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










Database
Type of study
Language
Publication year range
1.
Mol Ther Methods Clin Dev ; 2: 15017, 2015.
Article in English | MEDLINE | ID: mdl-26029728

ABSTRACT

It is a current regulatory requirement to demonstrate absence of detectable replication-competent lentivirus (RCL) in lentiviral vector products prior to use in clinical trials. Immune Design previously described an HIV-1-based integration-deficient lentiviral vector for use in cancer immunotherapy (VP02). VP02 is enveloped with E1001, a modified Sindbis virus glycoprotein which targets dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) expressed on dendritic cells in vivo. Vector enveloped with E1001 does not transduce T-cell lines used in standard HIV-1-based RCL assays, making current RCL testing formats unsuitable for testing VP02. We therefore developed a novel assay to test for RCL in clinical lots of VP02. This assay, which utilizes a murine leukemia positive control virus and a 293F cell line expressing the E1001 receptor DC-SIGN, meets a series of evaluation criteria defined in collaboration with US regulatory authorities and demonstrates the ability of the assay format to amplify and detect a hypothetical RCL derived from VP02 vector components. This assay was qualified and used to test six independent GMP production lots of VP02, in which no RCL was detected. We propose that the evaluation criteria used to rationally design this novel method should be considered when developing an RCL assay for any lentiviral vector.

2.
Hum Gene Ther Methods ; 23(5): 309-23, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23121195

ABSTRACT

The release of lentiviral vectors for clinical use requires the testing of vector material, production cells, and, if applicable, ex vivo-transduced cells for the presence of replication-competent lentivirus (RCL). Vectors derived from the nonprimate lentivirus equine infectious anemia virus (EIAV) have been directly administered to patients in several clinical trials, with no toxicity observed to date. Because EIAV does not replicate in human cells, and because putative RCLs derived from vector components within human vector production cells would most likely be human cell-tropic, we previously developed an RCL assay using amphotropic murine leukemia virus (MLV) as a surrogate positive control and human cells as RCL amplification/indicator cells. Here we report an additional RCL assay that tests for the presence of theoretical "equine-tropic" RCLs. This approach provides further assurance of safety by detecting putative RCLs with an equine cell-specific tropism that might not be efficiently amplified by the human cell-based RCL assay. We tested the ability of accessory gene-deficient EIAV mutant viruses to replicate in a highly permissive equine cell line to direct our choice of a suitable EIAV-derived positive control. In addition, we report for the first time the mathematical rationale for use of the Poisson distribution to calculate minimal infectious dose of positive control virus and for use in monitoring assay positive/spike control failures in accumulating data sets. No RCLs have been detected in Good Manufacturing Practice (GMP)-compliant RCL assays to date, further demonstrating that RCL formation is highly unlikely in contemporary minimal lentiviral vector systems.


Subject(s)
Genetic Vectors/genetics , Infectious Anemia Virus, Equine/genetics , Viral Tropism , Virus Replication , Animals , Biological Assay , Cell Line , Gene Order , Horses , Humans , Infectious Anemia Virus, Equine/physiology , Leukemia Virus, Murine , Mice , Reproducibility of Results , Transduction, Genetic
3.
Sci Transl Med ; 1(2): 2ra4, 2009 Oct 14.
Article in English | MEDLINE | ID: mdl-20368163

ABSTRACT

In Parkinson's disease, degeneration of specific neurons in the midbrain can cause severe motor deficits, including tremors and the inability to initiate movement. The standard treatment is administration of pharmacological agents that transiently increase concentrations of brain dopamine and thereby discontinuously modulate neuronal activity in the striatum, the primary target of dopaminergic neurons. The resulting intermittent dopamine alleviates parkinsonian symptoms but is also thought to cause abnormal involuntary movements, called dyskinesias. To investigate gene therapy for Parkinson's disease, we simulated the disease in macaque monkeys by treating them with the complex I mitochondrial inhibitor 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine, which induces selective degeneration of dopamine-producing neurons. In this model, we demonstrated that injection of a tricistronic lentiviral vector encoding the critical genes for dopamine synthesis (tyrosine hydroxylase, aromatic L-amino acid decarboxylase, and guanosine 5'-triphosphate cyclohydrolase 1) into the striatum safely restored extracellular concentrations of dopamine and corrected the motor deficits for 12 months without associated dyskinesias. Gene therapy-mediated dopamine replacement may be able to correct Parkinsonism in patients without the complications of dyskinesias.


Subject(s)
Disease Models, Animal , Dopamine/genetics , Genetic Therapy , Parkinson Disease/therapy , Animals , Dopamine/deficiency , Dyskinesias/complications , Genetic Vectors , Lentivirus/genetics , Macaca mulatta , Motor Activity/genetics , Parkinson Disease/complications
4.
J Gene Med ; 9(5): 345-56, 2007 May.
Article in English | MEDLINE | ID: mdl-17366519

ABSTRACT

Pseudotyping viral vectors with vesicular stomatitis virus glycoprotein (VSV-G) enables the transduction of an extensive range of cell types from different species. We have discovered two important parameters of the VSV-G-pseudotyping phenomenon that relate directly to the transduction potential of lentiviral vectors: (1) the glycosylation status of VSV-G, and (2) the quantity of glycoprotein associated with virions. We measured production-cell and virion-associated quantities of two isoform variants of VSV-G, which differ in their glycosylation status, VSV-G1 and VSV-G2, and assessed the impact of this difference on the efficiency of mammalian cell transduction by lentiviral vectors. The glycosylation of VSV-G at N336 allowed greater maximal expression of VSV-G in HEK293T cells, thus facilitating vector pseudotyping. The transduction of primate cell lines was substantially affected (up to 50-fold) by the degree of VSV-G1 or VSV-G2 incorporation, whereas other cell lines, such as D17 (canine), were less sensitive to virion-associated VSV-G1/2 quantities. These data indicate that the minimum required concentration of virion-associated VSV-G differs substantially between cell species/types. The implications of these data with regard to VSV-G-pseudotyped vector production, titration, and use in host-cell restriction studies, are discussed.


Subject(s)
Genetic Vectors , Lentivirus/genetics , Membrane Glycoproteins/genetics , Transduction, Genetic , Vesicular stomatitis Indiana virus/genetics , Viral Envelope Proteins/genetics , Animals , Cell Line , Glycosylation , Humans , Membrane Glycoproteins/analysis , Membrane Glycoproteins/chemistry , Protein Isoforms/analysis , Protein Isoforms/chemistry , Species Specificity , Viral Envelope Proteins/analysis , Viral Envelope Proteins/chemistry , Viral Proteins/analysis , Viral Proteins/chemistry , Viral Proteins/genetics , Virion/chemistry
5.
Mol Ther ; 14(4): 536-45, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16950499

ABSTRACT

Lentiviral vectors based on equine infectious anemia virus (EIAV) stably integrate into dividing and nondividing cells such as neurons, conferring long-term expression of their transgene. The integration profile of an EIAV vector was analyzed in dividing HEK293T cells, alongside an HIV-1 vector as a control, and compared to a random dataset generated in silico. A multivariate regression model was generated and the influence of the following parameters on integration site selection determined: (a) within/not within a gene, (b) GC content within 20 kb, (c) within 10 kb of a CpG island, (d) gene density within a 2-Mb window, and (e) chromosome number. The majority of the EIAV integration sites (68%; n = 458) and HIV-1 integration sites (72%; n = 162) were within a gene, and both vectors favored AT-rich regions. Sites within genes were examined using a second model to determine the influence of the gene-specific parameters, gene region, and transcriptional activity. Both EIAV and HIV-1 vectors preferentially integrated within active genes. Unlike the gammaretrovirus MLV, EIAV and HIV-1 vectors do not integrate preferentially into the promoter region or the 5' end of the transcription unit.


Subject(s)
Genetic Vectors/genetics , Infectious Anemia Virus, Equine/genetics , Base Composition/genetics , Base Sequence , Cell Line , Chromosomes, Human/genetics , Gene Expression , Genome, Human/genetics , Humans , Transcription, Genetic/genetics
6.
J Virol ; 76(19): 9991-9, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12208975

ABSTRACT

The African swine fever virus (ASFV) j4R protein is expressed late during the virus replication cycle and is present in both the nucleus and the cytoplasm of infected cells. By using the yeast two-hybrid system, direct binding, and coprecipitation from cells, we showed that the j4R protein binds to the alpha chain of nascent polypeptide-associated complex (alpha NAC). Confocal microscopy indicated that a proportion of j4R and alpha NAC interact in areas close to the plasma membrane, as well as through the cytoplasm in cells. In vitro binding studies suggested that binding of j4R to alpha NAC did not interfere with the binding of alpha- and beta NAC subunits (the BTF3 transcription factor).


Subject(s)
African Swine Fever Virus/chemistry , Trans-Activators/metabolism , Viral Proteins/metabolism , African Swine Fever Virus/genetics , Animals , Cell Nucleus/chemistry , Chlorocebus aethiops , Cytoplasm/chemistry , Molecular Chaperones , Nuclear Proteins , Open Reading Frames , Transcription Factors/metabolism , Transfection , Two-Hybrid System Techniques , Vero Cells
7.
Microbiology (Reading) ; 143 ( Pt 5): 1745-1755, 1997 May.
Article in English | MEDLINE | ID: mdl-9168624

ABSTRACT

Five independent clones of the Propionibacterium acnes P-37 lipase gene (gehA) were obtained in Escherichia coli, and the gene was localized to a 2.75 kb Xhol fragment by subcloning. The five clones were shown to contain the same gene by Southern blotting with a DIG-labelled probe to gehA. The nucleotide sequence of gehA was determined, and shown to contain a single ORF of 1017 kb, encoding a protein of 339 amino acids. The predicted molecular mass was 36 kDa. A 33 kDa (PAGE) radiolabelled polypeptide was detected from E. coli minicell preparations harbouring gehA, which could correspond to GehA after cleavage of the putative 26 amino acid residue signal peptide. gehA was overexpressed in E. coli under the control of the bacteriophage T7 promoter, and the corresponding polypeptide was found to be present in insoluble aggregates. Active lipase was produced when the overexpressing strain was incubated at a reduced temperature in the presence of sucrose. Purification of lipase from P. acnes culture supernatant fluids confirmed the production of a 33 kDa (PAGE) lipase.


Subject(s)
Genes, Bacterial , Lipase/genetics , Propionibacterium acnes/enzymology , Amino Acid Sequence , Bacteriophage T7/genetics , Base Sequence , Chromosome Mapping , Chromosomes, Bacterial/genetics , Cloning, Molecular , Enzyme Induction , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression Regulation, Bacterial , Lipid Metabolism , Molecular Sequence Data , Molecular Weight , Open Reading Frames , Promoter Regions, Genetic , Propionibacterium acnes/genetics , Protein Sorting Signals/metabolism , Recombinant Fusion Proteins/biosynthesis , Sequence Alignment , Sequence Homology, Amino Acid , Species Specificity , Sucrose/metabolism , Temperature
SELECTION OF CITATIONS
SEARCH DETAIL
...