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.
Int J Radiat Biol ; 79(7): 561-8, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14530165

ABSTRACT

PURPOSE: We explored and quantified the therapeutic potential of using dominant-negative EGFR transduction with replication-incompetent adenovirus (Ad-EGFR-CD533 or Ad-CD533) as a genetic approach for radiosensitization in different carcinoma and malignant glioma cell lines in vitro and in established tumour xenografts in vivo. MATERIAL AND METHODS: The cell lines MDA-MB-231, A-431, U-373 MG, U-87 MG and T47D were used. The ErbB expression profiles were quantified by Western blotting. MAPK immune complex assay measured MAPK activity with or without EGFR-CD533 expression after ionizing radiation. Radiosensitization was determined and quantified in vitro by colony-formation assays, in vivo by use of an ex vivo-in vitro colony-formation assay after intratumoral infusion of the adenoviral vectors expressing EGFR-CD533 or the control LacZ. RESULTS: Western blotting demonstrated widely varied expression levels of the ErbB receptors in the tested cell lines. Expression of EGFR-CD533 effectively blocked the radiation-induced activation of MAPK, leading to significant radiosensitization in vitro and in vivo. CONCLUSIONS: The radiation-induced ErbB activation can be effectively modulated by a gene therapeutic approach of over-expressing EGFR-CD533 leading to tumour cell radiosensitization after single and repeated radiation exposures both in vitro and in vivo.


Subject(s)
ErbB Receptors/genetics , Genetic Therapy , Neoplasms/radiotherapy , Radiation Tolerance , Adenoviridae/genetics , Cell Line, Tumor , Enzyme Activation , ErbB Receptors/analysis , ErbB Receptors/antagonists & inhibitors , Humans , Mitogen-Activated Protein Kinases/metabolism , Neoplasms/chemistry , Receptor, ErbB-2/analysis , Receptor, ErbB-3/analysis , Receptor, ErbB-4
2.
Int J Radiat Oncol Biol Phys ; 51(3): 775-84, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11697324

ABSTRACT

PURPOSE: Epidermal growth factor receptor (EGFR) and other members of the ErbB family of receptor tyrosine kinases (RTK) mediate autocrine growth regulation in a wide spectrum of human tumor cells. We have previously demonstrated that in stably transfected mammary carcinoma cells a dominant negative (DN) mutant of EGFR, EGFR-CD533 is a potent inhibitor of EGFR and its cytoprotective signaling after exposure to ionizing radiation. In the present study, we further investigate the capacity of a genetic approach, using replication-incompetent adenovirus (Ad)-mediated transfer of EGFR-CD533 (Ad-EGFR-CD533), to enhance the radiosensitivity in vitro of four cell lines representative of three major cancer phenotypes. METHODS AND MATERIALS: The cell lines MDA-MB-231 and T-47D mammary carcinoma, A-431 squamous carcinoma, and U-373 MG malignant glioma cells were used. The ErbB expression profiles and the EGFR tyrosine phosphorylation (Tyr-P) levels following irradiation were quantified by Western blotting. The relative radiosensitivities of tumor cells were assessed by standard colony formation assays after infection with control vector (Ad-LacZ) or Ad-EGFR-CD533. RESULTS: The expression profiles demonstrated varying levels of EGFR, ErbB2, ErbB3, and ErbB4 expression. The overexpression of EGFR-CD533 after infection with Ad-EGFR-CD533 completely inhibited the radiation-induced stimulation of EGFR Tyr-P relative to the immediate 2.4- to 3.1-fold increases in EGFR Tyr-P in control infected cells (Ad-LacZ). Ad-EGFR-CD533-infected cells demonstrated significant (p < 0.001) radiosensitization over a range of radiation doses (1-8 Gy), yielding dose-enhancement ratios (DER) between 1.4 and 1.7. This radiosensitization was maintained under conditions of repeated radiation exposures, using 3 x 2 Gy, yielding DERs of 1.6 and 1.7 for MDA-MB-231 and U-373 cells, respectively. CONCLUSIONS: Overexpression of EGFR-CD533 significantly sensitizes human carcinoma and glioma cells to single and repeated radiation exposures irrespective of their ErbB expression levels. Therefore, transduction of human tumor cells with EGFR-CD533 holds promise as a gene therapeutic approach for the radiosensitization of neoplastic cells that are growth-regulated by EGFR or other ErbB receptors.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Squamous Cell/metabolism , ErbB Receptors/metabolism , Genetic Therapy/methods , Glioma/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Adenoviridae/genetics , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/therapy , ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Genes, Dominant , Glioma/genetics , Glioma/therapy , Humans , Phosphorylation , Radiation Tolerance , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics , Receptor, ErbB-4 , Tumor Cells, Cultured/radiation effects , Tumor Stem Cell Assay
3.
J Natl Cancer Inst ; 93(12): 921-9, 2001 Jun 20.
Article in English | MEDLINE | ID: mdl-11416113

