Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters










Publication year range
1.
Gene Ther ; 18(7): 727-33, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21390070

ABSTRACT

Small interfering RNAs (siRNAs) are emerging as promising therapeutic tools. However, the widespread clinical application of such molecules as modulators of gene expression is still dependent on several aspects that limit their bioavailability. One of the most promising strategies to overcome the barriers faced by gene silencing molecules involves the use of lipid-based nanoparticles (LNPs) and viral vectors, such as adenoviruses (Ads). The primary obstacle for translating gene silencing technology from an effective research tool into a feasible therapeutic strategy remains its efficient delivery to the targeted cell type in vivo. In this study, we tested the capability of LNPs and Ad to transduce and treat locally tumors in vivo. Efficient knockdown of a surrogate reporter (luciferase) and therapeutic target genes such as the kinesin spindle protein (KIF11) and polo-like kinase 1 were observed. Most importantly, this activity led to a cell cycle block as a consequence and slowed down tumor progression in tumor-bearing animals. Our data indicate that it is possible to achieve tumor transduction with si/short hairpin RNAs and further improve the delivery strategy that likely in the future will lead to the ideal non-viral particle for targeted cancer gene silencing.


Subject(s)
Cholesterol/analogs & derivatives , Disease Progression , Gene Targeting , Genes, cdc , Liposomes , Nanoparticles , Neoplasms/genetics , Neoplasms/therapy , Polyethylene Glycols/administration & dosage , RNA Interference , Adenoviridae/genetics , Animals , Cell Line, Tumor , Cholesterol/administration & dosage , Gene Silencing , Genetic Vectors , Humans , Male , Mice , Mice, Nude , Transduction, Genetic
2.
Cancer Gene Ther ; 16(5): 462-72, 2009 May.
Article in English | MEDLINE | ID: mdl-18989354

ABSTRACT

In vivo electroporation of plasmid DNA (DNA-EP) is an efficient and safe method for vaccines resulting in increased DNA uptake, enhanced protein expression and increased immune responses to the target antigen in a variety of species. To further enhance the efficacy of DNA-EP, we have evaluated the toll-like receptor7 (TLR7) agonist-2, 9, substituted 8-hydroxyadenosine derivative or SM360320--as an adjuvant to vaccines against HER2/neu and CEA in BALB-neuT and CEA transgenic mice (CEA.Tg), respectively. SM360320 induced in vivo secretion of interferon alpha (IFNalpha) and exerted a significant antitumor effect in CEA.Tg mice challenged with a syngenic tumor cell line expressing CEA and an additive effect with a CEA vaccine. Additionally, combination of SM360320 with plasmid encoding the extracellular and transmembrane domain of ratHER2/neu affected the spontaneous tumor progression in BALB-neuT mice treated in an advanced disease setting. The antitumor effect in mice treated with DNA-EP and SM360320 was associated with an anti-CEA and anti-p185(neu) antibody isotype switch from IgG1 to IgG2a. These data demonstrate that SM360320 exerts significant antitumor effects and can act in association with DNA-EP for CEA-positive colon cancer and HER2-positive mammary carcinoma. These observations therefore emphasize the potential of SM360320 as immunological adjuvant for therapeutic DNA vaccines.


Subject(s)
Adenine/analogs & derivatives , Adjuvants, Immunologic/pharmacology , Antineoplastic Agents/pharmacology , Cancer Vaccines/immunology , Toll-Like Receptor 7/agonists , Vaccines, DNA/therapeutic use , Adenine/pharmacokinetics , Adenine/pharmacology , Adjuvants, Immunologic/pharmacokinetics , Administration, Oral , Animals , Antibody Formation/drug effects , Antineoplastic Agents/pharmacokinetics , Cancer Vaccines/pharmacology , Cancer Vaccines/therapeutic use , Disease Models, Animal , Female , Humans , Immunity, Cellular/drug effects , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Toll-Like Receptor 7/immunology , Vaccines, DNA/immunology
3.
Biochim Biophys Acta ; 1782(10): 574-80, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18675343

