Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Gene Ther ; 9(4): 256-62, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11896464

ABSTRACT

Single chain antibodies (scFv) represent powerful interventional agents for the achievement of targeted therapeutics. The practical utility of these agents have been limited, however, by difficulties related to production of recombinant scFv and the achievement of effective and sustained levels of scFv in situ. To circumvent these limitations, we have developed an approach to express scFv in vivo. An anti-erbB2 scFv was engineered for secretion by eukaryotic cells. The secreted scFv could bind to its target and specifically suppress cell growth of erbB2-positive cells in vitro. Adenoviral vectors expressing the cDNA for the secretory scFv likewise could induce target cells to produce an anti-tumor anti-erbB2 scFv. In vivo gene transfer via the anti-erbB2 scFv encoding adenovirus also showed anti-tumor effects. Thus, by virtue of engineering a secreted version of the anti-tumor anti-erbB-2 scFv, and in vivo expression via adenoviral vector, effective concentrations of scFv were achieved. In vivo gene transfer clearly represents a powerful means to realize effective scFv-based approaches. This method will likely have applicability for a range of disorders amenable to targeted therapeutic approaches.


Subject(s)
Adenoviridae/genetics , Antibodies, Monoclonal/genetics , Genetic Therapy/methods , Genetic Vectors , Receptor, ErbB-2/immunology , Animals , Antibodies, Monoclonal/blood , Female , Gene Targeting/methods , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/prevention & control , Transfection , Tumor Cells, Cultured
2.
Gynecol Oncol ; 83(2): 370-7, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11606099

ABSTRACT

OBJECTIVE: The transfer of tumor suppressor genes has been shown to revert the malignant phenotype. In this regard, bax is a pro-apoptotic molecule that also functions as a tumor suppressor. The purpose of this study was to evaluate bax as a gene therapeutic in the context of cervical cancer. METHODS: Efficiency of viral transduction in cervical cancer cell lines and primary cervical cancer cells was evaluated with an adenoviral vector encoding green fluorescent protein and luciferase, respectively. We generated a recombinant adenoviral vector that encodes the bax gene under inducible conditions. To this end, expression of this pro-apoptotic gene was controlled by a Cre-LoxP system. Following infection with the recombinant bax adenovirus, the viability of cervical cancer cell lines and primary cervical cancer cells was evaluated using crystal violet staining and FACS analysis. Apoptotic cell death was monitored using annexin V staining. RESULTS: High levels of viral infection were observed in all cervical cancer cell lines (>85%) and primary cervical cancer cells. Significant cytotoxicity was seen in all cervical cancer cells lines and, more importantly, patient-derived primary cervical cancer cells. Moreover, bax-mediated cell death occurred via an apoptotic pathway. CONCLUSIONS: Our results indicate that a bax recombinant adenoviral vector causes cell death mediated via an apoptotic pathway in multiple cervical cancer cell lines and primary cervical cancer cells. These data suggest that bax may be a candidate for human gene therapy in the setting of cervical carcinoma.


Subject(s)
Apoptosis/genetics , Genetic Therapy/methods , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/genetics , Uterine Cervical Neoplasms/therapy , Adenoviridae/genetics , Female , Genes, Tumor Suppressor , Genetic Vectors/genetics , HeLa Cells , Humans , Transduction, Genetic , Tumor Cells, Cultured , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , bcl-2-Associated X Protein
3.
Clin Cancer Res ; 7(8): 2496-504, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11489832

ABSTRACT

PURPOSE: The purpose of the study was to determine the capability of the midkine (MK) and cycooxygenase-2 (cox-2) gene promoter regions to function as tumor-specific promoters for use in targeted gene therapy of ovarian cancer. EXPERIMENTAL DESIGN: Established and primary ovarian cancer and mesothelial cells were transduced by adenoviral vectors containing a reporter or thymidine kinase gene expressed under the control of the MK, cox-2, or cytomegalovirus (CMV) promoters. SCID or C57BL/6 mice were injected i.p. with these same vectors. In vitro reporter gene expression and cellular cytotoxicity was determined using luciferase and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays, respectively. Acute toxicity in vivo was assessed by histological evaluation of harvested tissues. RESULTS: Consistent activation of the MK and cox-2 promoters was noted in all of the ovarian cancer cell lines in addition to primary ovarian cancer cells. In contrast, reduced reporter activity was reported in mesothelial cells transduced with adenoviruses containing the test promoters, which was especially apparent for the cox-2 promoter. Additionally, the cox-2 promoter exhibited significantly lower reporter gene levels in liver and peritoneum than the control promoter in in vivo experiments. Tumor-cell killing induced by Adcox-2 MTK was comparable to that observed with AdCMVTK. However, a clear differential toxicity pattern was observed in favor of animals treated with Adcox-2 MTK when compared with controls. CONCLUSIONS: These data clearly demonstrate that the transcriptional control afforded by the cox-2 promoter is tumor-specific and is able to mitigate associated toxicity in normal tissue while maintaining therapeutic efficacy in the context of an ovarian cancer molecular chemotherapeutic approach.


Subject(s)
Cytokines , Genetic Therapy/methods , Ovarian Neoplasms/therapy , Transgenes/genetics , Animals , Carrier Proteins/genetics , Cell Line , Cell Survival/drug effects , Cell Survival/genetics , Cyclooxygenase 2 , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Ganciclovir/therapeutic use , Gene Expression , Gene Transfer Techniques , Hepatitis/etiology , Hepatitis/genetics , Hepatitis/pathology , Herpesvirus 1, Human/genetics , Humans , Isoenzymes/genetics , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, SCID , Midkine , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Peritonitis/etiology , Peritonitis/genetics , Peritonitis/pathology , Plasmids/administration & dosage , Plasmids/genetics , Promoter Regions, Genetic/genetics , Prostaglandin-Endoperoxide Synthases/genetics , Thymidine Kinase/genetics , Tumor Cells, Cultured
4.
Clin Cancer Res ; 6(11): 4442-8, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11106265

ABSTRACT

We and others have proposed mammalian cells as gene delivery vehicles with the potential for overcoming physiological barriers to viral vectors. To that end, we previously have shown the potential of CD34+ endothelial progenitors for systemic gene delivery in a primate angiogenesis model. Here we seek to explore the utility of CD34+ cells of human origin as vehicles for toxin genes and, in particular, to measure their capacity to effect a cytotoxic bystander effect in human endothelium and tumor cells. To this end, CD34+ cells were transduced with TOZ.1, a nonreplicative herpes simplex vector encoding thymidine kinase. To test the capacity of CD34+ cells to induce a cytotoxic bystander effect in target cells, we performed mixing experiments, whereby TOZ.1-transduced CD34+ cells were mixed with either human vascular endothelial cells or human ovarian tumor cells (SKOV3.ip1). Cell viability was measured by the MTS assay. Lastly, mixtures of TOZ.1-transduced CD34+ cells and SKOV3.ip1 tumor cells were injected s.c. to evaluate the bystander effect in vivo. After transduction of CD34+ cells with TOZ.1, treatment with ganciclovir induced the killing of 99% of cells. In cell-mixing experiments, a linear correlation was observed between the percentages of TOZ.1-transduced CD34+ cells and total cell killing. For example, when 50% of CD34+ transduced cells were mixed with nontransduced SKOV3.ip1, >70% of all cells died. Similarly, when the same percentage was mixed with human vascular endothelial cells, >80% of the total number of cells died. In vivo studies showed an abrogation of tumor formation when TOZ.1-transduced CD34+ cells and ganciclovir were administered. Our observations establish the feasibility of a method for cell-based toxin gene delivery into disseminated areas of tumor angiogenesis.


Subject(s)
Antigens, CD34/analysis , Endothelium, Vascular/metabolism , Genetic Therapy , Hematopoietic Stem Cells/physiology , Neoplasms/therapy , Animals , Endothelium, Vascular/cytology , Female , Ganciclovir , Humans , Lymphocytes, Tumor-Infiltrating/physiology , Mice , Simplexvirus/genetics , Tumor Cells, Cultured
5.
Mol Ther ; 1(6): 545-54, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10933979

ABSTRACT

Overexpression of proapoptotic Bax favors death in cells resistant to ionizing radiation. We hypothesized that expression of Bax via adenoviral-mediated gene delivery could sensitize radiation-refractory cells to radiotherapy. An inducible Bax recombinant adenovirus (Ad/Bax) had been generated using the Cre/loxp system. Human ovarian cancer cell lines and primary, patient-derived cancer cells from ascites were irradiated and infected with the Ad/Bax and an expression-inducing vector, Ad/Cre. Cell death was evaluated by crystal violet staining, fluorescence-activated cell sorter analysis of Annexin V, and colony formation assay (cell lines only). To further characterize the mechanism of death, cell morphology was examined by nuclear staining with Hoechst 33258. Lastly, to evaluate the capacity of the combined treatment to inhibit tumor growth, mice were injected subcutaneously with ovarian cancer cells exposed to Bax, radiation therapy (RT), or both, and tumor size was measured periodically. Infection of the cancer cell lines and primary cells with both Ad/Bax and Ad/Cre significantly enhanced sensitivity to ionizing radiation, achieving high levels of cell killing in short-term assays. In addition, the combination of Bax and radiotherapy reduced the survival fraction of cell lines 2 logs in standard colony-forming assays. Investigation into the involved mechanism suggests that Bax-mediated radiosensitization occurs through both apoptosis and necrosis pathways. Further, mice subcutaneously injected with ovarian tumor cells previously treated with radiation, or with radiation and irrelevant viruses, consistently developed tumor nodules. In addition, approximately 80% of injections were followed by tumor formation after treatment with Ad/Bax and Ad/Cre alone. In contrast, tumor formation was completely inhibited after combined treatment with Ad/Bax and Ad/Cre and radiation. Augmentation of the effect of radiotherapy on human ovarian cancer cells and primary cancer cells from patients via a recombinant adenovirus encoding Bax is feasible.


Subject(s)
Adenoviridae/genetics , Apoptosis/genetics , Ovarian Neoplasms/therapy , Proto-Oncogene Proteins c-bcl-2 , Proto-Oncogene Proteins/genetics , Viral Proteins , Animals , Combined Modality Therapy , Female , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Humans , Integrases/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Ovarian Neoplasms/pathology , Ovarian Neoplasms/radiotherapy , Radiation Tolerance/genetics , Transplantation, Heterologous , Tumor Cells, Cultured , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...