Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
2.
Cell Transplant ; 9(5): 585-94, 2000.
Article in English | MEDLINE | ID: mdl-11144956

ABSTRACT

Adeno-associated virus (AAV)-based vectors are being tested in animal models as viable treatments for glioma and neurodegenerative disease and could potentially be employed to target a variety of central nervous system disorders. The relationship between dose of injected vector and its resulting distribution in brain tissue has not been previously reported nor has the most efficient method of delivery been determined. Here we report that convection-enhanced delivery (CED) of 2.5 x 10(8), 2.5 x 10(9), or 2.5 x 10(10) particles of AAV-thymidine kinase (AAV-TK) into rat brain revealed a clear dose response. In the high-dose group, a volume of 300 mm3 of brain tissue was partially transduced. Results showed that infusion pump and subcutaneous osmotic pumps were both capable of delivering vector via CED and that total particle number was the most important determining factor in obtaining efficient expression. Results further showed differences in histopathology between the delivery groups. While administration of vector using infusion pump had relatively benign effects, the use of osmotic pumps resulted in notable toxicity to the surrounding brain tissue. To determine tissue distribution of vector following intracranial delivery, PCR analysis was performed on tissues from rats that received high doses of AAV-TK. Three weeks following CED, vector could be detected in both hemispheres of the brain, spinal cord, spleen, and kidney.


Subject(s)
Brain , Convection , Dependovirus/genetics , Thymidine Kinase/genetics , Transgenes , Animals , Brain/metabolism , Brain/pathology , Female , Gene Expression , Gene Targeting , Genetic Vectors/toxicity , Image Processing, Computer-Assisted , Immunoenzyme Techniques , Infusion Pumps , Necrosis , Osmotic Pressure , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Thymidine Kinase/immunology , Thymidine Kinase/metabolism , Transduction, Genetic
3.
Clin Immunol ; 92(1): 67-75, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10413654

ABSTRACT

Recombinant adeno-associated virus (rAAV) is a replication-defective parvovirus which is being explored as a vector for gene therapy because of its broad host range, excellent safety profile, and durable transgene expression in infected hosts. rAAV has also been reported by several groups to induce little or no immune response to its encoded transgene products. In this study we examined the immunogenicity of rAAV by studying the immune response of C57BL/6 mice to a single dose of rAAV-encoding ovalbumin (AAV-Ova) administered by a variety of routes. Mice injected with AAV-Ova intraperitoneally (ip), intravenously, or subcutaneously developed potent ovalbumin-specific cytotoxic T lymphocytes (CTL) as well as anti-ovalbumin antibodies and antibodies to AAV. In contrast, mice injected with AAV-Ova intramuscularly developed a humoral response to the virus and the transgene but minimal ovalbumin-specific CTLs. The induced CTL response after ip administration of AAV-Ova protected mice against a subsequent tumor challenge with an ovalbumin-transfected B16 melanoma cell line. Studies of the mechanism by which AAV-Ova induces CTL confirmed that the virus delivers the transgene product into the classical MHC class I pathway of antigen processing. Mice that previously had been exposed to rAAV vectors failed to develop ovalbumin-specific CTL following administration of AAV-Ova. Analysis of these mice revealed the presence of circulating anti-AAV antibodies that blocked rAAV transduction in vitro and inhibited CTL induction in vivo. These results suggest a possible role for rAAV in the immunotherapy of malignancies and viral infections, although induced antibody responses to AAV may limit its ability to be administered for repeated vaccinations.


Subject(s)
Antigens, Viral/administration & dosage , Dependovirus/immunology , Animals , Antibody Formation , Drug Administration Routes , Female , Immunity, Cellular/immunology , Injections, Intramuscular , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Recombinant Proteins/administration & dosage
4.
Nat Med ; 5(1): 56-63, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9883840

ABSTRACT

Hemophilia B is a severe X-linked bleeding diathesis caused by the absence of functional blood coagulation factor IX, and is an excellent candidate for treatment of a genetic disease by gene therapy. Using an adeno-associated viral vector, we demonstrate sustained expression (>17 months) of factor IX in a large-animal model at levels that would have a therapeutic effect in humans (up to 70 ng/ml, adequate to achieve phenotypic correction, in an animal injected with 8.5x10(12) vector particles/kg). The five hemophilia B dogs treated showed stable, vector dose-dependent partial correction of the whole blood clotting time and, at higher doses, of the activated partial thromboplastin time. In contrast to other viral gene delivery systems, this minimally invasive procedure, consisting of a series of percutaneous intramuscular injections at a single timepoint, was not associated with local or systemic toxicity. Efficient gene transfer to muscle was shown by immunofluorescence staining and DNA analysis of biopsied tissue. Immune responses against factor IX were either absent or transient. These data provide strong support for the feasibility of the approach for therapy of human subjects.


