Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
PLoS One ; 3(6): e2409, 2008 Jun 18.
Article in English | MEDLINE | ID: mdl-18560559

ABSTRACT

BACKGROUND: Viral-mediated oncolysis is a novel cancer therapeutic approach with the potential to be more effective and less toxic than current therapies due to the agents selective growth and amplification in tumor cells. To date, these agents have been highly safe in patients but have generally fallen short of their expected therapeutic value as monotherapies. Consequently, new approaches to generating highly potent oncolytic viruses are needed. To address this need, we developed a new method that we term "Directed Evolution" for creating highly potent oncolytic viruses. METHODOLOGY/PRINCIPAL FINDINGS: Taking the "Directed Evolution" approach, viral diversity was increased by pooling an array of serotypes, then passaging the pools under conditions that invite recombination between serotypes. These highly diverse viral pools were then placed under stringent directed selection to generate and identify highly potent agents. ColoAd1, a complex Ad3/Ad11p chimeric virus, was the initial oncolytic virus derived by this novel methodology. ColoAd1, the first described non-Ad5-based oncolytic Ad, is 2-3 logs more potent and selective than the parent serotypes or the most clinically advanced oncolytic Ad, ONYX-015, in vitro. ColoAd1's efficacy was further tested in vivo in a colon cancer liver metastasis xenograft model following intravenous injection and its ex vivo selectivity was demonstrated on surgically-derived human colorectal tumor tissues. Lastly, we demonstrated the ability to arm ColoAd1 with an exogenous gene establishing the potential to impact the treatment of cancer on multiple levels from a single agent. CONCLUSIONS/SIGNIFICANCE: Using the "Directed Evolution" methodology, we have generated ColoAd1, a novel chimeric oncolytic virus. In vitro, this virus demonstrated a >2 log increase in both potency and selectivity when compared to ONYX-015 on colon cancer cells. These results were further supported by in vivo and ex vivo studies. Furthermore, these results have validated this methodology as a new general approach for deriving clinically-relevant, highly potent anti-cancer virotherapies.


Subject(s)
Colonic Neoplasms/therapy , Directed Molecular Evolution , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Adenoviridae/physiology , Animals , Cell Line, Tumor , Humans , Liver Neoplasms/secondary , Mice , Neoplasm Transplantation , Oncolytic Viruses/genetics
2.
Cancer Res ; 67(12): 5929-39, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575163

ABSTRACT

Accumulating data indicate that tumor-infiltrating regulatory T cells (Treg) are present in human tumors and locally suppress antitumor immune cells. In this study, we found an increased Treg/CD8 ratio in human breast and cervical cancers. A similar intratumoral lymphocyte pattern was observed in a mouse model for cervical cancer (TC-1 cells). In this model, systemic Treg depletion was inefficient in controlling tumor growth. Furthermore, systemic CTL-associated antigen-4 (CTLA-4) blockade, an approach that can induce tumor immunity in other tumor models, did not result in TC-1 tumor regression but led to spontaneous development of autoimmune hepatitis. We hypothesized that continuous expression of an anti-CTLA-4 antibody localized to the tumor site could overcome Treg-mediated immunosuppression and locally activate tumor-reactive CD8+ cells, without induction of autoimmunity. To test this hypothesis, we created TC-1 cells that secrete a functional anti-CTLA-4 antibody (TC-1/alphaCTLA-4-gamma1 cells). When injected into immunocompetent mice, the growth of TC-1/alphaCTLA-4-gamma1 tumors was delayed compared with control TC-1 cells and accompanied by a reversion of the intratumoral Treg/CD8 ratio due to an increase in tumor-infiltrating IFNgamma-producing CD8+ cells. When local anti-CTLA-4 antibody production was combined with Treg inhibition, permanent TC-1 tumor regression and immunity was induced. Importantly, no signs of autoimmunity were detected in mice that received local CTLA-4 blockade alone or in combination with Treg depletion.


Subject(s)
Antigens, CD/metabolism , Antigens, Differentiation/metabolism , Breast Neoplasms/immunology , Immunotherapy/methods , T-Lymphocytes, Regulatory/immunology , Uterine Cervical Neoplasms/immunology , Animals , Antigens, CD/immunology , Antigens, Differentiation/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/transplantation , CTLA-4 Antigen , Female , Flow Cytometry , Humans , Immunohistochemistry , Lymphocyte Depletion , Lymphocyte Subsets/immunology , Lymphocyte Subsets/transplantation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/transplantation , Mice , T-Lymphocytes, Regulatory/transplantation
3.
J Virol ; 81(9): 4866-71, 2007 May.
Article in English | MEDLINE | ID: mdl-17301138

ABSTRACT

Intravenous (i.v.) delivery of recombinant adenovirus serotype 5 (Ad5) vectors for gene therapy is hindered by safety and efficacy problems. We have discovered a new pathway involved in unspecific Ad5 sequestration and degradation. After i.v. administration, Ad5 rapidly binds to circulating platelets, which causes their activation/aggregation and subsequent entrapment in liver sinusoids. Virus-platelet aggregates are taken up by Kupffer cells and degraded. Ad sequestration in organs can be reduced by platelet depletion prior to vector injection. Identification of this new sequestration mechanism and construction of vectors that avoid it could improve levels of target cell transduction at lower vector doses.


