Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cancer Gene Ther ; 22(9): 445-53, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26251031

ABSTRACT

We examined cytotoxicity of replication-competent type 5 adenoviruses (Ad5) in human pancreatic carcinoma cells with a p53-defective genotype. The replication-competent Ad5 of which E1A gene was activated by exogenous transcriptional regulatory sequences, derived from the midkine and survivin genes, achieved cytotoxicity to the pancreatic carcinoma. These cells were susceptible to replication-incompetent Ad5 expressing the wild-type p53 gene. We also produced the replication-competent Ad5 bearing the same exogenous regulatory sequences and the type 35 Ad-derived fiber-knob region, and showed that the cytotoxicity was comparable to that of the replication-competent Ad5 prototype. We then investigated possible combinatory effects of the fiber-modified replication-competent Ad and Ad5 expressing the wild-type p53 gene, both of which did not interfere respective infections. The combination produced synergistic cytotoxic effects with enhanced cleavages of caspase-3 and PARP molecules, and with increased sub-G1 fractions and annexin V-positive populations although the viral production of the replication-competent Ad was rather suppressed by expressed p53. Pancreatic cells infected with both Ad showed increase of p53 and decrease of MDM2 and p21 levels, compared with those infected with Ad expressing the p53 gene. These data collectively indicated that replication-competent Ad augmented susceptibility of pancreatic cells to apoptosis through upregulated p53 expression.


Subject(s)
Adenocarcinoma/pathology , Adenoviruses, Human/physiology , Inhibitor of Apoptosis Proteins/physiology , Nerve Growth Factors/physiology , Pancreatic Neoplasms/pathology , Tumor Suppressor Protein p53/biosynthesis , Adenovirus E1A Proteins/deficiency , Adenoviruses, Human/genetics , Apoptosis , Capsid Proteins/genetics , Caspase 3/metabolism , Cell Cycle , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cytopathogenic Effect, Viral , Defective Viruses/physiology , Genes, p53 , HEK293 Cells , Humans , Inhibitor of Apoptosis Proteins/genetics , Midkine , Nerve Growth Factors/genetics , Poly(ADP-ribose) Polymerases/metabolism , Proto-Oncogene Proteins c-mdm2/biosynthesis , Proto-Oncogene Proteins c-mdm2/genetics , Survivin , Virus Replication/genetics
2.
Clin Cancer Res ; 14(20): 6505-14, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18927290

ABSTRACT

PURPOSE: Anaplastic thyroid carcinoma is a prime target for innovative therapy because it represents one of the most lethal human neoplasms and is refractory to conventional treatments such as chemotherapy and radiotherapy. We have evaluated a novel therapeutic approach based on the oncolytic replication-selective adenovirus dl922-947. EXPERIMENTAL DESIGN: The antitumor efficacies of the E1ADeltaCR2 (dl922-947) and DeltaE1B55K (dl1520) mutants were compared in human thyroid anaplastic carcinoma cells in culture and in xenografts in vivo. To enhance the effects of dl922-947, anaplastic thyroid carcinoma tumor xenografts were treated with dl922-947 in combination with bevacizumab. RESULTS: We showed that the efficacy of dl922-947 exceeded that of dl1520 in all tested anaplastic thyroid carcinoma cells in vitro and in vivo. Furthermore, bevacizumab in combination with dl922-947 significantly reduced tumor growth compared with single treatments alone. Bevacizumab treatment significantly improved viral distribution in neoplastic tissues. CONCLUSIONS: Our data showed that dl922-947 had a higher oncolytic activity compared with dl1520 in anaplastic thyroid carcinoma cell lines and might represent a better option for virotherapy of anaplastic thyroid carcinoma. Moreover, bevacizumab increased the oncolytic effects of dl922-947 by enhancing viral distribution in tumors. The results described herein encourage the use of the dl922-947 virus in combination with bevacizumab.


Subject(s)
Adenoviridae/drug effects , Angiogenesis Inhibitors/therapeutic use , Antibodies, Monoclonal/therapeutic use , Carcinoma/therapy , Oncolytic Virotherapy , Thyroid Neoplasms/therapy , Adenoviridae/genetics , Adenoviridae Infections/pathology , Adenoviridae Infections/therapy , Adenoviridae Infections/virology , Adenovirus E1A Proteins/deficiency , Animals , Antibodies, Monoclonal, Humanized , Bevacizumab , Carcinoma/pathology , Carcinoma/virology , Cells, Cultured , Combined Modality Therapy , Drug Synergism , Humans , Male , Mice , Mice, Nude , Thyroid Neoplasms/pathology , Thyroid Neoplasms/virology , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/immunology , Vascular Endothelial Growth Factor A/metabolism , Virus Replication/drug effects , Xenograft Model Antitumor Assays
3.
Mol Ther ; 13(5): 928-937, 2006 May.
Article in English | MEDLINE | ID: mdl-16459149

ABSTRACT

DNA-damage checkpoints are activated to arrest cells and promote survival upon genotoxic challenge. Efforts have been taken to target checkpoint kinase 1 (chk1; approved gene symbol CHEK1), a crucial checkpoint modulator, for therapeutic intervention. However, improvement of the potency and specificity of such therapeutics remains a major challenge. This prompted us to develop a novel chk1-targeting strategy by constructing a potent oncolytic adenovirus (M2). M2 was generated by combining two artificial features into a wild-type adenovirus type 5 genome. First, M2 was engineered with a 27-bp deletion in the E1A region to confer tumor-selective replication. Second, an antisense chk1 cDNA was substituted for viral E3 6.7K and gp19K genes. In this design, M2 exploited the adenovirus E3 promoters to express antisense chk1 cDNA in a viral replication-dependent fashion and preferentially silenced the chk1 gene in tumor cells. By virtue of combining oncolysis with chk1 targeting, M2 exhibited potent antitumoral efficacy in vitro and in vivo. Systemic administration of M2, plus a low dose of cisplatin, cured 80% of orthotopic hepatic carcinoma mouse models that were otherwise resistant to cisplatin. These findings have directed us toward the development of novel oncolytic adenoviruses that will be potentially applicable to a wide range of molecular-based therapeutics.


Subject(s)
Adenoviridae/genetics , Adenovirus E1A Proteins/deficiency , Carcinoma, Hepatocellular/therapy , Gene Targeting , Liver Neoplasms/therapy , Protein Kinases/genetics , Adenoviridae/physiology , Adenovirus E1A Proteins/genetics , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/virology , Cell Line , Cell Line, Tumor , Cell Survival , Checkpoint Kinase 1 , Cisplatin/therapeutic use , Cytopathogenic Effect, Viral/physiology , DNA, Antisense/genetics , Female , Genetic Vectors/administration & dosage , Genetic Vectors/therapeutic use , HeLa Cells , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/virology , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Genetic , Promoter Regions, Genetic , Protein Kinases/metabolism , Virus Replication , Xenograft Model Antitumor Assays
4.
Clin Cancer Res ; 11(23): 8431-40, 2005 Dec 01.
Article in English | MEDLINE | ID: mdl-16322306

ABSTRACT

PURPOSE: Polo-like kinase 1 (plk1) is a serine/threonine protein kinase essential for multiple mitotic processes. Previous observations have validated plk1 as a promising therapeutic target. Despite being conceptually attractive, the potency and specificity of current plk1-based therapies remain limited. We sought to develop a novel plk1-targeting strategy by constructing an oncolytic adenovirus to selectively silence plk1 in tumor cells. EXPERIMENTAL DESIGN: Two artificial features were engineered into one wild-type adenovirus type 5 (wt-Adv5) genome to generate a new oncolytic adenovirus (M1). First, M1 contains a 27-bp deletion in E1A region, which confers potent, oncolytic efficacy. Second, M1 is armed with a fragment of antisense plk1 cDNA that substitutes the E3 region encoding 6.7K and gp19K. In this design, tumor-selective replication of M1 would activate the native adenovirus E3 promoters to express the antisense plk1 cDNA preferentially in tumor cells and silence tumor-associated plk1 protein. RESULTS: By virtue of combining oncolysis with plk1 targeting, M1 exhibited potent antitumoral efficacy in vitro and in vivo. Systemic administration of M1 plus cisplatin induced complete tumor regression in 80% of orthotopic hepatic carcinoma model mice that were otherwise resistant to cisplatin and disseminated metastases. CONCLUSIONS: Coupling plk1 targeting with oncolysis had shown superior antitumor efficacy. Present findings would benefit the development of novel oncolytic adenoviruses generally applicable to a wide range of molecule-based therapeutics.


Subject(s)
Adenoviridae/physiology , Adenovirus E1A Proteins/deficiency , Carcinoma, Hepatocellular/therapy , Cell Cycle Proteins/antagonists & inhibitors , Cytopathogenic Effect, Viral/physiology , Liver Neoplasms/therapy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Adenovirus E1A Proteins/genetics , Adenovirus E3 Proteins/genetics , Animals , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/virology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Survival , Cisplatin/therapeutic use , DNA, Antisense/genetics , Female , Genetic Vectors/genetics , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/virology , Mice , Mice, Inbred BALB C , Mice, Nude , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Tumor Cells, Cultured , Virus Replication , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
5.
Neoplasia ; 7(1): 48-56, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15720816

ABSTRACT

Replication-competent oncolytic adenoviruses hold considerable promise for treating malignant gliomas. The toxicity of the clinically tested E1B-55 kDa mutant virus is negligible; however, its full clinical potential is still being evaluated. The purpose of the present study is to compare the antiglioma activity in vitro and in vivo between Delta-24, an E1A mutant adenovirus, and RA55, an E1B-55 kDa mutant adenovirus. We selected human glioma cell lines that were tumorigenic in nude mice and express wild-type p53 (U-87 MG, D54 MG) or mutant p53 (U-251 MG, U-373 MG) protein. Our studies demonstrated that Delta-24 induced a more potent antiglioma effect in vitro than RA55. Moreover, Delta-24 replicated markedly more efficiently than RA55 in both wild-type and mutant p53 scenarios. Importantly, direct intratumoral injection of Delta-24, but not RA55, significantly suppresses tumor growth in intracranial (U-87 MG, U-251 MG) or subcutaneous (D54 MG) animal models. Staining for hexon protein detected replicating adenoviruses in xenografts infected with Delta-24, but not with RA55. Collectively, these data indicate that E1A mutant adenoviruses targeting the Rb pathway are more powerful putative agents for antiglioma therapy than E1B mutant adenoviruses, and suggest that E1A mutant adenoviruses should be tested in the clinical setting for patients with malignant gliomas.


Subject(s)
Adenovirus E1A Proteins/genetics , Adenovirus E1B Proteins/genetics , Adenoviruses, Human/genetics , Glioma/pathology , Glioma/virology , Adenoviridae/genetics , Adenovirus E1A Proteins/deficiency , Adenovirus E1B Proteins/deficiency , Animals , Cell Line, Tumor , Cell Survival , Gene Deletion , Glioma/metabolism , Humans , Mice , Mice, Nude , Transplantation, Heterologous , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Virus Replication
6.
Neurology ; 63(3): 418-26, 2004 Aug 10.
Article in English | MEDLINE | ID: mdl-15304571

ABSTRACT

Oncolytic or tumor-selective adenoviruses are constructed as novel antiglioma therapies. After infection, the invading genetic adenoviral material is activated within the host cell. E1A and E1B adenoviral proteins are expressed immediately. E1A protein interacts with cell cycle regulatory proteins, such as retinoblastoma (Rb), driving the cell into the S phase and ensuing viral replication. The action of E1A stimulates the cellular p53 tumor suppressor system, which results in growth arrest or apoptosis, and halts adenovirus replication. However, adenoviral E1B interacts with p53 protein, preventing the DNA replication process from being abrogated by the induction of p53-mediated apoptosis. It was subsequently hypothesized that mutant adenoviruses that were unable to express wild-type E1A or E1B proteins could not replicate in normal cells with functional Rb or p53 pathways but instead would replicate and kill glioma cells that had defects in the regulation of these tumor suppressor pathways. Mutant E1B adenoviruses have already entered the clinical setting as an experimental treatment for patients with malignant gliomas. Mutant E1A adenoviruses are now in preclinical development as antiglioma therapy. In this review, the authors describe the mechanisms underlying the production of oncolytic adenoviruses, preclinical and clinical experiences with specific oncolytic adenoviruses, and the possibilities of combining mutant oncolytic adenoviruses with gene therapy or conventional therapies for managing malignant gliomas.


Subject(s)
Adenovirus E1A Proteins/deficiency , Adenovirus E1B Proteins/deficiency , Adenoviruses, Human/physiology , Biological Therapy/methods , Brain Neoplasms/therapy , Defective Viruses/physiology , Glioma/therapy , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/physiology , Adenovirus E1B Proteins/genetics , Adenovirus E1B Proteins/physiology , Adenoviruses, Human/genetics , Apoptosis , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/radiotherapy , Cell Cycle , Combined Modality Therapy , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Cranial Irradiation , Cytopathogenic Effect, Viral , Defective Viruses/genetics , Gene Expression Regulation, Viral , Genes, Viral/genetics , Genetic Therapy , Glioma/drug therapy , Glioma/genetics , Glioma/metabolism , Glioma/radiotherapy , Models, Neurological , Oligopeptides/genetics , Promoter Regions, Genetic/genetics , Receptors, Virus/deficiency , Retinoblastoma Protein/deficiency , Retinoblastoma Protein/physiology , Species Specificity , Transcription, Genetic , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/physiology , Virus Replication
7.
Proc Natl Acad Sci U S A ; 101(9): 3124-9, 2004 Mar 02.
Article in English | MEDLINE | ID: mdl-14976240

ABSTRACT

Replication-defective human adenovirus (Ad) group C transducing vectors, most of which have the E1A, E1B, and E3 genes deleted, are highly inflammatory despite the fact that the parental viruses typically cause subclinical or mild infections. To investigate this paradox, the roles that the E1A, E1B, and E3 genes play in inflammation were tested by using replication-incompetent viruses carrying a deletion of the preterminal protein gene. The viruses were injected into BALB/c mouse ears, and edema was monitored as a sensitive surrogate marker of inflammation. A virus deleted for the E1A 289R (transcription activating) protein was noninflammatory, and inhibited edema induced by empty virus particles. The E1A 243R and E1B 55-kDa (p53 binding) proteins play the most important roles in inhibition of inflammation by the noninflammatory virus. The E1B 19-kDa antiapoptotic protein inhibited edema when both the E1A 243R and E1B 55-kDa proteins were expressed but strongly induced edema when only one was expressed. E3 proteins had their greatest effect on the inhibition of edema induced by the E1A 289R protein. The results support a model in which inflammation is countered through a mechanism that involves complex genetic interactions between Ad early region proteins and offer promise for the design and construction of noninflammatory Ad gene therapy vectors that are relatively easy to grow and purify.


Subject(s)
Adenoviridae Infections/physiopathology , Adenovirus E1A Proteins/genetics , Adenovirus E1B Proteins/genetics , Adenoviruses, Human/genetics , Adenoviridae Infections/prevention & control , Adenovirus E1A Proteins/deficiency , Adenovirus E1B Proteins/deficiency , Animals , Base Sequence , Cell Line , DNA Primers , Edema/virology , Gene Deletion , Genotype , Humans , Inflammation/physiopathology , Inflammation/prevention & control , Kidney , Kinetics , Mice , Mice, Inbred BALB C , Phenotype
8.
Cancer Res ; 63(12): 3073-8, 2003 Jun 15.
Article in English | MEDLINE | ID: mdl-12810631

ABSTRACT

The adenovirus E1A proteins are involved in the transcriptional activation of viral and cellular genes needed for controlling cell cycle and virus replication. Undifferentiated embryonic carcinoma cells have the ability to produce an E1A-like activity that can induce the expression of E1A-targeted adenoviral and cellular genes in the absence of the E1A products. Differentiated embryonic carcinoma cells lose the ability to produce the E1A-like activity. In this study, we investigated the E1A-like activity in cancer cells with an adenovirus having a mutated E1a gene. The mutation is generated by the insertion of a large DNA fragment in the E1a gene and interrupts the COOH-terminal region of both the E1A 12S and 13S proteins. The E1a-mutated virus can efficiently replicate in HepG2 and Hep3B liver cancer cells and produce high titers of virus. Replication of the E1a-mutated virus inhibits tumor formation and destroys tumors in vivo. The results obtained in this study imply that cancer cells may produce an E1A-like activity to support the selective replication of mutated virus in cancer cells. In addition, we found that although the E1a-mutated virus could not replicate in Huh1.cl2 liver cells, the viral DNA could amplify in the cells. This result suggests that replication of adenoviral DNA is necessary, but not sufficient, for generating infectious viral progeny and destroying tumor cells.


Subject(s)
Adenovirus E1A Proteins/genetics , Biological Therapy , Carcinoma, Hepatocellular/pathology , Defective Viruses/physiology , Liver Neoplasms/pathology , Mastadenovirus/physiology , Adenovirus E1A Proteins/deficiency , Adenovirus E1A Proteins/physiology , Animals , Carcinoma, Hepatocellular/therapy , Cytopathogenic Effect, Viral , DNA, Viral/genetics , Defective Viruses/genetics , Humans , Liver Neoplasms/therapy , Liver Neoplasms/virology , Mastadenovirus/genetics , Mice , Mice, Nude , Mutagenesis, Insertional , Tumor Cells, Cultured/virology , Virus Integration , Virus Replication , Xenograft Model Antitumor Assays
9.
Hum Gene Ther ; 13(15): 1873-85, 2002 Oct 10.
Article in English | MEDLINE | ID: mdl-12396619

ABSTRACT

Local overexpression of genes that promote lung defense or repair may be helpful in protecting the immature neonatal lung from injuries, but whether the vectors used to administer these genes affect physiological postnatal lung growth has not been investigated. We explored the effect on alveolarization of E1-deleted Adnull vector (Ad5-LMP-null) given intratracheally to 3-day-old rats. Three Adnull doses were evaluated 10(8), 5 x 10(8), and 10(9) TCID(50). Lung morphometry on day 21 showed significant growth disorders with the two higher doses. With 5 x 10(8) TCID(50), absolute lung volume increased significantly (+16%), as did absolute (+20%) and specific (+32%) alveolar airspace volumes, whereas alveolar surface density decreased by 13% (p < 0.009 for all parameters). Lung inflammation was mild, nonsignificant, and occurred mainly with the highest Adnull dose, indicating that it was unlikely to contribute to our results. Adnull instillation induced a significant#10; decrease in terminal bronchiolar cell proliferation as evaluated by proliferating cell nuclear antigen immunostaining (p = 0.02), as well as a 23% decrease in absolute parenchyma elastic fiber length (p = 0.02). Furthermore, lung tropoelastin mRNA content decreased by 25% (p < 0.02). In conclusion, E1-deleted adenoviral vectors can induce lung growth disorders when instilled into the airways of neonatal rats. Interactions with lung matrix turnover may be the main explanation to these deleterious effects.


Subject(s)
Adenoviruses, Human/physiology , Genetic Vectors/toxicity , Lung/virology , Adenovirus E1A Proteins/deficiency , Adenovirus E1A Proteins/physiology , Adenoviruses, Human/genetics , Animals , Animals, Newborn , Bronchi/pathology , Bronchi/virology , Bronchoalveolar Lavage Fluid , Cell Division , Defective Viruses/genetics , Defective Viruses/physiology , Dose-Response Relationship, Drug , Elastic Tissue/pathology , Elastin/metabolism , Epithelial Cells/pathology , Gene Expression Regulation, Viral , Genes, Reporter , Genetic Vectors/pharmacology , Inflammation , Instillation, Drug , Lung/metabolism , Lung/pathology , Lung Volume Measurements , RNA, Messenger/biosynthesis , Rats , Recombinant Fusion Proteins/biosynthesis , Trachea , Transfection , Tropoelastin/biosynthesis , Tropoelastin/genetics , beta-Galactosidase/biosynthesis , beta-Galactosidase/genetics
10.
Hum Gene Ther ; 13(15): 1887-900, 2002 Oct 10.
Article in English | MEDLINE | ID: mdl-12396620

ABSTRACT

Systemic administration of adenoviral vectors leads to activation of innate and antigen-specific immunity. In an attempt to diminish T and B cell-specific immune responses to E1-deleted adenoviral vectors, capsid proteins were modified with various activated monomethoxypolyethylene glycols (MPEGs). The impact of this modification was studied in a murine model of liver-directed gene transfer in which an E1-deleted adenovirus expressing the lacZ gene was given intravenously. The efficiency of vector transduction of hepatocytes in vivo was not compromised by any of the polymer chemistries. PEGylation of the virus, however, diminished the activation of cytotoxic T lymphocytes and helper T cells of the type 1 subset (Th1 cells) against native viral antigens; neutralizing antibodies to native virus were also diminished. PEGylation prolonged transgene expression and allowed partial readministration with native virus or with a virus PEGylated with a heterologous chemical moiety. Apparently, modification of the capsid leads to a shift in antigenic epitopes because vector readministration was not possible when the immunizing vector had been modified by the same PEGylation chemistry used to modify the second vector. In light of these results, the concept of improving the performance of adenoviral vectors through modification of the capsid with PEG shows promise.


Subject(s)
Adenoviruses, Human/physiology , Capsid Proteins/chemistry , Defective Viruses/physiology , Gene Expression Regulation, Viral , Genetic Vectors/pharmacokinetics , Liver/virology , Polyethylene Glycols/pharmacokinetics , Sulfones/pharmacokinetics , Transduction, Genetic/methods , Adenovirus E1A Proteins/deficiency , Adenovirus E1A Proteins/genetics , Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Animals , Antibodies, Viral/biosynthesis , Cytokines/metabolism , Defective Viruses/genetics , Defective Viruses/immunology , Genes, Reporter , Genetic Vectors/administration & dosage , Genetic Vectors/physiology , Green Fluorescent Proteins , Hepatocytes/metabolism , Hepatocytes/virology , Injections, Intravenous , Lac Operon , Liver/metabolism , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Mice , Mice, Inbred C57BL , Neutralization Tests , Polyethylene Glycols/administration & dosage , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Sulfones/administration & dosage , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism
11.
Nat Genet ; 4(1): 27-34, 1993 May.
Article in English | MEDLINE | ID: mdl-7685651

ABSTRACT

We describe the use of a human bronchial xenograft model for studying the efficiency and biology of in vivo gene transfer into human bronchial epithelia with recombinant E1 deleted adenoviruses. All cell types in the surface epithelium except basal cells efficiently expressed the adenoviral transduced recombinant genes, lacZ and CFTR, for 3-5 weeks. Stable transgene expression was associated with high level expression of the early adenoviral gene, E2a, in a subset of transgene expressing cells and virtually undetectable expression of the late adenoviral genes encoding the structural proteins, hexon and fiber. These studies begin to address important issues that relate to safety and in vivo efficacy of recombinant adenoviruses for gene delivery into the human airway.


Subject(s)
Adenoviruses, Human/genetics , Bronchi/transplantation , Defective Viruses/genetics , Genetic Therapy/methods , Genetic Vectors , Membrane Proteins/genetics , Transfection , Transplantation, Heterologous , Adenovirus E1A Proteins/deficiency , Adenovirus E1A Proteins/genetics , Adenoviruses, Human/isolation & purification , Animals , Bronchi/metabolism , Bronchi/microbiology , Cell Differentiation , Cystic Fibrosis Transmembrane Conductance Regulator , Defective Viruses/isolation & purification , Epithelium/metabolism , Epithelium/microbiology , Epithelium/transplantation , Gene Expression , Humans , Leg , Male , Membrane Proteins/biosynthesis , Mice , Mice, Nude , Rats , Rats, Inbred F344 , Recombinant Fusion Proteins/biosynthesis , Safety , Trachea/transplantation , Transplantation, Heterotopic , Viral Proteins/biosynthesis , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...