Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Gene Ther ; 12(13): 1042-8, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15789059

ABSTRACT

Targeted therapies directed to tumor-associated antigens are being investigated for the treatment of cancer. However, there are few suitable animal models for testing the ability to target these tumor markers. Therefore, we have exploited mice transgenic for the human coxsackie and adenovirus receptor (hCAR) to establish a new model for transient expression of human tumor-associated antigens in the pulmonary vasculature. Systemic administration of Ad in hCAR mice resulted in an increase in transgene expression in the lungs compared to wild-type mice, as determined using a luciferase reporter gene. To reduce transgene expression in the liver, the predominant organ of ectopic Ad localization and transgene expression following systemic administration, we utilized the endothelial-specific flt-1 promoter, which resulted in a further increased lung-to-liver ratio of luciferase expression. Administration of an adenoviral vector encoding the tumor-associated antigen carcinoembryonic antigen (CEA) under transcriptional control of the flt-1 promoter resulted in selective expression of this antigen in the pulmonary vasculature of hCAR mice. Feasibility of targeting to expressed CEA was subsequently demonstrated using adenoviral vectors preincubated with a bifunctional adapter molecule recognizing this tumor-associated antigen, thus demonstrating utility of this transient transgenic animal model.


Subject(s)
Adenoviridae/genetics , Carcinoembryonic Antigen/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Pulmonary Circulation/physiology , Animals , Extracellular Matrix Proteins , Gene Expression , Gene Targeting/methods , Genetic Vectors/genetics , Immunohistochemistry/methods , Liver/metabolism , Luciferases/genetics , Mice , Mice, Transgenic , Myosin Heavy Chains , Nonmuscle Myosin Type IIB , Promoter Regions, Genetic , Proteins/genetics , Receptors, Virus/genetics , Vascular Endothelial Growth Factor Receptor-1
2.
Gene Ther ; 11(7): 645-8, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15029227

ABSTRACT

Adenoviral vectors are considered to be good gene delivery vectors for cancer gene therapy due to their wide host tissue range and cell cycle-independent infectivity. However, the disadvantages include the lack of specificity for cancer cells and the high liver accumulation in vivo. The human CXCR4 gene is expressed at high levels in many types of cancers, but is repressed in the liver. We explored the CXCR4 promoter as a candidate to restrict adenoviral transgene expression to tumor cells with a low expression in host tissues. The luciferase activities in multiple cancer cell lines infected with recombinant adenovirus reAdGL3BCXCR4 or the control vector reAdGL3BCMV revealed that the CXCR4 promoter exhibited relatively high transcriptional activity in a breast cancer cell line, MDA-MB-361, and two ovarian cancer cell lines, OVCAR-3 and SKOV3. ip1, 65% (P=0.0087), 16.7% (P=0.1) and 20% (P=0.0079) compared to that of the CMV promoter, respectively, and low expression, 4.9 and 0.1%, respectively, in both normal cell lines HFBC and HMEC. In addition, CXCR4 had a low expression of luciferase (0.32%) compared to that of the CMV promoter in mouse liver in vivo. The data also revealed that the CXCR4 promoter was a stronger tumor-specific promoter (TSP) than the Cox-2M promoter in primary melanomas obtained from two patients. The CXCR4 promoter is shown to have a 'tumor-on' and 'liver-off' status in vitro and in vivo, and CXCR4 may prove to be a good candidate TSP for cancer gene therapy approaches for melanoma and breast cancers.


Subject(s)
Adenoviridae/genetics , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Neoplasms/therapy , Receptors, CXCR4/genetics , Transcription, Genetic , Animals , Breast Neoplasms/therapy , Cell Line, Tumor , Female , Gene Expression , Humans , Liver/metabolism , Melanoma/therapy , Mice , Ovarian Neoplasms/therapy , Promoter Regions, Genetic , Skin Neoplasms/therapy
3.
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
4.
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
5.
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
6.
Clin Cancer Res ; 7(7): 2057-66, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11448924

ABSTRACT

PURPOSE: We hypothesized that adenovirus-mediated soluble fms-like tyrosine kinase receptor (sFLT-1) gene therapy can inhibit the ovarian tumor growth and increase survival of mice in the context of ovarian carcinoma. EXPERIMENTAL DESIGN: We constructed an infectivity-enhanced recombinant adenovirus (AdRGDGFPsFLT-1) expressing soluble FLT-1 and green fluorescent protein (GFP). An adenovirus AdRGDGFP expressing GFP alone was used as control. The functional validation of adenovirus-mediated sFLT-1 was determined by an in vitro human umbilical vein endothelial cell proliferation inhibition assay. To evaluate the therapeutic potential of adenovirus-expressed sFLT-1 to inhibit the growth of ovarian tumors and to increase the survival duration of mice with ovarian tumors, two tumor models were used. First, SKOV3.ip1 ovarian carcinoma cells were infected ex vivo with either AdRGDGFPsFLT-1 or AdRGDGFP or uninfected and then inoculated s.c. into BALB/c nude mice, and tumor growth was monitored. Second, SKOV3.ip1 cells were inoculated i.p. into CB17 SCID mice and then treated with two doses of either AdRGDGFPsFLT-1 or AdRGDGFP or with PBS on days 1 and 14 after inoculation of cells, and the survival duration was monitored. RESULTS: Treatment with adenovirus-expressed sFLT-1 significantly inhibited the proliferation of human umbilical vein endothelial cells. The s.c. tumor nodules in mice derived from cells infected with AdRGDGFPsFLT-1 were significantly smaller than those infected with either AdRGDGFP or uninfected. In addition, i.p. administration of the AdRGDGFPsFLT-1 resulted in a significant increase in the survival times of mice compared with AdRGDGFP- or PBS-treated mice. CONCLUSIONS: Our results suggest that adenovirus-mediated sFLT-1 gene therapy can effectively inhibit ovarian tumor growth and increase survival in a murine model of ovarian carcinoma.


Subject(s)
Genetic Therapy , Ovarian Neoplasms/therapy , Proto-Oncogene Proteins/genetics , Receptor Protein-Tyrosine Kinases/genetics , Adenoviridae/genetics , Animals , Cell Division/genetics , Cell Line , Cloning, Molecular , DNA, Complementary/genetics , Female , Genetic Vectors/genetics , Green Fluorescent Proteins , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, SCID , Neoplasm Transplantation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Solubility , Survival Analysis , Transplantation, Heterologous , Tumor Cells, Cultured , Vascular Endothelial Growth Factor Receptor-1
7.
Infect Immun ; 65(1): 211-8, 1997 Jan.
Article in English | MEDLINE | ID: mdl-8975914

ABSTRACT

This study was conducted to partially characterize and identify the purity of two major outer membrane proteins (OMPs) (with molecular weights of 32,000 and 35,000 [32K and 35K, respectively]) of Pasteurella haemolytica. The 35K and 32K major OMPs, designated Pasteurella outer membrane proteins A and B (PomA and PomB, respectively), were extracted from P. haemolytica by solubilization in N-octyl polyoxyl ethylene. The P. haemolytica strain used was a mutant serotype A1 from which the genes expressing the 30-kDa lipoproteins had been deleted. PomA and PomB were separated and partially purified by anion-exchange chromatography. PomA but not PomB was heat modifiable. The N-terminal amino acid sequences of the two proteins were determined and compared with reported sequences of other known proteins. PomA had significant N-terminal sequence homology with the OmpA protein of Escherichia coli and related proteins from other gram-negative bacteria. Moreover, polyclonal antiserum raised against the E. coli OmpA protein reacted with this protein. PomA was surface exposed, was conserved among P. haemolytica biotype A serotypes, and had porin activity in planar bilayers. No homology between the N-terminal amino acid sequence of PomB and those of other known bacterial proteins was found. Cattle vaccinated with live P. haemolytica developed a significant increase in serum antibodies to partially purified PomA, as shown by enzyme-linked immunosorbent assays, and to purified PomA and PomB, as detected on Western blots and by densitometry.


Subject(s)
Bacterial Outer Membrane Proteins/chemistry , Lipoproteins/chemistry , Mannheimia haemolytica/chemistry , Amino Acid Sequence , Animals , Antibodies, Bacterial/biosynthesis , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/immunology , Cattle , Detergents , Lipid Bilayers , Lipoproteins/immunology , Lung/pathology , Mannheimia haemolytica/classification , Mannheimia haemolytica/immunology , Molecular Sequence Data , Pasteurella Infections/immunology , Pasteurella Infections/pathology , Polyethylene Glycols , Sequence Analysis , Sequence Homology, Amino Acid , Serotyping , Solubility
SELECTION OF CITATIONS
SEARCH DETAIL
...