Subject(s)
Adenoviridae/metabolism , Blood Platelets/metabolism , Genetic Therapy/methods , Genetic Vectors/metabolism , Liver/virology , Mononuclear Phagocyte System/virology , Animals , Genetic Vectors/blood , Mice , Mice, Transgenic
4.
Mol Ther ; 15(1): 193-202, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17164791

ABSTRACT

Baculovirus vectors are able to transduce a large variety of mammalian cell types and express transgenes placed under the control of heterologous promoters. In this study, we evaluated the potential of baculovirus vectors for malaria vaccination. To induce efficient CD4(+) and CD8(+) T-cell responses, we produced a series of vectors that display the Plasmodium falciparum circumsporozoite (CS) protein in the virion envelope and/or allow for CS expression upon transduction of mammalian cells. We found that baculovirus vectors can transduce professional antigen-presenting cells and trigger their maturation, which is a prerequisite for efficient antigen presentation. Upon intramuscular injection into mice, the vector that both displayed and expressed CS induced higher anti-CS antibody titers (of the immunoglobulin (IgG)1 and IgG2a type) and a higher frequency of interferon-gamma-producing T cells specific to CS, than the vectors which either only displayed or only expressed CS. The baculovirus CS display/expression vector was also superior in inducing CS-specific CD4(+) and CD8(+) T-cell responses in vitro using human peripheral blood mononuclear cells from naive donors. This, together with the absence of pre-existing immunity to baculoviruses in humans, the absence of viral gene expression in mammalian cells, and the relative low immunogenicity of baculovirus virions, makes these vectors promising tools for vaccination. Furthermore, the ability to produce large amounts in serum-free medium at a low cost adds a further advantage to this vector system.


Subject(s)
Baculoviridae/genetics , Baculoviridae/immunology , Genetic Vectors/genetics , Protozoan Proteins/immunology , Vaccination , Animals , Antibodies, Protozoan/immunology , Cell Line , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Leukocytes/immunology , Leukocytes/metabolism , Mice , Mice, Inbred BALB C , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Spodoptera
5.
Blood ; 108(8): 2554-61, 2006 Oct 15.
Article in English | MEDLINE | ID: mdl-16788098

ABSTRACT

Upon local delivery, adenovirus (Ad) serotype 5 viruses use the coxsackie and Ad receptor (CAR) for cell binding and alpha(v) integrins for internalization. When administered systemically, however, their role in liver tropism is limited because CAR-permissive and mutated viruses show similar biodistribution, a finding recently attributed to blood coagulation factor (F) IX or complement protein C4BP binding to the adenovirus fiber and "bridging" to either low-density lipoprotein receptor-related protein or heparan sulfate proteoglycans. Here, we show that hepatocyte transduction in vitro can be enhanced by the vitamin K-dependent factors FX, protein C, and FVII in addition to FIX but not by prothrombin (FII), FXI, and FXII. This phenomenon was not dependent on proteolytic activation or cell signaling activity and for FX was mediated by direct virus-factor binding. Human FX substantially enhanced hepatocyte transduction by CAR-permissive and mutated viruses in an ex vivo liver perfusion model. In vivo, global down-regulation of vitamin K-dependent zymogens by warfarin significantly diminished liver uptake of CAR-deleted Ads; however, this phenomenon was fully rescued by acute infusion of human FX. Our results indicate a common and pivotal role for distinct vitamin K-dependent coagulation factors in mediating hepatocyte transduction by adenoviruses in vitro and in vivo.


Subject(s)
Adenoviruses, Human/genetics , Blood Coagulation Factors/metabolism , Gene Transfer Techniques , Hepatocytes/metabolism , Hepatocytes/virology , Vitamin K/metabolism , Animals , Cell Line , Coxsackie and Adenovirus Receptor-Like Membrane Protein , Enzyme Precursors/metabolism , Factor IX/genetics , Factor IX/metabolism , Factor IX/pharmacology , Factor X/metabolism , Hepatocytes/drug effects , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Virus/metabolism , Signal Transduction , Transduction, Genetic , Warfarin/pharmacology
6.
Mol Ther ; 13(4): 756-65, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16461009

ABSTRACT

In contrast to commonly used serotype 5-based adenovirus (Ad) vectors, Ad's containing fibers derived from B-group serotype 35 (Ad5/35) efficiently transduce human DCs ex vivo and appear to target antigen-presenting cells after intravenous injection into baboons. Based on this, Ad5/35 vectors could be valuable tools for immunotherapy and vaccination. On the other hand, a number of studies indicate that signaling through the B-group Ad receptor, CD46, can cause tolerance or immunosuppression. Since mice do not express CD46 in a human-like pattern, we studied the in vivo properties of Ad5/35 in transgenic mice that express CD46 in a pattern and at a level similar to those of humans. Hypersensitivity assays and analyses of frequencies of regulatory T cells and T cell responses did not indicate that Ad5/35 injection exerts detrimental effects on the host's immune system. An Ad5/35 vector expressing a model antigen was able to trigger a strong T cell response against the test antigen after intramuscular injection. Overall, compared to Ad5 vectors, Ad5/35 vectors had a better safety profile, reflected by lower serum levels of proinflammatory cytokines.


Subject(s)
Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Vaccination , Viral Vaccines , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Dendritic Cells/immunology , Dendritic Cells/virology , Escherichia coli/genetics , Female , Fluorescent Antibody Technique , Gene Expression , Genome, Viral , Green Fluorescent Proteins/metabolism , Humans , Immunity, Innate , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Membrane Cofactor Protein/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Transduction, Genetic , Transgenes
7.
Cancer Res ; 66(2): 960-9, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16424031

ABSTRACT

Heat shock proteins such as gp96 have the ability to chaperone peptides and activate antigen-presenting cells. In this study, we tested whether adenovirus-mediated overexpression of secreted or membrane-associated forms of gp96 in tumor cells would stimulate an antitumor immune response. Studies were carried out in C57Bl/6 mice bearing aggressively growing s.c. tumors derived from syngeneic TC-1 cells, a cell line that expresses HPV16 E6 and E7 proteins. We found that secreted gp96 can induce protective and therapeutic antitumor immune responses. Our data also indicate that the antitumor effect of sgp96 expression seems to be limited by the induction of suppressive regulatory T cells (Treg). TC-1 tumor transplantation increased the number of splenic and tumor-infiltrating Tregs. Importantly, treatment of mice with low-dose cyclophosphamide decreased the number of Tregs and enhanced the immunostimulatory effect of sgp96 expression. We also tested whether an oncolytic vector (Ad.IR-E1A/TRAIL), that is able to induce tumor cell apoptosis and, potentially, release cryptic tumor epitopes in immunogenic form, could stimulate antitumor immune responses. Although tumor cells infected ex vivo with Ad.IR-E1A/TRAIL had no antitumor effect when used as a vaccine alone, the additional treatment with low-dose cyclophosphamide resulted in the elimination of pre-established tumors. This study gives a rationale for testing approaches that suppress Tregs in combination with oncolytic or immunostimulatory vectors.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Cyclophosphamide/pharmacology , Heat-Shock Proteins/biosynthesis , Adenoviridae/genetics , Animals , Antigen-Presenting Cells/immunology , Cancer Vaccines , Dose-Response Relationship, Drug , Genetic Vectors , Human papillomavirus 16 , Humans , Membrane Glycoproteins , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/therapy , Oncogene Proteins, Viral , Oncolytic Virotherapy , Repressor Proteins , Transplantation, Heterologous , Tumor Cells, Cultured/immunology
8.
Hum Gene Ther ; 16(6): 664-77, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15960598

ABSTRACT

Vectors containing group B adenovirus (Ad) fibers are able to efficiently transduce gene therapy targets that are refractory to infection with standard Ad serotype 5 (Ad5) vectors, including malignant tumor cells, hematopoietic stem cells, and dendritic cells. Preliminary studies in mice indicate that, after intravenous injection, B-group fiber-containing Ads do not efficiently transduce most organs and cause less acute toxicity than Ad5 vectors. However, biodistribution and safety studies in mice are of limited value because the mouse analog of the B-group Ad receptor, CD46, is expressed only in the testis, whereas in humans, CD46 is expressed on all nucleated cells. Unlike mice, baboons have CD46 expression patterns and levels that closely mimic those in humans. We conducted a biodistribution and toxicity study of group B Ad fiber-containing vectors in baboons. Animals received phosphate-buffered saline, Ad5-bGal (a first-generation Ad5 vector), or B-group fiber-containing Ads (Ad5/35-bGal and Ad5/11-bGal) at a dose of 2 x 10(12) VP/kg, and vector biodistribution and safety was analyzed over 3 days. The amount of Ad5/35-bGal and Ad5/11-bGal vector genomes was in most tissues one to three orders of magnitude below that of Ad5. Significant Ad5/35- and Ad5/11-mediated transgene (beta-galactosidase) expression was seen only in the marginal zone of splenic follicles. Compared with the animal that received Ad5-bGal, all animals injected with B-group fiber-containing Ad vectors had lower elevations in serum proinflammatory cytokine levels. Gross and histopathology were normal in animals that received B-group Ad fiber-containing Ads, in contrast to the Ad5-infused animal, which showed widespread endothelial damage and inflammation. In a further study, a chimeric Ad5/35 vector carrying proapoptotic TRAIL and Ad E1A genes under tumor-specific regulation was well tolerated in a 30-day toxicity study. No major clinical, serologic, or pathologic abnormalities were noticed in this animal.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/pharmacokinetics , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , DNA Helicases/genetics , Escherichia coli Proteins , Genetic Vectors/administration & dosage , Injections, Intravenous , Male , Papio , Tissue Distribution , Toxicity Tests , beta-Galactosidase/blood , beta-Galactosidase/genetics , beta-Galactosidase/pharmacokinetics
9.
J Virol ; 79(12): 7478-91, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15919903

ABSTRACT

Adenoviruses (Ad) are efficient vehicles for gene delivery in vitro and in vivo. Therefore, they are a promising tool in gene therapy, particularly in the treatment of cancer and cardiovascular diseases. However, preclinical and clinical studies undertaken during the last decade have revealed a series of problems that limit both the safety and efficacy of Ad vectors, specifically after intravenous application. Major obstacles to clinical use include innate toxicity and Ad sequestration by nontarget tissues. The factors and mechanisms underlying these processes are poorly understood. The majority of intravenously injected Ad particles are sequestered by the liver, which in turn causes an inflammatory response characterized by acute transaminitis and vascular damage. Here, we describe a novel pathway that is used by Ad for infection of hepatocytes and Kupffer cells upon intravenous virus application in mice. We found that blood factors play a major role in targeting Ad vectors to hepatic cells. We demonstrated that coagulation factor IX and complement component C4-binding protein can bind the Ad fiber knob domain and provide a bridge for virus uptake through cell surface heparan sulfate proteoglycans and low-density lipoprotein receptor-related protein. An Ad vector, Ad5mut, which contained mutations in the fiber knob domain ablating blood factor binding, demonstrated significantly reduced infection of liver cells and liver toxicity in vivo. This study contributes to a better understanding of adenovirus-host interactions for intravenously applied vectors. It also provides a rationale for novel strategies to target adenovirus vector to specific tissues and to reduce virus-associated toxicity after systemic application.


Subject(s)
Adenoviruses, Human/pathogenicity , Factor IX/metabolism , Genetic Vectors , Hepatocytes/virology , Histocompatibility Antigens/metabolism , Kupffer Cells/virology , Adenoviruses, Human/genetics , Adenoviruses, Human/metabolism , Animals , Blood Coagulation Factors/metabolism , Capsid Proteins/metabolism , Cell Line , Cricetinae , Humans , Mice , Mice, Inbred C57BL , Transduction, Genetic
10.
Cancer Res ; 65(10): 4343-52, 2005 May 15.
Article in English | MEDLINE | ID: mdl-15899826

ABSTRACT

In this study, we identified murine breast cancer cell lines that support DNA replication of E1-deleted adenovirus vectors and which can be killed by an oncolytic adenovirus expressing adenovirus E1A and tumor necrosis factor (TNF)-related apoptosis inducing ligand (TRAIL) in a replication-dependent manner (Ad.IR-E1A/TRAIL). We showed that systemic or intratumoral (i.t.) injection of adenovirus vectors into mice increases plasma levels of proinflammatory cytokines and chemokines, including TNF-alpha, INF-gamma, and MCP-1, which are potent inducers of dendritic cell maturation. Furthermore, we showed that in vivo expression of Flt3L from an adenovirus vector increases the number of CD11b+ and CD11c+ cells (populations that include dendritic cells) in the blood circulation. Based on these findings, we tested whether Ad.IR-E1A/TRAIL induced killing of tumor cells in combination with dendritic cell mobilization by Ad.Flt3L or, for comparison, Ad.GM-CSF would have an additive antitumor effect. As a model, we used immunocompetent C3H mice with syngeneic s.c. tumors derived from C3L5 cells. We found that vaccination of mice with C3L5 cells that underwent viral oncolysis in combination with Flt3L or granulocyte-macrophage colony-stimulating factor (GM-CSF) expression induces a systemic antitumor immune response. I.t. injection of the oncolytic and Flt3L expressing vectors into established tumors delayed tumor growth but did not cause efficient tumor elimination. This study shows the effectiveness of a combined oncolytic/immunostimulatory tumor therapy approach.


Subject(s)
Adenoviridae/physiology , Cancer Vaccines/immunology , Immunotherapy/methods , Mammary Neoplasms, Experimental/therapy , Adenoviridae/genetics , Adenoviridae/immunology , Adenoviridae/metabolism , Adenovirus E1A Proteins/biosynthesis , Adenovirus E1A Proteins/genetics , Animals , Apoptosis Regulatory Proteins , CD11b Antigen/immunology , CD11b Antigen/metabolism , CD11c Antigen/immunology , CD11c Antigen/metabolism , Cancer Vaccines/genetics , DNA Replication , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Humans , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/virology , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
11.
J Immunol ; 174(11): 7310-9, 2005 Jun 01.
Article in English | MEDLINE | ID: mdl-15905578

ABSTRACT

The safety of gene therapy vectors is a major concern when novel viral or nonviral therapeutics are proposed for applications in humans. Adenovirus (Ad) vectors have been extensively used as efficient gene delivery vehicles in vitro over the last two decades. However, upon i.v. application, they elicit robust innate and inflammatory responses that may be fatal for the host. To date, the primary cytokines and chemokines involved in the initiation of these host responses remain illusive. In this study, we demonstrate that IL-1 is a major mediator involved in the initiation of immediate host responses toward i.v. applied Ad vectors. Using mice in which IL-1 signaling was genetically eliminated (IL-1RI-KO), or wild-type animals for which signaling was blocked by anti-IL-1 Abs, we found that i.v. applied Ad vectors elicited dramatically reduced acute inflammatory responses when compared with control animals. Importantly, the efficiency of Ad gene transfer in vivo was not significantly affected by interfering with IL-1 signaling. Using an in situ hybridization technique, we found that hepatocytes and Kupffer cells trigger IL-1 transcription in liver tissue after i.v. Ad vector administration. We also found that expression of the MIP-2 chemokine gene (which is responsible for recruitment of neutrophils to the liver) depends on IL-1 activation. Our data indicate that immediate innate and inflammatory host responses toward i.v. applied Ad vectors can be pharmacologically controlled through interference with IL-1 signaling pathways.


Subject(s)
Adenoviruses, Human/genetics , Adenoviruses, Human/immunology , Genetic Vectors/toxicity , Interleukin-1/physiology , Liver/immunology , Liver/virology , Signal Transduction/immunology , Animals , Chemokines/biosynthesis , Chemokines/genetics , Cytokines/biosynthesis , Cytokines/genetics , Gene Expression Regulation/immunology , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Genetic Vectors/immunology , Inflammation Mediators/administration & dosage , Inflammation Mediators/immunology , Inflammation Mediators/toxicity , Injections, Intravenous , Interleukin-1/antagonists & inhibitors , Interleukin-1/biosynthesis , Interleukin-1/genetics , Kupffer Cells/immunology , Kupffer Cells/metabolism , Kupffer Cells/virology , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Interleukin-1/deficiency , Receptors, Interleukin-1/genetics , Receptors, Interleukin-1/physiology , Signal Transduction/genetics , Tumor Necrosis Factor-alpha/physiology , Up-Regulation/genetics , Up-Regulation/immunology
12.
J Virol ; 79(8): 5090-104, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15795294

ABSTRACT

Adenovirus vectors based on human serotype 5 (Ad5) have successfully been used as gene transfer vectors in many gene therapy-based approaches to treat disease. Despite their widespread application, many potential therapeutic applications are limited by the widespread prevalence of vector-neutralizing antibodies within the human population and the inability of Ad5-based vectors to transduce important therapeutic target cell types. In an attempt to circumvent these problems, we have developed Ad vectors based on human Ad serotype 11 (Ad11), since the prevalence of neutralizing antibodies to Ad11 in humans is low. E1-deleted Ad11 vector genomes were generated by homologous recombination in 293 cells expressing the Ad11-E1B55K protein or by recombination in Escherichia coli. E1-deleted Ad11 genomes did not display transforming activity in rodent cells. Transduction of primary human CD34+ hematopoietic progenitor cells and immature dendritic cells was more efficient with Ad11 vectors than with Ad5 vectors. Thirty minutes after intravenous injection into mice that express one of the Ad11 receptors (CD46), we found, in a pattern and at a level comparable to what is found in humans, Ad11 vector genomes in all analyzed organs, with the highest amounts in liver, lung, kidney, and spleen. Neither Ad11 genomes nor Ad11 vector-mediated transgene expression were, however, detected at 72 h postinfusion. A large number of Ad11 particles were also found to be associated with circulating blood cells. We also discovered differences in in vitro transduction efficiencies and in vivo biodistributions between Ad11 vectors and chimeric Ad5 vectors possessing Ad11 fibers, indicating that Ad11 capsid proteins other than fibers influence viral infectivity and tropism. Overall, our study provides a basis for the application of Ad11 vectors for in vitro and in vivo gene transfer and for gaining an understanding of the factors that determine Ad tropism.


Subject(s)
Adenoviruses, Human/classification , Adenoviruses, Human/genetics , Gene Transfer Techniques , Genetic Vectors , Adenoviruses, Human/ultrastructure , Base Sequence , Cell Line , Dendritic Cells/virology , Genetic Complementation Test , Hematopoietic Stem Cells/virology , Humans , Kidney , Plasmids/genetics , Polymerase Chain Reaction , Recombinant Fusion Proteins/metabolism , Sequence Deletion , Serotyping
13.
Mol Ther ; 9(5): 650-7, 2004 May.
Article in English | MEDLINE | ID: mdl-15120325

ABSTRACT

The improvement of initial tumor cell transduction with viral vectors is a major task in tumor gene therapy. We have developed mouse tumor models with hepatic metastases to study transduction of tumor cells after systemic adenovirus vector application. The tumor models were established by intraportal transplantation of human tumor cell lines into immunodeficient mice. Liver metastases derived from cervix, colon, breast, and liver cancer lines were analyzed for distribution of extracellular matrix, vascularization, and transgene expression after tail vein injection of adenovirus vectors. Overall, xenografts resembled the morphology of corresponding tumors in cancer patients. Adenovirus-mediated gene delivery depended on tumor vascularization and direct contact between blood vessels and tumor cells. These models represent important tools for studying and improving tumor gene therapy approaches.


Subject(s)
Adenoviridae/genetics , Disease Models, Animal , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Mice , Transduction, Genetic , Xenograft Model Antitumor Assays , Animals , Antibodies/immunology , Carcinoma/pathology , Carcinoma/secondary , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Colonic Neoplasms/pathology , Female , Genetic Vectors , Humans , Immunochemistry , Laminin/immunology , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Transplantation, Heterologous , Uterine Cervical Neoplasms/pathology , beta-Galactosidase/immunology
14.
J Virol ; 78(10): 5368-81, 2004 May.
Article in English | MEDLINE | ID: mdl-15113916

ABSTRACT

After intravenous administration, adenovirus (Ad) vectors are predominantly sequestered by the liver. Delineating the mechanisms for Ad accumulation in the liver is crucial for a better understanding of Ad clearance and Ad-associated innate toxicity. To help address these issues, in this study, we used Ad vectors with different fiber shaft lengths and either coxsackievirus-Ad receptor (CAR)-interacting Ad serotype 9 (Ad9) or non-CAR-interacting Ad35 fiber knob domains. We analyzed the kinetics of Ad vector accumulation in the liver, uptake into hepatocytes and Kupffer cells, and induction of cytokine expression and release in response to systemic vector application. Immediately after intravenous injection, all Ad vectors accumulated equally efficiently in the liver; however, only genomes of long-shafted Ads were maintained in the liver tissue over time. We found that Kupffer cell uptake of long-shafted Ads was mediated by the fiber knob domain and was CAR independent. The short-shafted Ads were unable to efficiently interact with hepatocellular receptors and were not taken up by Kupffer cells. Moreover, our studies indicated that Kupffer cells were not the major reservoir for the observed accumulation of Ads (used in this study) in the liver within the first 30 min after virus infusion. The lower level of liver cell transduction by short-shafted Ads correlated with a significantly reduced inflammatory anti-Ad response as well as liver damage induced by the systemic administration of these vectors. This study contributes to a better understanding of the biology of systemically applied Ad and will help in designing safer vectors that can efficiently transduce target tissues.


Subject(s)
Adenoviridae/genetics , Genetic Vectors/toxicity , Hepatocytes/virology , Liver/virology , Adenoviridae/pathogenicity , Alanine Transaminase/blood , Animals , Cytokines/genetics , Genetic Vectors/genetics , Interleukin-6/blood , Kupffer Cells/virology , Mice , Mice, Inbred C57BL , Receptors, Virus/physiology , Tumor Necrosis Factor-alpha/analysis
15.
Mol Ther ; 9(4): 496-509, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15093180

ABSTRACT

We have constructed a new capsid-modified adenovirus (Ad) vector that specifically replicates in tumor cells and expresses TNF-related apoptosis-inducing ligand (TRAIL). The Ad capsid contains short-shafted fibers derived from Ad serotype 35, which allow for efficient infection of malignant tumor cells, and largely avoids innate toxicity after intravenous application. Replication-dependent homologous recombination in Ad genomes was used to achieve tumor-specific expression of Ad E1a (to mediate viral replication) and TRAIL (to mediate apoptosis and enhance release of progeny virus from infected cells). We demonstrated that our oncolytic vector (Ad5/35.IR-E1A/TRAIL) induced apoptosis in human tumor cell lines derived from colorectal, lung, prostate, and liver cancer. Both in vitro and in vivo tumor models showed efficient intratumoral spread of this vector. In a model for metastatic colon cancer, tail vein infusion of Ad5/35.IR-E1A/TRAIL resulted in elimination of preestablished liver metastases. Intravenous injection of this vector caused a transient elevation of serum glutamic pyruvic transaminase in tumor-bearing mice, which we attributed to factors released from apoptotic tumor cells. Liver histology analyzed at day 14 after virus injection did not show signs of hepatocellular damage. This new oncolytic vector represents a potentially efficient means for gene therapy of metastatic cancer.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Liver Neoplasms/diagnosis , Liver Neoplasms/secondary , Adenovirus E1A Proteins/metabolism , Alanine Transaminase/blood , Animals , Antigens, CD/genetics , Antineoplastic Agents/pharmacology , Apoptosis , Apoptosis Regulatory Proteins , Capsid , Caspase 3 , Caspases/metabolism , Cell Line , Cell Line, Tumor , Cells, Cultured , Colonic Neoplasms/pathology , Female , Genetic Vectors , Genome, Viral , Humans , Immunohistochemistry , Membrane Cofactor Protein , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mice , Mice, Transgenic , Models, Genetic , Neoplasm Metastasis , Neoplasm Transplantation , Plasmids/metabolism , TNF-Related Apoptosis-Inducing Ligand , Time Factors , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/metabolism , Uterine Cervical Neoplasms/metabolism
16.
Mol Ther ; 8(5): 746-55, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14599807

ABSTRACT

Avoiding transduction of normal tissue after intravenous application of oncolytic adenoviruses (Ad) is an important strategy to improve the safety and efficacy of these vectors in gene therapy. As a model for a targeted vector, we used Ad vectors with type 35 fibers (Ad5/35), which efficiently transduce human cervical carcinoma cells but not liver cells. In an in vitro model of liver metastases, in which small nests of HeLa cells were surrounded by mouse hepatocytes, we showed that an Ad5/35-based conditionally replicating vector regulated by DNA replication-dependent recombination conferred increased gene transfer to tumor cells and enhanced viral replication and tumor cell lysis compared to the nontargeted Ad5 vector. Intravenous injection of Ad5/35 vectors into mice bearing liver metastases derived from HeLa cells caused markedly less hepatotoxicity than Ad5 vectors; however, it did not result in enhanced tumor cell transduction, viral replication, or oncolysis. Apparently, other factors, including the stability of virus in the blood, trapping within the liver sinusoids, transendothelial transfer, and/or vector diffusion of viral particles to tumor cells, limit tumor transduction, even if the vector is not taken up by liver cells.


Subject(s)
Adenoviridae/genetics , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Neoplasms/therapy , Animals , Cell Line , Cell Separation , Coculture Techniques , DNA/metabolism , Flow Cytometry , Green Fluorescent Proteins , HeLa Cells , Hepatocytes/metabolism , Humans , Immunohistochemistry , Liver/metabolism , Liver Neoplasms/secondary , Luminescent Proteins/metabolism , Mice , Neoplasm Metastasis , Plasmids/metabolism , Recombination, Genetic , Tissue Distribution , beta-Galactosidase/metabolism
17.
Biochem Biophys Res Commun ; 307(4): 759-64, 2003 Aug 08.
Article in English | MEDLINE | ID: mdl-12878174

ABSTRACT

We previously reported that the HS-4 insulator, derived from the chicken beta-globin locus, was able to shield a downstream inducible promoter from viral enhancers or silencers present in the genome of adenovirus vectors. In this study, we constructed two recombinant adenoviruses (Ad) that express an alkaline phosphatase (AP) reporter gene driven by an alpha-fetoprotein (AFP) enhancer/promoter with and without HS-4 insulator (Ad.HS4.AFP-AP and Ad.AFP-AP). The insulated vector, Ad.HS4.AFP-AP, conferred significantly higher AP expression than Ad.AFP-AP in all AFP-producing hepatocellular carcinoma cell lines (HepG2, Hep3B, and HuH7) examined. AP expression from Ad.HS4.AFP-AP was specific to hepatoma cells and barely detectable in AFP-negative tumor cell lines and normal human cells, including human hepatocytes. Intravenous infusion of viral vectors into mice with liver metastasis derived from Hep3B hepatoma cells resulted in AP expression exclusively localized to tumor cells. The number of tumor cells with detectable AP expression was significantly higher in mice infused with Ad.HS4.AFP-AP than in mice that received the non-insulated vector. This study demonstrates that the HS-4 insulator in the context of an Ad vector can increase the activity of the AFP promoter, while maintaining its tumor-specificity in vitro and in vivo. Considering that the anti-tumor activity of oncolytic vectors often depends on the level of pro-apoptotic or suicide gene expression, insulators might be a useful tool to improve the efficacy and specificity of these vectors.


Subject(s)
Adenoviridae/genetics , Carcinoma, Hepatocellular/metabolism , Genetic Vectors , Insulator Elements , Liver Neoplasms/metabolism , Alkaline Phosphatase/genetics , Animals , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cells, Cultured , Gene Expression Regulation , Genes, Reporter , Genetic Therapy , Globins/genetics , Humans , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Mice , Promoter Regions, Genetic , Transcription, Genetic , Tumor Cells, Cultured , alpha-Fetoproteins/genetics
18.
Cancer Res ; 62(21): 6089-98, 2002 Nov 01.
Article in English | MEDLINE | ID: mdl-12414633

ABSTRACT

Adenoviruses (Ads) that selectively replicate in tumor cells have shown promising preliminary results in clinical trials, especially in combination with chemotherapy. Here, we describe a system that combines the antitumor synergy of Ads and chemotherapeutic agents with the benefits of enzyme-activated prodrug therapy. In this system, a functional transgene expression cassette is created by homologous recombination during adenoviral DNA replication. Transgene expression is strictly dependent on viral DNA replication, which in turn is tumor specific. We constructed replication-activated Ad vectors to express a secreted form of beta-glucuronidase and a cytosine deaminase/uracil phosphoribosyltransferase, which activate the prodrugs 9-aminocamptothecin glucuronide to 9-aminocamptothecin and 5-fluorocytosine to 5-fluorouracil (5-FU) and further to 5-fluoro-UMP, respectively. We demonstrated replication-dependent transgene expression, prodrug activation, and induction of tumor cell toxicity by secreted beta-glucuronidase and cytosine deaminase/uracil phosphoribosyltransferase. Furthermore, exposure of cells to activated prodrug or drug at subtoxic concentrations enhanced viral DNA replication. Characteristically, these agents induced changes in the cell cycle status of exposed cells (G(2) arrest), which closely resembled the effect of wild-type Ad infection, and are thought to be favorable for viral replication. We tested a number of cytostatic drugs (camptothecin, etoposide, daunorubicin, cisplatin, 5-fluorouracil, hydroxyurea, Taxol, and actinomycin D) for their effect on viral DNA replication and found considerable differences between individual agents. Finally, we show that the combination of viral and prodrug therapy enhances viral replication and spread in liver metastases derived from human colon carcinoma or cervical carcinoma in a mouse model. Our data indicate that specific vector/drug combinations tailored to be synergistic may have the potential to improve the potency of either therapeutic approach. These data also provide a new rationale for expressing prodrug-activating enzymes from conditionally replicating Ads.


Subject(s)
Adenoviridae/physiology , Camptothecin/analogs & derivatives , Genetic Therapy/methods , Glucuronidase/metabolism , Nucleoside Deaminases/metabolism , Pentosyltransferases/metabolism , Prodrugs/pharmacokinetics , Adenoviridae/drug effects , Biotransformation , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Cell Cycle/drug effects , Combined Modality Therapy , Cytopathogenic Effect, Viral/drug effects , Cytosine Deaminase , DNA Replication/drug effects , Flucytosine/pharmacokinetics , Fluorouracil/pharmacology , Glucuronidase/genetics , Glucuronides/pharmacokinetics , HeLa Cells , Humans , Nucleoside Deaminases/genetics , Pentosyltransferases/genetics , Transgenes , Virus Replication/drug effects
19.
J Virol ; 76(21): 10994-1002, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12368342

ABSTRACT

We have developed a new class of adenovirus vectors that selectively replicate in tumor cells. The vector design is based on our recent observation that a variety of human tumor cell lines support DNA replication of adenovirus vectors with deletions of the E1A and E1B genes, whereas primary human cells or mouse liver cells in vivo do not. On the basis of this tumor-selective replication, we developed an adenovirus system that utilizes homologous recombination between inverted repeats to mediate precise rearrangements within the viral genome resulting in replication-dependent activation of transgene expression in tumors (Ad.IR vectors). Here, we used this system to achieve tumor-specific expression of adenoviral wild-type E1A in order to enhance viral DNA replication and spread within tumor metastases. In vitro DNA replication and cytotoxicity studies demonstrated that the mechanism of E1A-enhanced replication of Ad.IR-E1A vectors is efficiently and specifically activated in tumor cells, but not in nontransformed human cells. Systemic application of the Ad.IR-E1A vector into animals with liver metastases achieved transgene expression exclusively in tumors. The number of transgene-expressing tumor cells within metastases increased over time, indicating viral spread. Furthermore, the Ad.IR-E1A vector demonstrated antitumor efficacy in subcutaneous and metastatic models. These new Ad.IR-E1A vectors combine elements that allow for tumor-specific transgene expression, efficient viral replication, and spread in liver metastases after systemic vector application.


Subject(s)
Adenoviruses, Human/genetics , DNA Replication , DNA, Viral/biosynthesis , Genetic Vectors/genetics , Virus Replication , Adenovirus E1 Proteins/genetics , Adenovirus E3 Proteins/genetics , Adenoviruses, Human/physiology , Animals , Consumer Product Safety , Cytopathogenic Effect, Viral , Disease Models, Animal , Genetic Vectors/physiology , HeLa Cells , Humans , Kinetics , Liver Neoplasms/secondary , Mice , Mutagenesis , Neoplasms, Experimental/secondary , Recombination, Genetic , Time Factors
20.
Mol Ther ; 5(4): 420-6, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11945069

ABSTRACT

Stable gene transfer to the liver by viral vectors is inefficient. In an attempt to stimulate expansion of retrovirally transduced hepatocytes, we employed a synthetic drug (AP20187) that can reversibly dimerize and activate fusion proteins that contain a growth factor receptor signaling domain. Signaling domains derived from receptors for interleukin-6 (gp130), hepatocyte growth factor (c-met), epithelial growth factor (EGF-R), and thrombopoietin (mpl) triggered monkey hepatocytes to enter the cell cycle. However, mitosis occurred only upon activation of the gp130 and c-met signaling domains. Primary mouse hepatocytes expressing the gp130 fusion proliferated transiently in response to AP20187. AP20187-triggered activation of gp130 also stimulated the selective (>2-fold) expansion of retrovirally transduced hepatocytes in vivo, as shown by immunohistochemical staining and quantitative proviral DNA analysis. Drug-inducible in vivo expansion of genetically modified hepatocytes may have potential applications in hepatic gene transfer or in liver repopulation by transplanted hepatocytes or their progenitors. (c)2002 Elsevier Science (USA).


Subject(s)
Cell Division/physiology , Hepatocytes/cytology , Tacrolimus/pharmacology , Animals , Cell Division/drug effects , Cell Line , Dimerization , Feasibility Studies , Gene Transfer Techniques , Genetic Vectors , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Macaca , Mice , Retroviridae/genetics , Tacrolimus/analogs & derivatives
SELECTION OF CITATIONS
SEARCH DETAIL
...