ABSTRACT

BACKGROUND: Exposure of human cancer cells to ionizing radiation activates the epidermal growth factor receptor (EGFR), which, in turn, mediates a cytoprotective response that reduces the cells' sensitivity to ionizing radiation. Overexpression of a dominant-negative EGFR mutant, EGFR-CD533, disrupts the cytoprotective response by preventing radiation-induced activation of the receptor and its downstream effectors. To investigate whether gene therapy with EGFR-CD533 has the potential to increase tumor cell radiosensitivity, we introduced an adenoviral vector containing EGFR-CD533 into xenograft tumors in nude mice and evaluated the tumor response to ionizing radiation. METHODS: Xenograft tumors established from the human mammary carcinoma cell line MDA-MB-231 were transduced via infusion with the adenoviral vector Ad-EGFR-CD533 or a control vector containing the beta-galactosidase gene, Ad-LacZ. The transduced tumors were then exposed to radiation in the therapeutic dose range, and radiation-induced EGFR activation was assessed by examining the tyrosine phosphorylation of immunoprecipitated EGFR. Radiosensitization was determined in vitro by colony-formation assays. All statistical tests were two-sided. RESULTS: The transduction efficiency of MDA-MB-231 tumors by Ad-LacZ was 44%. Expression of EGFR-CD533 in tumors reduced radiation-induced EGFR activation by 2.94-fold (95% confidence interval [CI] = 2.23 to 4.14). The radiosensitivity of Ad-EGFR-CD533-transduced tumors was statistically significantly higher (46%; P<.001) than that of Ad-LacZ-transduced tumors, yielding a dose-enhancement ratio of 1.85 (95% CI = 1.54 to 2.51). CONCLUSIONS: Transduction of MDA-MB-231 xenograft tumors with Ad-EGFR-CD533 conferred a dominant-negative EGFR phenotype and induced tumor radiosensitization. Therefore, disruption of EGFR function through overexpression of EGFR-CD533 may hold promise as a gene therapeutic approach to enhance the sensitivity of tumor cells to ionizing radiation.


Subject(s)
Breast Neoplasms/therapy , ErbB Receptors/physiology , Genetic Therapy , Animals , Breast Neoplasms/pathology , Breast Neoplasms/radiotherapy , Cell Survival/drug effects , Cell Survival/radiation effects , Dose-Response Relationship, Radiation , Doxycycline/toxicity , ErbB Receptors/genetics , ErbB Receptors/radiation effects , Female , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Mice , Mice, Nude , Radiation Tolerance , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Stem Cell Assay
4.
Clin Cancer Res ; 7(3): 682-90, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11297265

ABSTRACT

The epidermal growth factor receptor (EGFR) plays an important role in neoplastic growth control of malignant gliomas. We have demonstrated that radiation activates EGFR Tyr-phosphorylation (EGFR Tyr-P) and the proliferation of surviving human carcinoma cells, a likely mechanism of accelerated cellular repopulation, a major cytoprotective response after radiation. We now investigate the importance of radiation-induced activation of EGFR on the radiosensitivity of the human malignant glioma cells U-87 MG and U-373 MG. The function of EGFR was inhibited through a genetic approach of transducing cells with an Adenovirus (Ad) vector containing dominant-negative (DN) EGFR-CD533 (Ad-EGFR-CD533) at efficiencies of 85-90%. The resulting cells are referred to as U-87-EGFR-CD533 and U-373-EGFR-CD533. After irradiation at 2 Gy, both of the cell lines exhibited a mean 3-fold increase in EGFR Tyr-P. The expression of EGFR-CD533 completely inhibited the radiation-induced activation of EGFR. In clonogenic survival assays after a single radiation exposure, the radiation dose for a survival of 37% (D37) for U-87-EGFR-CD533 cells was 1.4- to 1.5-fold lower, relative to cells transduced with AdLacZ or untransduced U-87 MG cells. This effect was amplified with repeated radiation exposures (3 x 2 Gy) yielding a D37 ratio of 1.8-2.0. In clonogenic survival studies with U-373 MG cells, the radiosensitizing effect of EGFR-CD533 was similar. Furthermore, in vivo studies with U-87 MG xenografts confirmed the effect of EGFR-CD533 on tumor radiosensitization (dose enhancement ratio, 1.8). We conclude that inhibition of EGFR function via Ad-mediated gene transfer of EGFR-CD533 results in significant radiosensitization. As underlying mechanism, we suggest the disruption of a major cytoprotective response involving EGFR and its downstream effectors, such as mitogen-activated protein kinase. The experiments demonstrate for the first time that radiosensitization of malignant glioma cells through disruption of EGFR function may be achieved by genetic therapy approaches.


Subject(s)
Brain Neoplasms/radiotherapy , ErbB Receptors/genetics , Glioma/radiotherapy , Radiation Tolerance , Adenoviridae/genetics , Animals , Blotting, Western , Cell Division , Cell Survival , Dose-Response Relationship, Drug , Dose-Response Relationship, Radiation , Female , Genes, Dominant , Genetic Therapy , Humans , Mice , Mice, Nude , Phosphorylation , Transduction, Genetic , Tumor Cells, Cultured
5.
Oncogene ; 18(33): 4756-66, 1999 Aug 19.
Article in English | MEDLINE | ID: mdl-10467423

ABSTRACT

Exposure of MDA-MB-231 human mammary carcinoma cells to an ionizing radiation dose of 2 Gy results in immediate activation and Tyr phosphorylation of the epidermal growth factor receptor (EGFR). Doxycycline induced expression of a dominant negative EGFR-CD533 mutant, lacking the COOH-terminal 533 amino acids, in MDA-TR15-EGFR-CD533 cells was used to characterize intracellular signaling responses following irradiation. Within 10 min, radiation exposure caused an immediate, transient activation of mitogen activated protein kinase (MAPK) which was completely blocked by expression of EGFR-CD533. The same radiation treatment also induced an immediate activation of the c-Jun-NH2-terminal kinase 1 (JNK1) pathway that was followed by an extended rise in kinase activity after 30 min. Expression of EGFR-CD533 did not block the immediate JNK1 response but completely inhibited the later activation. Treatment of MDA-TR15-EGFR-CD533 cells with the MEK1/2 inhibitor, PD98059, resulted in approximately 70% inhibition of radiation-induced MAPK activity, and potentiated the radiation-induced increase of immediate JNK1 activation twofold. Inhibition of Ras farnesylation with a concomitant inhibition of Ras function completely blocked radiation-induced MAPK and JNK1 activation. Modulation of EGFR and MAPK functions also altered overall cellular responses of growth and apoptosis. Induction of EGFR-CD533 or treatment with PD98059 caused a 3-5-fold increase in radiation toxicity in a novel repeated radiation exposure growth assay by interfering with cell proliferation and potentiating apoptosis. In summary, this data demonstrates that both MAPK and JNK1 activation in response to radiation occur through EGFR-dependent and -independent mechanisms, and are mediated by signaling through Ras. Furthermore, we have demonstrated that radiation-induced activation of EGFR results in downstream activation of MAPK which may affect the radiosensitivity of carcinoma cells.


Subject(s)
Breast Neoplasms/radiotherapy , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Carcinoma/radiotherapy , ErbB Receptors/genetics , Mitogen-Activated Protein Kinases , Alkyl and Aryl Transferases/antagonists & inhibitors , Apoptosis/radiation effects , Cell Division , Enzyme Activation , Farnesyltranstransferase , Female , Flavonoids/pharmacology , Humans , JNK Mitogen-Activated Protein Kinases , Models, Biological , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism , Radiation, Ionizing , Signal Transduction , Tumor Cells, Cultured , ras Proteins/metabolism
6.
Clin Cancer Res ; 5(2): 405-11, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10037190

ABSTRACT

Ionizing radiation activates the epidermal growth factor receptor (EGFR) and downstream signaling involving the cytoprotective mitogen-activated protein kinase (MAPK) pathway. In our effort to investigate the role of EGFR in cellular responses to radiation, we generated mammary carcinoma cell clones, MCF-TR5-EGFR-CD533 and MDA-TR15-EGFR-CD533, that inducibly express EGFR-CD533, a truncated EGFR mutant lacking mitogenic and transformation activity. EGFR-CD533 expression inhibits radiation- and EGF-induced EGFR autophosphorylation and MAPK activation and, therefore, functions as a dominant-negative mutant without blocking the expression of EGFR or erbB-2, another member of the erbB receptor Tyr kinase family. Expression of EGFR-CD533 only minimally inhibited cell growth and did not alter radiosensitivity to single radiation exposures. However, repeated 2 Gy radiation exposures of cells, under conditions of EGFR-CD533 expression, essentially abolished their ability for subsequent cell growth. These results identify the inhibition of EGFR function through genetic manipulation as a potential therapeutic maneuver. The concept of such an intervention would be the radiosensitization of cells by counteracting a radiation-induced cytoprotective proliferation response.


Subject(s)
ErbB Receptors/biosynthesis , Radiation Tolerance , Breast Neoplasms , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Division/drug effects , Cell Division/radiation effects , Dose-Response Relationship, Drug , Doxycycline/pharmacology , ErbB Receptors/genetics , Female , Gene Deletion , Humans , Kinetics , RNA, Messenger/biosynthesis , Tumor Cells, Cultured
8.
Oncogene ; 15(10): 1191-7, 1997 Sep 04.
Article in English | MEDLINE | ID: mdl-9294612

ABSTRACT

Accelerated cellular repopulation has been described as a response of tumors to fractionated irradiation in both normal tissue and tumor systems. To identify the mechanisms by which cells enhance their proliferative rate in response to clinically used doses of ionizing radiation (IR) we have studied human mammary and squamous carcinoma cells which are autocrine growth regulated by the epidermal growth factor receptor (EGFR) and its ligands, transforming growth factor-alpha and EGF. Both EGF and IR induced EGFR autophosphorylation, comparable levels of phospholipase C gamma activation as measured by inositol-1,4,5-triphosphate production, and as a consequence oscillations in cytosolic [Ca2+]. Activities of Raf-1 and mitogen-activated protein kinase (MAPK) were also stimulated by EGF and IR by Ca(2+)-dependent mechanisms. All these responses to EGF and IR were dependent upon activation of EGFR as judged by the use of the specific inhibitor of EGFR autophosphorylation, tyrphostin AG1478. Importantly, IR-induced proliferation of A431 cells was also inhibited by AG1478. This is the first report which demonstrates a link between IR-induced activation of proliferative signal transduction pathways and enhanced proliferation. We propose that accelerated repopulation of tumors whose growth is regulated by EGFR is initiated by an IR-induced EGFR activation mechanism that mimics the effects of growth factors.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Division/drug effects , ErbB Receptors/metabolism , Tyrphostins , Breast Neoplasms/pathology , Calcium/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Enzyme Activation/radiation effects , Enzyme Inhibitors/pharmacology , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Isoenzymes/metabolism , Nitriles/pharmacology , Phospholipase C gamma , Phosphotyrosine/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-raf , Quinazolines/pharmacology , Radiation, Ionizing , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Signal Transduction , Tumor Cells, Cultured , Type C Phospholipases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL