Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Genes Dev ; 28(10): 1085-100, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24788093

ABSTRACT

Glioblastomas are the most prevalent and lethal primary brain tumor and are comprised of hierarchies with self-renewing cancer stem cells (CSCs) at the apex. Like neural stem cells (NSCs), CSCs reside in functional niches that provide essential cues to maintain the cellular hierarchy. Bone morphogenetic proteins (BMPs) instruct NSCs to adopt an astrocyte fate and are proposed as anti-CSC therapies to induce differentiation, but, paradoxically, tumors express high levels of BMPs. Here we demonstrate that the BMP antagonist Gremlin1 is specifically expressed by CSCs as protection from endogenous BMPs. Gremlin1 colocalizes with CSCs in vitro and in vivo. Furthermore, Gremlin1 blocks prodifferentiation effects of BMPs, and overexpression of Gremlin1 in non-CSCs decreases their endogenous BMP signaling to promote stem-like features. Consequently, Gremlin1-overexpressing cells display increased growth and tumor formation abilities. Targeting Gremlin1 in CSCs results in impaired growth and self-renewal. Transcriptional profiling demonstrated that Gremlin1 effects were associated with inhibition of p21(WAF1/CIP1), a key CSC signaling node. This study establishes CSC-derived Gremlin1 as a driving force in maintaining glioblastoma tumor proliferation and glioblastoma hierarchies through the modulation of endogenous prodifferentiation signals.


Subject(s)
Glioma/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neoplastic Stem Cells/metabolism , Animals , Bone Morphogenetic Protein 2/metabolism , Cell Cycle/genetics , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioma/genetics , Glioma/pathology , Heterografts , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Neoplastic Stem Cells/pathology , Signal Transduction
2.
Nature ; 478(7368): 197-203, 2011 Oct 05.
Article in English | MEDLINE | ID: mdl-21976023

ABSTRACT

Activation of the aryl hydrocarbon receptor (AHR) by environmental xenobiotic toxic chemicals, for instance 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin), has been implicated in a variety of cellular processes such as embryogenesis, transformation, tumorigenesis and inflammation. But the identity of an endogenous ligand activating the AHR under physiological conditions in the absence of environmental toxic chemicals is still unknown. Here we identify the tryptophan (Trp) catabolite kynurenine (Kyn) as an endogenous ligand of the human AHR that is constitutively generated by human tumour cells via tryptophan-2,3-dioxygenase (TDO), a liver- and neuron-derived Trp-degrading enzyme not yet implicated in cancer biology. TDO-derived Kyn suppresses antitumour immune responses and promotes tumour-cell survival and motility through the AHR in an autocrine/paracrine fashion. The TDO-AHR pathway is active in human brain tumours and is associated with malignant progression and poor survival. Because Kyn is produced during cancer progression and inflammation in the local microenvironment in amounts sufficient for activating the human AHR, these results provide evidence for a previously unidentified pathophysiological function of the AHR with profound implications for cancer and immune biology.


Subject(s)
Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Glioma/metabolism , Glioma/pathology , Kynurenine/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Animals , Autocrine Communication , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Cell Line, Tumor , Cell Survival , Disease Progression , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/immunology , Humans , Kynurenine/immunology , Kynurenine/pharmacology , Ligands , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Paracrine Communication , Receptors, Aryl Hydrocarbon/immunology , Tryptophan/metabolism , Tryptophan Oxygenase/deficiency , Tryptophan Oxygenase/genetics , Tryptophan Oxygenase/metabolism
3.
Brain ; 136(Pt 2): 564-76, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23378223

ABSTRACT

Transforming growth factor-ß is a central mediator of the malignant phenotype of glioblastoma, the most common and malignant form of intrinsic brain tumours. Transforming growth factor-ß promotes invasiveness and angiogenesis, maintains cancer cell stemness and induces profound immunosuppression in the host. Integrins regulate cellular adhesion and transmit signals important for cell survival, proliferation, differentiation and motility, and may be involved in the activation of transforming growth factor-ß. We report that αvß3, αvß5 and αvß8 integrins are broadly expressed not only in glioblastoma blood vessels but also in tumour cells. Exposure to αv, ß3 or ß5 neutralizing antibodies, RNA interference-mediated integrin gene silencing or pharmacological integrin inhibition using the cyclic RGD peptide EMD 121974 (cilengitide) results in reduced phosphorylation of Smad2 in most glioma cell lines, including glioma-initiating cell lines and reduced transforming growth factor-ß-mediated reporter gene activity, coinciding with reduced transforming growth factor-ß protein levels in the supernatant. Time course experiments indicated that the loss of transforming growth factor-ß bioactivity due to integrin inhibition likely results from two distinct mechanisms: an early effect on activation of preformed inactive protein, and second, major effect on transforming growth factor-ß gene transcription as confirmed by decreased activity of the transforming growth factor-ß gene promoter and decreased transforming growth factor-ß(1) and transforming growth factor-ß(2) messenger RNA expression levels. In vivo, EMD 121974 (cilengitide), which is currently in late clinical development as an antiangiogenic agent in newly diagnosed glioblastoma, was a weak antagonist of pSmad2 phosphorylation. These results validate integrin inhibition as a promising strategy not only to inhibit angiogenesis, but also to block transforming growth factor-ß-controlled features of malignancy including invasiveness, stemness and immunosuppression in human glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Integrins/physiology , Transforming Growth Factor beta1/biosynthesis , Animals , Brain Neoplasms/drug therapy , Cell Line, Tumor , Glioblastoma/drug therapy , Humans , Integrins/antagonists & inhibitors , Mice , Mice, Nude , Mink , Neural Pathways/physiology , Snake Venoms/pharmacology , Snake Venoms/therapeutic use , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/physiology
4.
Proteomics Clin Appl ; 16(5): e2100049, 2022 09.
Article in English | MEDLINE | ID: mdl-35462455

ABSTRACT

PURPOSE: The study aim is a comparative proteome-based analysis of different autologous bone entities (alveolar bone [AB], iliac cortical [IC] bone, and iliac spongiosa [IS]) used for alveolar onlay grafting. EXPERIMENTAL DESIGN: Site-matched bone samples of AB, IC, and IS were harvested during alveolar onlay grafting. Proteins were extracted using a detergent-based (sodium dodecyl sulfate) strategy and trypsinized. Proteome analysis was performed using liquid chromatography-tandem mass spectrometry (LC-MS/MS). MaxQuant was used for peptide-to-spectrum matching, peak detection, and quantitation. Linear models for microarray analysis (LIMMA) were used to detect differentially abundant peptides and proteins. RESULTS: A total of 1730 different proteins were identified across the 15 samples at a false discovery rate of 1%. Partial least-squares discriminant analysis approved segregation of AB, IC, and IS protein profiles. LIMMA statistics highlighted 66 proteins that were more abundant in AB then in IC (vs. 92 proteins were enriched in IC over AB). Gene Ontology enrichment analysis revealed a matrisomal versus an immune-related proteome fingerprint in AB versus IC. CONCLUSION AND CLINICAL RELEVANCE: This pilot study demonstrates an ECM protein-related proteome fingerprint in AB and an immune-related proteome fingerprint in IS and IC.


Subject(s)
Proteome , Proteomics , Chromatography, Liquid , Detergents/analysis , Humans , Pilot Projects , Proteome/metabolism , Proteomics/methods , Sodium Dodecyl Sulfate , Tandem Mass Spectrometry
5.
Int J Cancer ; 126(6): 1513-20, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-19688824

ABSTRACT

Peritoneal carcinomatosis is a frequent finding in gastric cancer associated with a poor prognosis. The features that enable gastric tumors to disseminate are poorly understood until now. Previously, we showed elevated mRNA levels of phosphoglycerate kinase 1 (PGK1), an adenosine triphosphate-generating enzyme in the glycolytic pathway, the chemokine receptor 4 (CXCR4), the corresponding chemokine ligand 12 (CXCL12) and beta-catenin in specimens from gastric cancer patients with peritoneal carcinomatosis. In this study, the influence of PGK1 on CXCR4 and beta-catenin was assessed as well as the invasiveness of PGK1 overexpressing cancer cells. In this current study, we found that PGK1 regulates the expression of CXCR4 and beta-catenin at the mRNA and protein levels. On the other hand, CXCR4 regulates the expression of PGK1. Plasmid-mediated overexpression of PGK1 dramatically increased the invasiveness of gastric cancer cells. Interestingly, inhibition of CXCR4 in cells overexpressing PGK1 produced only a moderate reduction of invasiveness suggesting that, PGK1 itself has a critical role in tumor invasiveness. Immunohistochemistry in specimens from diffuse gastric cancer patients also revealed an overexpression of PGK1 in patients with development of peritoneal carcinomatosis. Therefore, PGK1 may be a crucial enzyme in peritoneal dissemination. Together these findings suggest that the enhanced expression of PGK1 and its signaling targets CXCR4 and beta-catenin in gastric cancer cells promote peritoneal carcinomatosis. Thus, PGK1 may serve as prognostic marker and/or be a potential therapeutic target to prevent dissemination of gastric carcinoma cells into the peritoneum.


Subject(s)
Gene Expression Regulation, Neoplastic , Peritoneal Neoplasms/secondary , Phosphoglycerate Kinase/genetics , Stomach Neoplasms/pathology , Blotting, Western , Cell Line, Tumor , Cell Movement/genetics , Cell Movement/physiology , Cell Survival/genetics , Cell Survival/physiology , Humans , Immunohistochemistry , Models, Biological , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Phosphoglycerate Kinase/metabolism , RNA Interference , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics , Signal Transduction/physiology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , beta Catenin/genetics , beta Catenin/metabolism
6.
Stem Cells ; 27(4): 909-19, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19353519

ABSTRACT

Mesenchymal stem cells (MSC) display unique suppressive properties on T-cell immunity, thus representing an attractive vehicle for the treatment of conditions associated with harmful T-cell responses such as organ-specific autoimmunity and graft-versus-host disease. Toll-like receptors (TLR) are primarily expressed on antigen-presenting cells and recognize conserved pathogen-derived components. Ligation of TLR activates multiple innate and adaptive immune response pathways to eliminate and protect against invading pathogens. In this work, we show that TLR expressed on human bone marrow-derived MSC enhanced the immunosuppressive phenotype of MSC. Immunosuppression mediated by TLR was dependent on the production of immunosuppressive kynurenines by the tryptophan-degrading enzyme indoleamine-2,3-dioxygenase-1 (IDO1). Induction of IDO1 by TLR involved an autocrine interferon (IFN)-beta signaling loop, which was dependent on protein kinase R (PKR), but independent of IFN-gamma. These data define a new role for TLR in MSC immunobiology, which is to augment the immunosuppressive properties of MSC in the absence of IFN-gamma rather than inducing proinflammatory immune response pathways. PKR and IFN-beta play a central, previously unidentified role in orchestrating the production of immunosuppressive kynurenines by MSC.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase/immunology , Interferon-beta/immunology , Mesenchymal Stem Cells/immunology , Signal Transduction/immunology , Toll-Like Receptors/immunology , eIF-2 Kinase/immunology , Blotting, Western , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Cell Differentiation/immunology , Chromatography, High Pressure Liquid , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-beta/metabolism , Kynurenine/biosynthesis , Kynurenine/immunology , Lymphocyte Culture Test, Mixed , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Toll-Like Receptors/metabolism , eIF-2 Kinase/metabolism
7.
Int J Cancer ; 125(3): 530-40, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19431147

ABSTRACT

High biological activity of the transforming growth factor (TGF)-beta-Smad pathway characterizes the malignant phenotype of malignant gliomas and confers poor prognosis to glioma patients. Accordingly, TGF-beta has become a novel target for the experimental treatment of these tumors. TGF-beta is processed by furin-like proteases (FLP) and secreted from cells in a latent complex with its processed propeptide, the latency-associated peptide (LAP). Latent TGF-beta-binding protein 1 (LTBP-1) covalently binds to this small latent TGF-beta complex (SLC) and regulates its function, presumably via interaction with the extracellular matrix (ECM). We report here that the levels of LTBP-1 protein in vivo increase with the grade of malignancy in gliomas. LTBP-1 is associated with the ECM as well as secreted into the medium in cultured malignant glioma cells. The release of LTBP-1 into the medium is decreased by the inhibition of FLP activity. Gene-transfer mediated overexpression of LTBP-1 in glioma cell lines results in an increase inTGF-beta activity. Accordingly, Smad2 phosphorylation as an intracellular marker of TGF-beta activity is enhanced. Conversely, LTBP-1 gene silencing reduces TGF-beta activity and Smad2 phosphorylation without affecting TGF-beta protein levels. Collectively, we identify LTBP-1 as an important modulator of TGF-beta activation in glioma cells, which may contribute to the malignant phenotype of these tumors.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Latent TGF-beta Binding Proteins/metabolism , Signal Transduction , Smad2 Protein/metabolism , Transforming Growth Factor beta/metabolism , Astrocytoma/metabolism , Cell Line, Tumor , Cell Proliferation , Extracellular Matrix/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Humans , Immunoblotting , Immunohistochemistry , Oligonucleotide Array Sequence Analysis , Phenotype , Phosphorylation , Polymerase Chain Reaction , Up-Regulation
8.
Neuro Oncol ; 17(2): 254-65, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25165192

ABSTRACT

BACKGROUND: The transforming growth factor (TGF)-ß and vascular endothelial growth factor (VEGF) pathways have a major role in the pathogenesis of glioblastoma, notably immunosuppression, migration, and angiogenesis, but their interactions have remained poorly understood. METHODS: We characterized TGF-ß pathway activity in 9 long-term glioma cell lines (LTCs) and 4 glioma-initiating cell lines (GICs) in relation to constitutive and exogenous TGF-ß-induced VEGF release. Results were validated using The Cancer Genome Atlas transcriptomics data. RESULTS: Glioma cells exhibit heterogeneous patterns of constitutive TGF-ß pathway activation reflected by phosphorylation not only of SMAD2 and SMAD3 but also of SMAD1/5/8. Constitutive TGF-ß pathway activity depends on the type I TGF-ß receptor, ALK-5, and accounts for up to 69% of constitutive VEGF release, which is positively regulated by SMAD2/3 and negatively regulated by SMAD1/5/8 signaling in a cell line-specific manner. Exogenous TGF-ß induces VEGF release in most cell lines in a SMAD- and ALK-5-dependent manner. There is no correlation between the fold induction of VEGF secretion induced by TGF-ß compared with hypoxia. The role of SMAD5 signaling is highly context and cell-line dependent with a VEGF inhibitory effect at low TGF-ß and pSMAD2 levels and a stimulatory effect when TGF-ß is abundant. CONCLUSIONS: TGF-ß regulates VEGF release by glioma cells in an ALK-5-dependent manner involving SMAD2, SMAD3, and SMAD1/5/8 signaling. This crosstalk between the TGF-ß and VEGF pathways may open up new avenues of biomarker-driven exploratory clinical trials focusing on the microenvironment in glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad Proteins, Receptor-Regulated/metabolism , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor A/metabolism , Cell Line, Tumor , Gene Expression Regulation , Humans , RNA, Messenger/metabolism , Receptor, Transforming Growth Factor-beta Type I , Smad1 Protein/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Tumor Cells, Cultured
9.
Oncotarget ; 6(8): 5963-77, 2015 Mar 20.
Article in English | MEDLINE | ID: mdl-25849941

ABSTRACT

Transforming growth factor (TGF)-ß is a central molecule maintaining the malignant phenotype of glioblastoma. Anti-TGF-ß strategies are currently being explored in early clinical trials. Yet, there is little contemporary data on the differential expression of TGF-ß isoforms at the mRNA and protein level or TGF-ß/Smad pathway activity in glioblastomas in vivo.Here we studied 64 newly diagnosed and 16 recurrent glioblastomas for the expression of TGF-ß1-3, platelet-derived growth factor (PDGF)-B, and plasminogen activator inhibitor (PAI)-1 mRNA by RT-PCR and for the levels of TGF-ß1-3 protein, phosphorylated Smad2 (pSmad2), pSmad1/5/8 and PAI-1 by immunohistochemistry.Among the TGF-ß isoforms, TGF-ß1 mRNA was the most, whereas TGF-ß3 mRNA was the least abundant. TGF-ß1-3 mRNA expression was strongly correlated, as was the expression of TGF-ß1-3 mRNA, and of the TGF-ß1-3 target genes, PDGF-B and PAI-1. TGF-ß2 and TGF-ß3 protein levels correlated well, whereas the comparison of the other TGF-ßisoforms did not. Positive correlation was also observed between TGF-ß1 and pSmad1/5/8 and between pSmad2 and pSmad1/5/8. Survival analyses indicated that a group of patients with high expression levels of TGF-ß2 mRNA or pSmad1/5/8 protein have inferior outcome.We thus provide potential biomarkers for patient stratification in clinical trials of anti-TGF-ß therapies in glioblastoma.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Transforming Growth Factor beta/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/genetics , Cells, Cultured , Child , Child, Preschool , Female , Glioblastoma/genetics , Humans , Infant , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Phosphorylation , Young Adult
10.
Neuro Oncol ; 16(3): 382-91, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24327582

ABSTRACT

BACKGROUND: There are emerging reports that the family of a disintegrin and metalloproteinases (ADAM) are involved in the maintenance of the malignant phenotype of glioblastomas. Notably, ADAM proteases 10 and 17 might impair the immune recognition of glioma cells via the activating immunoreceptor NKG2D by cleavage of its ligands from the cell surface. Glioblastoma-initiating cells (GIC) with stem cell properties have been identified as an attractive target for immunotherapy. However, GIC immunogenicity seems to be low. METHODS AND RESULTS: Here,we show that ADAM10 and ADAM17 are expressed on the cell surface of GIC and contribute to an immunosuppressive phenotype by cleavage of ULBP2. The cell surface expression of ULBP2 is enhanced upon blocking ADAM10 and ADAM17, and treatment with ADAM10 and ADAM17specific inhibitors leads to enhanced immunerecognition of GIC by natural killer cells. CONCLUSIONS: Therefore, ADAM10 and ADAM17 constitute suitable targets to boost an immune response against GIC.


Subject(s)
ADAM Proteins/metabolism , Amyloid Precursor Protein Secretases/metabolism , Brain Neoplasms/immunology , Glioblastoma/immunology , Membrane Proteins/metabolism , Neoplastic Stem Cells/immunology , ADAM Proteins/antagonists & inhibitors , ADAM10 Protein , ADAM17 Protein , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cell Line, Tumor , Cell Survival , Dipeptides/pharmacology , GPI-Linked Proteins/metabolism , Humans , Hydroxamic Acids/pharmacology , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Receptors, Transforming Growth Factor beta/antagonists & inhibitors
11.
Neurooncol Pract ; 1(4): 178-183, 2014 Dec.
Article in English | MEDLINE | ID: mdl-26034630

ABSTRACT

Lomustine is an oral alkylating drug commonly used for brain tumor patients. Recently, the lomustine-containing PCV polychemotherapy regime (procarbazine, CCNU/lomustine, and vincristine) in combination with radiotherapy has become the standard of care for anaplastic oligodendroglioma with 1p/19q codeletion and high-risk low-grade glioma. Here, we review the literature of all reported cases of lomustine overdose, highlight complications by exemplifying a case of inadvertent lomustine overdose, and outline the management of this potential complication of outpatient PCV therapy.

12.
Clin Cancer Res ; 16(15): 3851-9, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20534737

ABSTRACT

PURPOSE: Growth and differentiation factor (GDF)-15 is a member of the transforming growth factor (TGF)-beta family. GDF-15 is necessary for the maintenance of pregnancy but has also been linked to other physiologic and pathologic conditions. EXPERIMENTAL DESIGN: The expression of GDF-15 in glioma cell lines was assessed by quantitative reverse transcriptase-PCR and immunoblot. GDF-15 levels in situ and in the peripheral blood of glioma patients were examined by immunohistochemistry and enzyme-linked immunosorbent assay, respectively. The effects of short hairpin RNA-mediated GDF-15 inhibition on proliferation and immunogenicity of SMA-560 glioma cells were investigated by [methyl-(3)H]thymidine incorporation and immune-mediated target cell lysis. The impact of GDF-15 on glioma growth in vivo was assessed in syngeneic mice. RESULTS: GDF-15 is expressed by gliomas of different WHO grades as assessed by immunohistochemistry. The high expression of GDF-15 in tumor tissue translates into elevated GDF-15 serum levels in glioblastoma patients compared with healthy controls. GDF-15 mRNA and protein are also detectable in human and mouse glioma cells in vitro. Silencing of GDF-15 by RNA interference reduces the proliferation of malignant glioma cells. Immunologically, the depletion of glioma-derived GDF-15 enhances the susceptibility of mouse glioma cells towards syngeneic natural killer cells and splenocytes. This results in a reduced in vivo tumorigenicity and increased T-cell infiltration of GDF-15-deficient glioma cells in syngeneic mice. CONCLUSIONS: Although previous studies focusing on ectopic overexpression of GDF-15 have proposed unclear or antitumorigenic effects of GDF-15 in glioma cells, we here show that GDF-15 at endogenous levels contributes to proliferation and immune escape of malignant gliomas in an immunocompetent host.


Subject(s)
Brain Neoplasms/metabolism , Cell Proliferation , Glioma/metabolism , Growth Differentiation Factor 15/metabolism , Tumor Escape/genetics , Animals , Blotting, Western , Brain Neoplasms/genetics , Brain Neoplasms/immunology , Cell Line, Tumor , Cytotoxicity, Immunologic , Enzyme-Linked Immunosorbent Assay , Glioma/genetics , Glioma/immunology , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/immunology , Humans , Immunohistochemistry , Mice , RNA, Small Interfering , Reverse Transcriptase Polymerase Chain Reaction , Tumor Escape/immunology
13.
Neuro Oncol ; 11(6): 747-56, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19221171

ABSTRACT

Cilengitide is a cyclic peptide antagonist of integrins alphavbeta3 and alphavbeta5 that is currently being evaluated as a novel therapeutic agent for recurrent and newly diagnosed glioblastoma. Its mode of action is thought to be mainly antiangiogenic but may include direct effects on tumor cells, notably on attachment, migration, invasion, and viability. In this study we found that, at clinically relevant concentrations, cilengitide (1-100 microM) induces detachment in some but not all glioma cell lines, while the effect on cell viability is modest. Detachment induced by cilengitide could not be predicted by the level of expression of the cilengitide target molecules, alphavbeta3 and alphavbeta5, at the cell surface. Glioma cell death induced by cilengitide was associated with the generation of caspase activity, but caspase activity was not required for cell death since ectopic expression of cytokine response modifier (crm)-A or coexposure to the broad-spectrum caspase inhibitor zVAD-fmk was not protective. Moreover, forced expression of the antiapoptotic protein marker Bcl-X(L) or altering the p53 status did not modulate cilengitide-induced cell death. No consistent effects of cilengitide on glioma cell migration or invasiveness were observed in vitro. Preliminary clinical results indicate a preferential benefit from cilengitide added to temozolomide-based radiochemotherapy in patients with O(6)-methylguanine DNA methyltransferase (MGMT) gene promoter methylation. Accordingly, we also examined whether the MGMT status determines glioma cell responses to cilengitide alone or in combination with temozolomide. Neither ectopic expression of MGMT in MGMT-negative cells nor silencing the MGMT gene in MGMT-positive cells altered glioma cell responses to cilengitide alone or to cilengitide in combination with temozolomide. These data suggest that the beneficial clinical effects derived from cilengitide in vivo may arise from altered perfusion, which promotes temozolomide delivery to glioma cells.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/pathology , Dacarbazine/analogs & derivatives , Glioma/pathology , Snake Venoms/pharmacology , Animals , Brain Neoplasms/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Methylation , Dacarbazine/pharmacology , Drug Therapy, Combination , Gene Silencing , Humans , Immunoblotting , In Vitro Techniques , Integrin alphaVbeta3/antagonists & inhibitors , Mice , NIH 3T3 Cells , Neoplasm Invasiveness , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , O(6)-Methylguanine-DNA Methyltransferase/genetics , Polymerase Chain Reaction , Promoter Regions, Genetic , Receptors, Vitronectin/antagonists & inhibitors , Temozolomide , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL