Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Cancer Res ; 66(16): 7864-9, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16912159

ABSTRACT

The epidermal growth factor receptor (EGFR) is commonly amplified, overexpressed, and mutated in glioblastoma, making it a compelling molecular target for therapy. We have recently shown that coexpression of EGFRvIII and PTEN protein by glioblastoma cells is strongly associated with clinical response to EGFR kinase inhibitor therapy. PTEN loss, by dissociating inhibition of the EGFR from downstream phosphatidylinositol 3-kinase (PI3K) pathway inhibition, seems to act as a resistance factor. Because 40% to 50% of glioblastomas are PTEN deficient, a critical challenge is to identify strategies that promote responsiveness to EGFR kinase inhibitors in patients whose tumors lack PTEN. Here, we show that the mammalian target of rapamycin (mTOR) inhibitor rapamycin enhances the sensitivity of PTEN-deficient tumor cells to the EGFR kinase inhibitor erlotinib. In two isogenic model systems (U87MG glioblastoma cells expressing EGFR, EGFRvIII, and PTEN in relevant combinations, and SF295 glioblastoma cells in which PTEN protein expression has been stably restored), we show that combined EGFR/mTOR kinase inhibition inhibits tumor cell growth and has an additive effect on inhibiting downstream PI3K pathway signaling. We also show that combination therapy provides added benefit in promoting cell death in PTEN-deficient tumor cells. These studies provide strong rationale for combined mTOR/EGFR kinase inhibitor therapy in glioblastoma patients, particularly those with PTEN-deficient tumors.


Subject(s)
ErbB Receptors/genetics , Glioblastoma/pathology , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Protein Kinase Inhibitors/pharmacology , Protein Kinases/physiology , Cell Division/drug effects , Cell Line, Tumor , Enzyme Activation , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Glioblastoma/genetics , Humans , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Quinazolines/pharmacology , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Transfection
2.
Cancer Res ; 66(1): 159-67, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16397228

ABSTRACT

Glioblastomas are invasive and aggressive tumors of the brain, generally considered to arise from glial cells. A subset of these cancers develops from lower-grade gliomas and can thus be clinically classified as "secondary," whereas some glioblastomas occur with no prior evidence of a lower-grade tumor and can be clinically classified as "primary." Substantial genetic differences between these groups of glioblastomas have been identified previously. We used large-scale expression analyses to identify glioblastoma-associated genes (GAG) that are associated with a more malignant phenotype via comparison with lower-grade astrocytomas. We have further defined gene expression differences that distinguish primary and secondary glioblastomas. GAGs distinct to primary or secondary tumors provided information on the heterogeneous properties and apparently distinct oncogenic mechanisms of these tumors. Secondary GAGs primarily include mitotic cell cycle components, suggesting the loss of function in prominent cell cycle regulators, whereas primary GAGs highlight genes typical of a stromal response, suggesting the importance of extracellular signaling. Immunohistochemical staining of glioblastoma tissue arrays confirmed expression differences. These data highlight that the development of gene pathway-targeted therapies may need to be specifically tailored to each subtype of glioblastoma.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Glioblastoma/secondary , Adipokines , Apoptosis/genetics , Astrocytoma/genetics , Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Cycle/genetics , Cell Growth Processes/genetics , Chitinase-3-Like Protein 1 , Gene Expression Profiling , Glioblastoma/metabolism , Glioblastoma/pathology , Glycoproteins/biosynthesis , Glycoproteins/genetics , Humans , Immunohistochemistry , Lectins , Mesoderm/pathology , Stromal Cells/pathology , Transcription, Genetic , Up-Regulation
3.
N Engl J Med ; 353(19): 2012-24, 2005 Nov 10.
Article in English | MEDLINE | ID: mdl-16282176

ABSTRACT

BACKGROUND: The epidermal growth factor receptor (EGFR) is frequently amplified, overexpressed, or mutated in glioblastomas, but only 10 to 20 percent of patients have a response to EGFR kinase inhibitors. The mechanism of responsiveness of glioblastomas to these inhibitors is unknown. METHODS: We sequenced kinase domains in the EGFR and human EGFR type 2 (Her2/neu) genes and analyzed the expression of EGFR, EGFR deletion mutant variant III (EGFRvIII), and the tumor-suppressor protein PTEN in recurrent malignant gliomas from patients who had received EGFR kinase inhibitors. We determined the molecular correlates of clinical response, validated them in an independent data set, and identified effects of the molecular abnormalities in vitro. RESULTS: Of 49 patients with recurrent malignant glioma who were treated with EGFR kinase inhibitors, 9 had tumor shrinkage of at least 25 percent. Pretreatment tissue was available for molecular analysis from 26 patients, 7 of whom had had a response and 19 of whom had rapid progression during therapy. No mutations in EGFR or Her2/neu kinase domains were detected in the tumors. Coexpression of EGFRvIII and PTEN was significantly associated with a clinical response (P<0.001; odds ratio, 51; 95 percent confidence interval, 4 to 669). These findings were validated in 33 patients who received similar treatment for glioblastoma at a different institution (P=0.001; odds ratio, 40; 95 percent confidence interval, 3 to 468). In vitro, coexpression of EGFRvIII and PTEN sensitized glioblastoma cells to erlotinib. CONCLUSIONS: Coexpression of EGFRvIII and PTEN by glioblastoma cells is associated with responsiveness to EGFR kinase inhibitors.


Subject(s)
ErbB Receptors/genetics , Glioblastoma/genetics , PTEN Phosphohydrolase/metabolism , Protein Kinase Inhibitors/therapeutic use , Adult , Aged , Antineoplastic Agents/therapeutic use , DNA, Neoplasm/analysis , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Female , Gefitinib , Gene Amplification , Gene Deletion , Gene Expression , Genes, erbB-1 , Genes, erbB-2 , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Male , Middle Aged , Mutation , Oligodendroglioma/drug therapy , Oligodendroglioma/genetics , Oligodendroglioma/metabolism , PTEN Phosphohydrolase/genetics , Polymerase Chain Reaction , Quinazolines/therapeutic use , Sequence Analysis, DNA , Signal Transduction
4.
J Biol Chem ; 280(29): 26953-64, 2005 Jul 22.
Article in English | MEDLINE | ID: mdl-15908427

ABSTRACT

Glioblastoma is the most common malignant brain tumor of adults and one of the most lethal cancers. The secreted growth factor pleiotrophin (PTN) promotes glioblastoma migration and proliferation, initiating its oncogenic activities through two cell surface receptors, the protein tyrosine phosphatase receptor zeta (PTPRZ1) and the anaplastic lymphoma kinase (ALK), respectively. Here, we report on the presence and purification of two naturally occurring forms of PTN (18 and 15 kDa) that differentially promote glioblastoma migration and proliferation. Using a panel of glioblastoma cell lines, including low passage patient-derived cultures, we demonstrate that PTN15 promotes glioblastoma proliferation in an ALK-dependent fashion, whereas immobilized PTN18 promotes haptotactic migration of glioblastoma cells in a PTPRZ1-dependent fashion. Mass spectrometric analysis indicated that PTN15 differs from PTN18 by processing of 12 C-terminal amino acids. To demonstrate clinical relevance, we show that PTN15, PTN18, and PTPRZ1 are significantly overexpressed in glioblastoma relative to normal brain at both mRNA and protein levels using microarray, Western blot, and tissue microarray analyses on human tumors. These results indicate that the PTN18-PTPRZ1 and the PTN15-ALK signaling pathways represent potentially important therapeutic targets for glioblastoma invasion and growth.


Subject(s)
Carrier Proteins/physiology , Cytokines/physiology , Glioblastoma/pathology , Adult , Anaplastic Lymphoma Kinase , Carrier Proteins/analysis , Carrier Proteins/metabolism , Cell Movement , Cell Proliferation , Cytokines/analysis , Cytokines/metabolism , Glioblastoma/chemistry , Humans , Neoplasm Invasiveness/pathology , Neoplasm Proteins/analysis , Nerve Growth Factors/physiology , Protein Processing, Post-Translational , Protein Tyrosine Phosphatases/analysis , Protein Tyrosine Phosphatases/physiology , Protein-Tyrosine Kinases , RNA, Neoplasm/analysis , Receptor Protein-Tyrosine Kinases , Receptor-Like Protein Tyrosine Phosphatases, Class 5 , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...