohistochemistry in 112 formalin fixed paraffin embedded astrocytomas and compared with tumor grade and overall patient survival. Moreover, double immunofluorescence stainings were performed with LAMP-1 and the astrocytic marker GFAP and the putative stem cell marker CD133 on ten glioblastomas. Most tumors expressed the LAMP-1 protein in the cytoplasm of the tumor cells, while the blood vessels were positive in all tumors. The percentage of LAMP-1 positive tumor cells and staining intensities increased with tumor grade but variations in tumors of the same grade were also found. No association was found between LAMP-1 expression and patient overall survival in the individual tumor grades. LAMP-1/GFAP showed pronounced co-expression and LAMP-1/CD133 was co-expressed as well suggesting that tumor cells including the proposed tumor stem cells contain lysosomes. The results suggest that high amounts of lysosomes are present in glioblastomas and in the proposed tumor stem cells. Targeting of lysosomes may be a promising novel therapeutic strategy against this highly malignant neoplasm.


Subject(s)
Astrocytoma/chemistry , Biomarkers, Tumor/analysis , Brain Neoplasms/chemistry , Lysosomal Membrane Proteins/analysis , Lysosomes/chemistry , AC133 Antigen , Antigens, CD/analysis , Astrocytoma/mortality , Astrocytoma/pathology , Astrocytoma/therapy , Biopsy , Brain Neoplasms/mortality , Brain Neoplasms/pathology , Brain Neoplasms/therapy , Glial Fibrillary Acidic Protein/analysis , Glycoproteins/analysis , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lysosomes/pathology , Multivariate Analysis , Neoplasm Grading , Neoplastic Stem Cells/chemistry , Peptides/analysis , Prognosis , Proportional Hazards Models
7.
Am J Pathol ; 182(2): 540-52, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23219427

ABSTRACT

Human leukocyte antigen-G (HLA-G) is a nonclassical major histocompatibility complex (MHC) class I molecule involved in immune tolerance processes, playing an important role in the maintenance of the semi-allogeneic fetus. Although HLA-G expression is restricted in normal tissues, it is broadly expressed in malignant tumors and may favor tumor immune escape. We analyzed HLA-G protein and mRNA expression in tumor samples from patients with glioblastoma collected in France, Denmark, and Brazil. We found HLA-G protein expression in 65 of 108 samples and mRNA in 20 of 21 samples. The absence of HLA-G protein expression was associated with a better long-term survival rate. The mechanisms underlying HLA-G gene expression were investigated in glioma cell lines U251MG, D247MG, and U138MG. Induction of HLA-G transcriptional activity was dependent of 5-aza-2'-deoxycytidine treatment and enhanced by interferon-γ. HLA-G protein expression was observed in U251MG cells only. These cells exhibited a permissive chromatin state at the HLA-G gene promoter and the highest levels of induced HLA-G transcriptional activity following 5-aza-2'-deoxycytidine treatment. Several antigen-presenting machinery components were up-regulated in U251MG cells after demethylating and IFN-γ treatments, suggesting an effect on the up-regulation of HLA-G cell surface expression. Therefore, because of its role in tumor tolerance, HLA-G found to be expressed in glioblastoma samples should be taken into consideration in clinical studies on the pathology and in the design of therapeutic strategies to prevent its expression in HLA-G-negative tumors.


Subject(s)
Azacitidine/analogs & derivatives , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Glioblastoma/genetics , Glioblastoma/immunology , HLA-G Antigens/genetics , Interferon-gamma/pharmacology , Acetylation/drug effects , Aged , Antigen Presentation/drug effects , Antigen Presentation/immunology , Azacitidine/pharmacology , Biopsy , Brain Neoplasms/pathology , Cell Line, Tumor , Chromatin Assembly and Disassembly/drug effects , DNA Methylation/drug effects , DNA Methylation/genetics , Decitabine , Female , Glioblastoma/pathology , HLA-G Antigens/metabolism , Humans , Male , Middle Aged , Paraffin Embedding , Prognosis , Promoter Regions, Genetic/genetics , Survival Analysis , Transcription, Genetic/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics , beta 2-Microglobulin/metabolism
8.
J Histochem Cytochem ; 59(4): 391-407, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21411810

ABSTRACT

The putative tumor stem cell marker CD133 is the marker of choice for identifying brain tumor stem cells in gliomas, but the use of different CD133 antibody clones possibly recognizing different CD133 splice variants with epitopes of different glycosylation status confuses the field. The aim was to investigate if current inconsistent CD133 observations could be a result of using different CD133 antibodies for immunohistochemical identification of CD133. Ten glioblastomas were immunohistochemically stained with four different CD133 antibody clones (AC133, W6B3C1, C24B9, and ab19898) and analyzed by quantitative stereology. Moreover, the CD133 staining pattern of each antibody clone was investigated in kidney, pancreas, and placenta tissue as well as in glioblastoma and retinoblastoma cultures and cell lines. All antibody clones revealed CD133+ niches and single cells in glioblastomas, but when using different clones, their distribution rarely corresponded. Morphology of identified single cells varied, and staining of various tissues, cultures, and cells lines was also inconsistent among the clones. In conclusion, the authors report inconsistent CD133 detection when using different primary CD133 antibody clones. Thus, direct comparison of studies using different antibody clones and conclusions based on CD133 immunohistochemistry should be performed with caution.


Subject(s)
Antibodies/immunology , Antigens, CD/biosynthesis , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Glycoproteins/biosynthesis , AC133 Antigen , Adult , Aged , Antigens, CD/immunology , Biomarkers, Tumor/biosynthesis , Biomarkers, Tumor/immunology , Brain Neoplasms/blood supply , Epitopes , Female , Glioblastoma/blood supply , Glycoproteins/immunology , Glycosylation , Humans , Immunohistochemistry , Kidney/metabolism , Male , Middle Aged , Neoplastic Stem Cells/metabolism , Organ Specificity , Pancreas/metabolism , Peptides/immunology , Placenta/metabolism , Pregnancy , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...