Subject(s)
Dependovirus , Factor IX/genetics , Gene Transfer Techniques , Genetic Vectors , Hemophilia B/therapy , Animals , DNA, Viral/analysis , Dependovirus/genetics , Disease Models, Animal , Dogs , Factor IX/immunology , Gene Expression , Hemophilia B/immunology , Humans , Injections, Intramuscular , Male , Time Factors , Tumor Cells, Cultured
5.
Gene Ther ; 5(7): 938-45, 1998 Jul.
Article in English | MEDLINE | ID: mdl-9813665

ABSTRACT

The purpose of this work was to develop an efficient method for the production of adeno-associated virus (AAV) vectors in the absence of helper virus. The adenovirus regions that mediate AAV vector replication were identified and assembled into a helper plasmid. These included the VA, E2A and E4 regions. When this helper plasmid was cotransfected into 293 cells, along with plasmids encoding the AAV vector, and rep and cap genes, AAV vector was produced as efficiently as when using adenovirus infection as a source of help. CMV-driven constructs expressing the E4orf6 and the 72-M(r), E2A proteins were able to functionally replace the E4 and E2A regions, respectively. Therefore the minimum set of genes required to produce AAV helper activity equivalent to that provided by adenovirus infection consists of, or is a subset of, the following genes: the E4orf6 gene, the 72-M(r), E2A protein gene, the VA RNA genes and the E1 region. AAV vector preparations made with adenovirus and by the helper virus-free method were essentially indistinguishable with respect to particle density, particle to infectivity ratio, capsimer ratio and efficiency of muscle transduction in vivo. Only AAV vector preparations made by the helper virus-free method were not reactive with anti-adenovirus sera.


Subject(s)
Dependovirus , Genetic Engineering , Genetic Vectors , Adenoviridae , Animals , Female , Helper Viruses , Mice , Mice, Inbred BALB C , Plasmids
6.
Gene Ther ; 5(12): 1642-9, 1998 Dec.
Article in English | MEDLINE | ID: mdl-10023443

ABSTRACT

Mucopolysaccharidosis type VII (MPS VII) is a lysosomal storage disease caused by a genetic deficiency of beta-glucuronidase (GUS). We used a recombinant adeno-associated virus vector (AAV-GUS) to deliver GUS cDNA to MPS VII mice. The route of vector administration had a dramatic effect on the extent and distribution of GUS activity. Intramuscular injection of AAV-GUS resulted in high, localized production of GUS, while intravenous administration produced low GUS activity in several tissues. This latter treatment of MPS VII mice reduced glycosaminoglycan levels in the liver to normal and reduced storage granules dramatically. We show that a single administration of AAV-GUS can provide sustained expression of GUS in a variety of cell types and is sufficient to reverse the disease phenotype at least in the liver.


Subject(s)
Dependovirus , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Glucuronidase/genetics , Mucopolysaccharidosis VII/therapy , Animals , Genetic Vectors/immunology , Glucuronidase/biosynthesis , Glucuronidase/immunology , Injections, Intramuscular , Injections, Intravenous , Liver/enzymology , Mice , Mice, Mutant Strains , Mucopolysaccharidosis VII/enzymology , Mucopolysaccharidosis VII/immunology
7.
J Virol ; 71(3): 1776-83, 1997 Mar.
Article in English | MEDLINE | ID: mdl-9032306

ABSTRACT

We tested the ability of a recombinant adeno-associated virus (rAAV) vector to express and integrate exogenous DNA into human hematopoietic cells in the absence of selection. We developed an rAAV vector, AAV-tNGFR, carrying a truncated rat nerve growth factor receptor (tNGFR) cDNA as a cell surface reporter under the control of the Moloney murine leukemia virus (MoMuLV) long terminal repeat. An analogous MoMuLV-based retroviral vector (L-tNGFR) was used in parallel, and gene transfer and expression in human hematopoietic cells were assessed by flow cytometry and DNA analyses. Following gene transfer into K562 cells with AAV-tNGFR at a multiplicity of infection (MOI) of 13 infectious units (IU), 26 to 38% of cells expressed tNGFR on the surface early after transduction, but the proportion of tNGFR expressing cells steadily declined to 3.0 to 3.5% over 1 month of culture. At an MOI of 130 IU, nearly all cells expressed tNGFR immediately posttransduction, but the proportion of cells expressing tNGFR declined to 62% over 2 months of culture. The decline in the proportion of AAV-tNGFR-expressing cells was associated with ongoing losses of vector genomes. In contrast, K562 cells transduced with the retroviral vector L-tNGFR expressed tNGFR in a constant fraction. Integration analyses on clones showed that integration occurred at different sites. Integration frequencies were estimated at about 49% at an MOI of 130 and 2% at an MOI of 1.3. Transduction of primary human CD34+ progenitor cells by AAV-tNGFR was less efficient than with K562 cells and showed a declining percentage of cells expressing tNGFR over 2 weeks of culture. Thus, purified rAAV caused very high gene transfer and expression in human hematopoietic cells early after transduction, which steadily declined during cell passage in the absence of selection. Although the efficiency of integration was low, overall integration was markedly improved at a high MOI. While prolonged episomal persistence may be adequate for gene therapy of nondividing cells, a very high MOI or improvements in basic aspects of AAV-based vectors may be necessary to improve integration frequency in the rapidly dividing hematopoietic cell population.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors , Receptors, Nerve Growth Factor/genetics , Virus Integration , Animals , Antigens, CD34 , Cell Line , Cells, Cultured , DNA/analysis , Gene Expression , Genes, Reporter , Genome, Viral , Humans , Rats , Recombination, Genetic , Selection, Genetic , Tumor Cells, Cultured
8.
Proc Natl Acad Sci U S A ; 93(24): 14082-7, 1996 Nov 26.
Article in English | MEDLINE | ID: mdl-8943064

ABSTRACT

Somatic gene therapy has been proposed as a means to achieve systemic delivery of therapeutic proteins. However, there is limited evidence that current methods of gene delivery can practically achieve this goal. In this study, we demonstrate that, following a single intramuscular administration of a recombinant adeno-associated virus (rAAV) vector containing the beta-galactosidase (AAV-lacZ) gene into adult BALB/c mice, protein expression was detected in myofibers for at least 32 weeks. A single intramuscular administration of an AAV vector containing a gene for human erythropoietin (AAV-Epo) into mice resulted in dose-dependent secretion of erythropoietin and corresponding increases in red blood cell production that persisted for up to 40 weeks. Primary human myotubes transduced in vitro with the AAV-Epo vector also showed dose-dependent production of Epo. These results demonstrate that rAAV vectors are able to transduce skeletal muscle and are capable of achieving sustained expression and systemic delivery of a therapeutic protein following a single intramuscular administration. Gene therapy using AAV vectors may provide a practical strategy for the treatment of inherited and acquired protein deficiencies.


Subject(s)
Erythropoiesis , Erythropoietin/biosynthesis , Gene Transfer Techniques , Genetic Therapy/methods , Muscle, Skeletal/physiology , beta-Galactosidase/biosynthesis , Animals , Cell Line , Dependovirus , Erythropoietin/genetics , Escherichia coli , Gene Expression , Genetic Vectors , Histocytochemistry , Humans , Injections, Intramuscular , Lac Operon , Mice , Mice, Inbred BALB C , Time Factors , beta-Galactosidase/genetics
9.
Blood ; 88(2): 492-504, 1996 Jul 15.
Article in English | MEDLINE | ID: mdl-8695797

ABSTRACT

Gene transfer vectors based on adeno-associated virus (AAV) appear promising because of their high transduction frequencies regardless of cell cycle status and ability to integrate into chromosomal DNA. We tested AAV-mediated gene transfer into a panel of human bone marrow or umbilical cord-derived CD34+ hematopoietic progenitor cells, using vectors encoding several transgenes under the control of viral and cellular promoters. Gene transfer was evaluated by (1) chromosomal integration of vector sequences and (2) analysis of transgene expression. Southern hybridization and fluorescence in situ hybridization analysis of transduced CD34 genomic DNA showed the presence of integrated vector sequences in chromosomal DNA in a portion of transduced cells and showed that integrated vector sequences were replicated along with cellular DNA during mitosis. Transgene expression in transduced CD34 cells in suspension cultures and in myeloid colonies differentiating in vitro from transduced CD34 cells approximated that predicted by the multiplicity of transduction. This was true in CD34 cells from different donors, regardless of the transgene or selective pressure. Comparisons of CD34 cell transduction either before or after cytokine stimulation showed similar gene transfer frequencies. Our findings suggest that AAV transduction of CD34+ hematopoietic progenitor cells is efficient, can lead to stable integration in a population of transduced cells, and may therefore provide the basis for safe and efficient ex vivo gene therapy of the hematopoietic system.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Hematopoietic Stem Cells/virology , Virus Integration , Amino Acid Sequence , Antigens, CD34/analysis , Bone Marrow Cells , Cell Cycle , Chromosome Mapping , DNA Replication , Fetal Blood/cytology , Hematopoietic Stem Cells/metabolism , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Recombinant Fusion Proteins/biosynthesis , Transfection
11.
J Virol ; 68(9): 5656-66, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8057446

ABSTRACT

Gene transfer vectors based on adeno-associated virus (AAV) are emerging as highly promising for use in human gene therapy by virtue of their characteristics of wide host range, high transduction efficiencies, and lack of cytopathogenicity. To better define the biology of AAV-mediated gene transfer, we tested the ability of an AAV vector to efficiently introduce transgenes into nonproliferating cell populations. Cells were induced into a nonproliferative state by treatment with the DNA synthesis inhibitors fluorodeoxyuridine and aphidicolin or by contact inhibition induced by confluence and serum starvation. Cells in logarithmic growth or DNA synthesis arrest were transduced with vCWR:beta gal, an AAV-based vector encoding beta-galactosidase under Rous sarcoma virus long terminal repeat promoter control. Under each condition tested, vCWR:beta Gal expression in nondividing cells was at least equivalent to that in actively proliferating cells, suggesting that mechanisms for virus attachment, nuclear transport, virion uncoating, and perhaps some limited second-strand synthesis of AAV vectors were present in nondividing cells. Southern hybridization analysis of vector sequences from cells transduced while in DNA synthetic arrest and expanded after release of the block confirmed ultimate integration of the vector genome into cellular chromosomal DNA. These findings may provide the basis for the use of AAV-based vectors for gene transfer into quiescent cell populations such as totipotent hematopoietic stem cells.


Subject(s)
Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors , Aphidicolin/pharmacology , Base Sequence , Cell Division , Cell Line , Contact Inhibition , DNA Primers/chemistry , Genetic Therapy , Humans , In Vitro Techniques , Molecular Sequence Data , Transduction, Genetic , Virus Integration
12.
Adv Exp Med Biol ; 312: 139-49, 1992.
Article in English | MEDLINE | ID: mdl-1325101

ABSTRACT

We have targeted mRNA sequences encompassing the translation initiation codon of the essential herpes simplex virus type 1 (HSV-1) IE3 gene with three kinds of anti-sense molecule. Addition of a 15mer oligodeoxyribonucleoside methylphosphonate to tissue culture cells resulted in suppression of viral replication. HSV-1 replication was also inhibited in cultured cells containing anti-sense vectors expressing transcripts complementary to the IE3 mRNA. We have also constructed a ribozyme which upon base pairing with the target IE3 mRNA induces cleavage at the predicted GUC site. A major obstacle to anti-sense studies in animals is drug delivery of preformed antisense molecules to ganglionic neurons, the site of HSV latency and reactivation. We speculate as to how this may be accomplished through carrier compounds which are taken up by nerve terminals and transported by retrograde axoplasmic flow. By the same route, HSV itself may be used as an anti-sense vector.


Subject(s)
Antiviral Agents/pharmacology , DNA, Antisense/pharmacology , RNA, Antisense/pharmacology , Simplexvirus/drug effects , Animals , Base Sequence , Humans , Molecular Sequence Data
SELECTION OF CITATIONS
SEARCH DETAIL
...