ABSTRACT

Mucopolysaccharidosis type II (MPSII) is an inherited disorder due to a deficiency of the lysosomal enzyme iduronate-2-sulfatase (IDS). The disease is characterized by a considerable deposition of heparan- and dermatan-sulfate, causing a general impairment of physiological functions. Most of the therapeutic protocols proposed so far are mainly based upon enzyme replacement therapy which is very expensive. There is a requirement for an alternative approach, and to this aim, we evaluated the feasibility of muscle electro gene transfer (EGT) performed in the IDS-knockout (IDS-ko) mouse model. EGT is a highly efficient method of delivering exogenous molecules into different tissues. More recently, pre-treatment with bovine hyaluronidase has shown to further improve transfection efficiency of muscle EGT. We here show that, by applying such procedure, IDS was very efficiently produced inside the muscle. However, no induced IDS activity was measured in the IDS-ko mice plasma, in contrast to matched healthy controls. In the same samples, an anticipated and rapidly increasing immune response against the recombinant protein was observed in the IDS-ko vs control mice, although reaching the same levels at 5 weeks post-injection. Additional experiments performed on healthy mice showed a significant contribution of hyaluronidase pre-treatment in increasing the immune response.


Subject(s)
Genetic Therapy/methods , Glycoproteins/metabolism , Mucopolysaccharidosis II/therapy , Quadriceps Muscle/metabolism , Animals , Antibody Formation/immunology , Cattle , Electric Stimulation/methods , Feasibility Studies , Glycoproteins/genetics , Glycoproteins/immunology , Glycosaminoglycans/metabolism , Glycosaminoglycans/pharmacology , Humans , Hyaluronoglucosaminidase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucopolysaccharidosis II/genetics , Quadriceps Muscle/drug effects , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
4.
Cancer Gene Ther ; 14(2): 201-10, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17053815

ABSTRACT

Cancer vaccines are a promising approach to treating tumors or preventing tumor relapse through induction of an immune response against tumor-associated antigens (TAA). One major obstacle to successful therapy is the immunological tolerance against self-antigens which limits an effective antitumor immune response. As a transient reduction of immunological tolerance may enable more effective vaccination against self-tumor antigens, we explored this hypothesis in a CEA tolerant animal model with an adenovirus expressing CEA vaccine in conjunction with inactivation of CD4(+)CD25(+) regulatory T cells. This vaccination modality resulted in increased CEA-specific CD8(+), CD4(+) T cells and antibody response. The appearance of a CD4(+) T-cell response correlated with a stronger memory response. The combined CD25(+) inactivation and genetic vaccination resulted in significant tumor protection in a metastatic tumor model. Non-invasive tumor visualization showed that not only primary tumors were reduced, but also hepatic metastases. Our results support the viability of this cancer vaccine strategy as an adjuvant treatment to prevent tumor relapse in cancer patients.


Subject(s)
Adenoviridae/genetics , CD4 Antigens/immunology , Cancer Vaccines/immunology , Interleukin-2 Receptor alpha Subunit/immunology , Lymphocyte Activation , T-Lymphocytes/immunology , Animals , Carcinoembryonic Antigen/immunology , Cell Line , Mice , Models, Animal
5.
Scand J Immunol ; 63(1): 35-41, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16398699

ABSTRACT

T-cell tolerance to tumor antigens is a considerable challenge to cancer immunotherapy. The existence of a murine model transgenic for human carcinoembryonic antigen (CEA) allows CEA vaccination efficacy to be studied in a physiologically tolerant context. Immunization of CEA-transgenic mice with an adenoviral vector coding for CEA induced a significant CD8+ T-cell response specific to CEA but failed to induce CEA-specific CD4+ T cells and antibodies. To overcome CD4+ T-cell tolerance, we explored the effect of adjuvants inducing in vivo dendritic cell maturation. Two different Toll-like receptor ligands, monophosphoryl lipid A (MPL) and CpG motif-containing oligodeoxynucleotides (CpG-ODN), were tested. CD4+-mediated IFN-gamma production was induced in the CEA-transgenic mice only when the genetic immunization was performed in the presence of these adjuvants. Moreover, CpG-ODN had a greater effect than MPL in inducing CD4+ T-cell response and enabling anti-CEA antibody production.


Subject(s)
Adenoviridae/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immune Tolerance/immunology , Lipid A/analogs & derivatives , Toll-Like Receptors/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic , Animals , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , CpG Islands , Genetic Vectors/genetics , Humans , Ligands , Lipid A/administration & dosage , Mice , Mice, Transgenic , Oligodeoxyribonucleotides/administration & dosage , Th1 Cells/immunology , Vaccination
6.
Scand J Immunol ; 62(3): 206-17, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16179007

ABSTRACT

Adenovirus vectors encoding carcinoembryonic antigen (Ad-CEA) or costimulatory molecules CD80, intercellular adhesion molecule-1 (ICAM-1) and leucocyte function-associated antigen-3 (LFA-3) (Ad-STIM) were used to transduce murine bone marrow-derived dendritic cells (BMDC). BMDC were characterized for expression of activation markers and for their ability to elicit protective immunity against MC38-CEA tumours in wildtype and CEA-transgenic (CEA-tg) mice. To determine optimal culture conditions, studies were conducted using BMDC cultured in heterologous bovine serum or autologous mouse serum. Transduction of cells grown in presence of heterologous serum increased the expression of costimulatory molecules, major histocompatibility complex class II, of IL-6 and IL-12. Upon vaccination, tumour protection was not specific and was observed also with untransduced cells. Transduced BMDC cultured in the presence of autologous serum showed low expression of the activation markers, did not express IL-6 and had reduced ability to stimulate T-cell proliferation. Nonetheless, CEA-specific CD8+ T-cell response was enhanced upon coinfection of Ad-STIM and Ad-CEA in both mouse strains, although this immune response was not sufficient to protect CEA-tg mice from tumour challenge. These studies support the use of BMDC transduced with Ad vectors encoding tumour antigens for cancer immunotherapy and demonstrate that culture conditions greatly affect the immunological properties of these cells.


Subject(s)
Adenoviridae/genetics , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Dendritic Cells/immunology , Neoplasms/prevention & control , Animals , B7-1 Antigen/genetics , Bone Marrow Cells/immunology , CD58 Antigens/genetics , CD8-Positive T-Lymphocytes/immunology , Cell Culture Techniques , Cells, Cultured , Genetic Vectors , Intercellular Adhesion Molecule-1/genetics , Interleukin-6/metabolism , Mice , Mice, Transgenic , Neoplasms/immunology , Transduction, Genetic
7.
Acta Paediatr Suppl ; 91(439): 100-4, 2002.
Article in English | MEDLINE | ID: mdl-12572851

ABSTRACT

AIMS: Hunter syndrome is a rare X-linked lysosomal storage disorder caused by the deficiency of the housekeeping enzyme iduronate-2-sulphatase (IDS). Deficiency of IDS causes accumulation of undegraded dermatan and heparan-sulphate in various tissues and organs. Approaches have been proposed for the symptomatic therapy of the disease, including bone marrow transplantation and, very recently, enzyme replacement. To date, gene therapy strategies have considered mainly retroviral and adenoviral transduction of the correct cDNA. In this paper, two non-viral somatic gene therapy approaches are proposed: encapsulated heterologous cells and muscle electro-gene transfer (EGT). METHODS: Hunter primary fibroblasts were co-cultured with either cell clones over-expressing the lacking enzyme or with the same incorporated in alginate microcapsules. For EGT, plasmid vector was injected into mouse quadriceps muscle, which was then immediately electro-stimulated. RESULTS: Co-culturing Hunter primary fibroblasts with cells over-expressing IDS resulted in a three- to fourfold increase in fibroblast enzyme activity with respect to control cells. Fibroblast IDS activity was also increased after co-culture with encapsulated cells. EGT was able to transduce genes in mouse muscle, resulting in at least a tenfold increase in IDS activity 1-5 weeks after treatment. CONCLUSION: Although preliminary, results from encapsulated heterologous cell clones and muscle EGT encourage further evaluations for possible application to gene therapy for Hunter syndrome.


Subject(s)
Gene Transfer Techniques , Genetic Therapy , Mucopolysaccharidosis II/genetics , Mucopolysaccharidosis II/therapy , Animals , Clone Cells/transplantation , Coculture Techniques , Disease Models, Animal , Fibroblasts/transplantation , Genetic Vectors/therapeutic use , Mice , Mice, Inbred C57BL , Myoblasts/transplantation , Transplantation, Heterologous
8.
Curr Opin Mol Ther ; 3(5): 464-7, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11699890

ABSTRACT

The use of baculovirus vectors for gene expression in mammalian cells is in continuous expansion. These vectors do not replicate in mammalian cells, do not cause a cytopathic effect upon infection and are able to carry large DNA inserts. Baculovirus vectors have been shown to transduce various cell types in vitro and in vivo with significant efficiency leading to stable gene expression. This review focuses on recent developments with baculovirus that highlight its potential usefor new gene therapy strategies.


Subject(s)
Baculoviridae/genetics , Genetic Therapy/methods , Genetic Vectors , Animals , Central Nervous System/metabolism , Gene Expression , Gene Transfer Techniques , Genetic Therapy/trends , Humans , In Vitro Techniques , Liver/metabolism , Muscle, Skeletal/metabolism , Neoplasms/therapy
9.
J Gene Med ; 3(4): 394-402, 2001.
Article in English | MEDLINE | ID: mdl-11529669

ABSTRACT

BACKGROUND: The hepatitis C virus (HCV) is responsible for a severe and widespread form of hepatitis for which a durable and effective therapy has not yet been established. The only approved therapy against hepatitis C, alpha-interferon protein intramuscular administration, presents numerous drawbacks that might be overcome by adopting a gene therapy approach. HCV exclusively infects humans and chimpanzees, hence an acceptable animal model for hepatitis C pharmacological studies is not available. Recently, tamarins infected by GB virus B (GBV-B) have been proposed as a surrogate animal model for HCV infection. The aim of the present study was the production of tamarin interferon (tIFN) through delivery of tIFN-coding DNA to evaluate the feasibility of a gene therapy approach based on IFN electro-gene transfer (EGT) in future studies with primates. METHODS: Production and biological activity of cloned tamarin interferon was monitored in cultured cells upon transfection and in mice upon muscle EGT of the corresponding plasmid DNA, respectively. RESULTS: A tamarin gene encoding a protein homologous to human interferon-alpha2 (hIFN-alpha2) has been cloned. The tamarin IFN-alpha (tIFN-alpha) protein shows antiviral activity in a cell-based assay. Upon EGT of the corresponding gene in mouse muscles, tIFN-alpha is detectable at high levels in serum for at least 4 months. Most important, activity of tIFN, measured as enhancement of mRNA levels of genes induced by type I IFNs, is also detectable in the liver of EGT-treated mice. CONCLUSION: The present study demonstrates that the delivery of tIFN-alpha DNA via intramuscular injection yields a functional protein able to produce biological effects inside a remote target organ, the liver. This finding, besides the specific purpose of the present study, is of general relevance with a view to establishing therapeutic protocols based on EGT.


Subject(s)
Interferon-alpha/genetics , Liver/physiology , Saguinus/genetics , 5' Untranslated Regions/genetics , Animals , Base Sequence , Cloning, Molecular , DNA Primers , DNA, Complementary/chemistry , DNA, Complementary/genetics , HeLa Cells , Humans , Interferon alpha-2 , Interferon-alpha/pharmacology , L Cells , Liver/drug effects , Mice , Mice, Transgenic , Molecular Sequence Data , Open Reading Frames , Phylogeny , Recombinant Proteins , Saguinus/classification , Sequence Alignment , Sequence Homology, Nucleic Acid , Tumor Cells, Cultured
10.
Hum Gene Ther ; 12(8): 871-81, 2001 May 20.
Article in English | MEDLINE | ID: mdl-11387053

ABSTRACT

Baculovirus vectors are efficient tools for gene transfer into mammalian cells in vitro. However, in vivo gene delivery by systemic administration is hindered by the vector inactivation mediated by the complement system. To characterize further the gene transfer efficacy of baculovirus we examined the vector transduction efficiency in skeletal muscle. Vectors expressing vesicular stomatitis virus glycoprotein (VSV-G) in the viral envelope were generated by inserting the VSV-G coding sequence downstream of the polyhedrin promoter. Two viruses were constructed to carry either the Escherichia coli beta-galactosidase (beta-Gal) gene or the mouse erythropoietin (EPO) cDNA cloned downstream of the cytomegalovirus immediate-early promoter and enhancer. The greater gene transduction efficiency of the Bac-G-betaGal vector was confirmed by comparing the beta-Gal expression level in a variety of human and mouse cell lines with that obtained on infection with Bac-betaGal, a vector that lacks VSV-G. Similarly, a 5- to 10-fold increase in beta-Gal expression between Bac-G-betaGal and Bac-betaGal was observed when mouse myoblasts and myotubes were infected. The same increase in beta-Gal expression was detected on injection of the Bac-G-betaGal vector in the quadriceps of BALB/c and C57BL/6 mice. In contrast, a 2-fold difference in transduction was observed between these two vectors in DBA/2J mouse strain. Last, expression of EPO cDNA was detected for at least 178 days in DBA/2J mice on Bac-G-EPO injection into the quadriceps whereas EPO expression declined to normal values by 35 days postinfection in BALB/c and C57BL/6 mice. Thus, these results indicate that baculovirus may be considered a useful vector for gene transfer in mouse skeletal muscle and that persistence of expression may depend on the mouse strain used.


Subject(s)
Baculoviridae/genetics , Gene Transfer Techniques , Genetic Vectors , Membrane Glycoproteins , Muscle, Skeletal/metabolism , Animals , Blotting, Western , Cell Line , DNA, Complementary/metabolism , Enhancer Elements, Genetic , Erythropoietin/genetics , Escherichia coli/enzymology , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Muscle, Smooth/cytology , Muscle, Smooth/metabolism , Promoter Regions, Genetic , Species Specificity , Time Factors , Transduction, Genetic , Tumor Cells, Cultured , Viral Envelope Proteins/biosynthesis , beta-Galactosidase/genetics
11.
Proc Natl Acad Sci U S A ; 98(11): 5986-91, 2001 May 22.
Article in English | MEDLINE | ID: mdl-11353820

ABSTRACT

Helper-dependent adenoviral vectors deleted of all viral coding sequences have shown an excellent gene expression profile in a variety of animal models, as well as a reduced toxicity after systemic delivery. What is still unclear is whether long-term expression and therapeutic dosages of these vectors can be obtained also in the presence of a preexisting immunity to adenovirus, a condition found in a high proportion of the adult human population. In this study we performed intramuscular delivery of helper-dependent vectors carrying mouse erythropoietin as a marker transgene. We found that low doses of helper-dependent adenoviral vectors can direct long-lasting gene expression in the muscles of fully immunocompetent mice. The best performance-i.e., 100% of treated animals showing sustained expression after 4 months-was achieved with the latest generation helper-dependent backbones, which replicate and package at high efficiency during vector propagation. Moreover, efficient and prolonged transgene expression after intramuscular injection was observed with limited vector load also in animals previously immunized against the same adenovirus serotype. These data suggest that human gene therapy by intramuscular delivery of helper-dependent adenoviral vectors is feasible.


Subject(s)
Adenoviruses, Human/immunology , Genetic Vectors/immunology , Helper Viruses/immunology , Animals , Erythropoietin/genetics , Gene Expression , Gene Transfer Techniques , HeLa Cells , Humans , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
12.
Cancer Gene Ther ; 8(1): 63-72, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11219495

ABSTRACT

Recent studies have shown that gene therapy with type I interferon (IFN) in an adenovirus vector is a powerful tool to suppress the growth of human tumors transplanted in immune-deficient mice. However, in these studies the host immune-mediated effects, which may be important in mediating the long-term control of tumor growth by these cytokines, was not studied. In this paper, we evaluate the antitumor efficacy of different adenoviral vectors containing mouse IFN-alpha genes (i.e., a first-generation replication-defective vector containing IFN-alpha1 and two different second-generation vectors containing IFN-alpha2) in immunocompetent DBA/2 mice transplanted with highly metastatic Friend leukemic cells resistant in vitro to type I IFN. We found that injection of all the different adenovirus vectors containing mouse IFN-alpha( genes resulted in a marked antitumor response in mice transplanted either subcutaneously or intravenously with IFN-resistant Friend leukemic cells compared to tumor-bearing animals inoculated with a control vector. Tumor growth inhibition after injection of IFN-adenovirus vectors was associated with a prolonged presence of high IFN levels in the sera of the injected mice. Suppression of metastatic tumor growth was also observed after a single injection of the IFN--adenovirus recombinant vectors, whereas a comparable antitumor response generally required several injections of high doses of IFN. Altogether, these results demonstrate that IFN--adenoviral vectors can efficiently inhibit metastatic tumor growth by host-mediated mechanisms and suggest that adenovirus-mediated IFN-alpha gene therapy may represent an attractive alternative to the conventional clinical use of this cytokine, which generally requires multiple injections of high IFN doses for a prolonged period of time.


Subject(s)
Adenoviridae/genetics , Interferon-alpha/genetics , Leukemia, Experimental/therapy , Animals , Friend murine leukemia virus , Genetic Therapy , Genetic Vectors , Injections, Intraperitoneal , Injections, Intravenous , Interferon-alpha/blood , Interferon-alpha/metabolism , Lac Operon/physiology , Leukemia, Experimental/immunology , Liver Neoplasms, Experimental/pathology , Liver Neoplasms, Experimental/prevention & control , Male , Mice , Mice, Inbred DBA , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Survival Analysis , Transfection , Tumor Cells, Cultured
13.
Gene Ther ; 8(24): 1817-25, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11821934

ABSTRACT

A major goal in gene therapy is to develop efficient gene transfer protocols that allow tissue-specific, long-term and tightly regulated expression of the desired transgene. This objective is becoming more attainable through the co-evolution of gene transfer vectors and regulation systems. The ideal vector should efficiently transduce non-dividing cells with minimal toxicity, thus endowing the system with persistent transgene expression. The helper-dependent adenovirus vectors meet these requirements, as demonstrated in various studies in the literature. The most promising regulation system is the tet-on system, which has low basal transcriptional activity and high inducibility. To explore the regulated transgene expression in the context of a helper-dependent vector, we constructed the HD-TET-IFN vector, containing the mIFN(alpha) gene under the control of the tetracycline inducible transactivator rtTA2(s)-S2. Mice injected with HD-TET-IFN showed high levels of serum mIFN(alpha) only upon transcriptional activation. The transgene expression was reinducible to the same high level up to 3 months p.i., and the amount of expressed cytokine could be regulated by dosing doxycycline. Transcriptional activation of mIFN(alpha) induced by doxycycline resulted in prolonged survival and reduced liver damage in HD-TET-IFN-injected mice challenged with a lethal dose of coronavirus. Activation of antiviral genes mediated by doxycycline-dependent mIFN(alpha) expression was also observed at low HD-TET-IFN doses. The possibility of controlling gene expression by the combination of HD vectors and the latest tet-on transactivator also holds promise for studying gene function in other animal models.


Subject(s)
Adenoviridae/genetics , Anti-Bacterial Agents/therapeutic use , Doxycycline/therapeutic use , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Interferon-alpha/genetics , Alanine Transaminase/blood , Animals , Carcinoma, Hepatocellular , Cell Line , Enzyme-Linked Immunosorbent Assay/methods , Female , Gene Expression , Gene Expression Regulation , Hepatitis, Viral, Animal/therapy , Humans , Interferon-alpha/analysis , Interferon-alpha/blood , Liver/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , RNA, Messenger/analysis , Trans-Activators/genetics , Transgenes
14.
J Virol ; 74(24): 11598-607, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11090158

ABSTRACT

We describe an improved genetic immunization strategy for eliciting a full spectrum of anti-hepatitis C virus (HCV) envelope 2 (E2) glycoprotein responses in mammals through electrical gene transfer (EGT) of plasmid DNA into muscle fibers. Intramuscular injection of a plasmid encoding a cross-reactive hypervariable region 1 (HVR1) peptide mimic fused at the N terminus of the E2 ectodomain, followed by electrical stimulation treatment in the form of high-frequency, low-voltage electric pulses, induced more than 10-fold-higher expression levels in the transfected mouse tissue. As a result of this substantial increment of in vivo antigen production, the humoral response induced in mice, rats, and rabbits ranged from 10- to 30-fold higher than that induced by conventional naked DNA immunization. Consequently, immune sera from EGT-treated mice displayed a broader cross-reactivity against HVR1 variants from natural isolates than sera from injected animals that were not subjected to electrical stimulation. Cellular response against E2 epitopes specific for helper and cytotoxic T cells was significantly improved by EGT. The EGT-mediated enhancement of humoral and cellular immunity is antigen independent, since comparable increases in antibody response against ciliary neurotrophic factor or in specific anti-human immunodeficiency virus type 1 gag CD8(+) T cells were obtained in rats and mice. Thus, the method described potentially provides a safe, low-cost treatment that may be scaled up to humans and may hold the key for future development of prophylactic or therapeutic vaccines against HCV and other infectious diseases.


Subject(s)
Adenovirus E2 Proteins/immunology , DNA, Viral/immunology , Hepacivirus/immunology , Hepatitis C/immunology , Viral Hepatitis Vaccines/immunology , Adenovirus E2 Proteins/genetics , Animals , B-Lymphocytes/immunology , B-Lymphocytes/virology , DNA, Viral/genetics , Electroporation , Hepatitis C/prevention & control , Immunity , Mice , Rabbits , Rats , T-Lymphocytes/immunology , T-Lymphocytes/virology , Transfection , Vaccines, DNA/immunology
15.
Hum Gene Ther ; 11(13): 1891-900, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10986561

ABSTRACT

We have investigated the efficacy of a gene transfer strategy based on plasmid DNA electroinjection for the correction of anemia associated with renal failure. An expression plasmid encoding the rat erythropoietin (EPO) cDNA under the control of the CMV promoter as constructed and utilized for this work. Electroinjection of pCMV/rEPO in different rat muscles yielded sustained and long-term EPO production and secretion. The muscle-produced EPO corrected the anemia in five of six nephrectomized rats, used as a model of renal failure. The efficiency of muscle transduction was comparable in rats and mice injected with equivalent amounts of DNA per kilogram of body weight. These results demonstrate that gene electrotransfer can be applied to produce therapeutically significant levels of erythropoietin in chronic renal failure.


Subject(s)
Anemia/therapy , Erythropoietin/genetics , Gene Transfer Techniques , Kidney Failure, Chronic/complications , Muscle, Skeletal/physiology , Anemia/etiology , Animals , Cytomegalovirus/genetics , Disease Models, Animal , Erythropoietin/metabolism , Erythropoietin/pharmacology , Genetic Therapy/methods , Hematocrit , Injections/methods , Mice , Mice, Inbred BALB C , Nephrectomy , Plasmids/pharmacology , Promoter Regions, Genetic , Rabbits , Rats , Rats, Sprague-Dawley , Transduction, Genetic
16.
J Virol ; 74(10): 4816-23, 2000 May.
Article in English | MEDLINE | ID: mdl-10775620

ABSTRACT

The current therapy for hepatitis B and C is based on systemic administration of recombinant human alpha interferon (r-hIFN-alpha). However, systemic delivery of r-hIFN-alpha is associated with severe side effects, but more importantly, it is effective in only a small percentage of patients. In an effort to maximize IFN-alpha antiviral efficacy, we have explored the therapeutic potential of murine IFN-alpha2 (mIFNalpha2) selectively expressed in the liver. To this end, we have developed a helper-dependent adenovirus vector (HD) containing the mIFN-alpha2 gene under the control of the liver-specific transthyretin promoter (HD-IFN). Comparison with a first-generation adenovirus carrying the same mIFN-alpha2 expression cassette indicates that at certain HD-IFN doses, induction of antiviral genes can be achieved in the absence of detectable circulating mIFN-alpha2. Challenge of injected mice with mouse hepatitis virus type 3 showed that HD-IFN provides high liver protection. Moreover, liver protection was also observed in acute nonviral liver inflammation hepatitis induced by concanavalin A at 1 month postinfection. These results hold promise for the development of a gene therapy treatment for chronic viral hepatitis based on liver-restricted expression of IFN-alpha2.


Subject(s)
Chemical and Drug Induced Liver Injury/therapy , Hepatitis, Viral, Animal/therapy , Interferon-alpha/genetics , Interferon-alpha/metabolism , Liver/metabolism , Adenoviridae/genetics , Adenoviridae/immunology , Animals , Chemical and Drug Induced Liver Injury/prevention & control , Concanavalin A/pharmacology , Female , Gene Expression , Genetic Therapy , Genetic Vectors , Helper Viruses/immunology , Hepatitis, Viral, Animal/prevention & control , Hepatitis, Viral, Animal/virology , Humans , Mice , Mice, Inbred C57BL , Murine hepatitis virus/pathogenicity
17.
Hum Gene Ther ; 11(6): 859-68, 2000 Apr 10.
Article in English | MEDLINE | ID: mdl-10779163

ABSTRACT

Helper-dependent (HD) adenoviral (Ad) vectors, in which all viral coding sequences are deleted, have been generated. We show here that intravenous delivery of a mouse EPO (mEPO) expression cassette cloned in an HD vector in immunocompetent mice is effective and long lasting, but not permanent. A precise dose-response relationship between the dose of injected virus and stable EPO serum levels was observed, together with a 100-fold increase in gene expression per infectious particle when compared with a first-generation Ad vector bearing the same cassette. As a direct consequence, therapeutic increases in hematocrit that lasted more than 6 months were achieved with minute amounts of virus, which caused no detectable production of neutralizing antibodies. Intravenous readministration of the HD-mEPO vector in the same mice was as effective as in naive animals without any need for prior immunosuppression. Finally, HD-mEPO injection in subtotally nephrectomized rats improved the anemic status induced by surgery. HD Ad vectors are thus excellent tools for EPO gene therapy.


Subject(s)
Adenoviridae/genetics , Erythropoietin/genetics , Gene Transfer Techniques , Sequence Deletion , Animals , Antibody Formation , Erythropoietin/immunology , Erythropoietin/metabolism , Female , Genetic Vectors , Hematocrit , Injections, Intravenous , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Nephrectomy , Rats , Rats, Sprague-Dawley , Species Specificity , Time Factors
18.
Proc Natl Acad Sci U S A ; 96(11): 6417-22, 1999 May 25.
Article in English | MEDLINE | ID: mdl-10339602

ABSTRACT

We show that an electric treatment in the form of high-frequency, low-voltage electric pulses can increase more than 100-fold the production and secretion of a recombinant protein from mouse skeletal muscle. Therapeutical erythopoietin (EPO) levels were achieved in mice with a single injection of as little as 1 microgram of plasmid DNA, and the increase in hematocrit after EPO production was stable and long-lasting. Pharmacological regulation through a tetracycline-inducible promoter allowed regulation of serum EPO and hematocrit levels. Tissue damage after stimulation was transient. The method described thus provides a potentially safe and low-cost treatment for serum protein deficiencies.


Subject(s)
Erythropoietin/genetics , Gene Transfer Techniques , Muscle, Skeletal/physiology , 5' Untranslated Regions/genetics , Animals , Cytomegalovirus/genetics , Electric Stimulation , Electroporation/methods , Erythropoietin/biosynthesis , Erythropoietin/blood , Female , Gene Expression Regulation , Kinetics , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Muscle, Skeletal/cytology , Plasmids , Promoter Regions, Genetic , Recombinant Proteins/biosynthesis , Sensitivity and Specificity , Time Factors
19.
Proc Natl Acad Sci U S A ; 96(6): 2615-20, 1999 Mar 16.
Article in English | MEDLINE | ID: mdl-10077559

ABSTRACT

Adenovirus (Ad) and adeno-associated virus (AAV) have attractive and complementary properties that can be exploited for gene transfer purposes. Ad vectors are probably the most efficient vehicles to deliver foreign genes both in vitro and in vivo. AAV exhibits the unique ability to establish latency by efficiently integrating at a specific locus of human chromosome 19 (AAVS1). Two viral elements are necessary for the integration at AAVS1: Rep68/78 and the inverted terminal repeats (AAV-ITRs). In this study, we report the development of two helper-dependent adenoviral (HD) vectors, one carrying the Rep78 gene, the other an AAV-ITR-flanked transgene. Although Rep proteins have been demonstrated to interfere with Ad replication, HD Rep78 vector was successfully amplified on serial passages in 293CRE4 cells with a yield of 50-100 transducing units per cell. DNA integration at the AAVS1 site also was demonstrated in hepatoma cells coinfected with the HD-expressing Rep78 and with the second HD vector carrying a transgene flanked by AAV-ITRs. The high transduction efficiency, large cloning capacity, and high titer of the HD, combined with the site-specific integration machinery provided by AAV-derived components, make the Ad/AAV hybrid viruses a promising vehicle for gene therapy.


Subject(s)
Adenoviridae , DNA-Binding Proteins , Dependovirus , Gene Transfer Techniques , Genetic Vectors , Base Sequence , DNA Helicases/genetics , HeLa Cells , Humans , In Situ Hybridization , Molecular Sequence Data , Reassortant Viruses , Trans-Activators/genetics
20.
Virology ; 255(2): 302-11, 1999 Mar 15.
Article in English | MEDLINE | ID: mdl-10069956

ABSTRACT

Although great progress has been made in the characterization of the biochemical and biological features of hepatitis C virus (HCV) gene expression, the elucidation of the HCV life cycle and the evaluation of novel antiviral strategies have been hindered by the lack of a suitable cell culture system. In this context, the development of an efficient HCV cDNA delivery method would contribute to the understanding of HCV replication. To assess the functionality of baculovirus mediated gene delivery for HCV expression, we have constructed recombinant baculoviruses encoding HCV cDNA under the control of the cytomegalovirus promoter. Transduction of the human hepatoma cell line Huh-7 with Bac-HCV vectors was efficient and HCV cDNA expression was enhanced by treatment of the infected cells with dexamethasone. HCV structural and nonstructural polypeptides were processed correctly and were found to localize in the cytoplasm in a pattern characteristic of the endoplasmic reticulum. The expression of the HCV proteins was detected for 49 days after infection. Thus, these results indicate that the recombinant Bac-HCV vectors are a useful tool for the delivery of HCV cDNA and can facilitate the analysis of structural and functional properties of the HCV proteins. In addition, the Bac-HCV vectors can provide important information on the evaluation of novel anti-HCV antiviral strategies.


Subject(s)
Baculoviridae , DNA, Viral , Genetic Vectors , Hepacivirus/genetics , Baculoviridae/genetics , Carcinoma, Hepatocellular , Cell Transformation, Viral , DNA, Complementary , Gene Expression , Genetic Vectors/genetics , Hepacivirus/immunology , Humans , Recombination, Genetic , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...