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










Database
Language
Publication year range
1.
PLoS One ; 19(3): e0298542, 2024.
Article in English | MEDLINE | ID: mdl-38457474

ABSTRACT

Drug-based antiretroviral therapies (ART) efficiently suppress HIV replication in humans, but the virus persists as integrated proviral reservoirs in small numbers of cells. Importantly, ART cannot eliminate HIV from an infected individual, since it does not target the integrated provirus. Therefore, genome editing-based strategies that can inactivate or excise HIV genomes would provide the technology for novel curative therapies. In fact, the HIV-1 LTR-specific designer-recombinase Brec1 has been shown to remove integrated proviruses from infected cells and is highly efficacious on clinical HIV-1 isolates in vitro and in vivo, suggesting that Brec1 has the potential for clinical development of advanced HIV-1 eradication strategies in people living with HIV. In line with the preparation of a first-in-human advanced therapy medicinal product gene therapy trial, we here present an extensive preclinical evaluation of Brec1 and lentiviral vectors expressing the Brec1 transgene. This included detailed functional analysis of potential genomic off-target sites, assessing vector safety by investigating vector copy number (VCN) and the risk for potential vector-related insertional mutagenesis, as well as analyzing the potential of Brec1 to trigger an undesired strong T cell immune response. In conclusion, the antiviral designer-recombinase Brec1 is shown to lack any detectable cytopathic, genotoxic or T cell-related immunogenic effects, thereby meeting an important precondition for clinical application of the therapeutic lentiviral vector LV-Brec1 in novel HIV-1 curative strategies.


Subject(s)
HIV Infections , HIV-1 , Humans , Lentivirus/genetics , Lentivirus/metabolism , Recombinases/metabolism , HIV-1/physiology , Proviruses/genetics , HIV Long Terminal Repeat/genetics , HIV Infections/therapy , Genetic Vectors/genetics
2.
Mol Ther ; 31(7): 2266-2285, 2023 07 05.
Article in English | MEDLINE | ID: mdl-36934299

ABSTRACT

The human T cell leukemia virus type 1 (HTLV-1) is a pathogenic retrovirus that persists as a provirus in the genome of infected cells and can lead to adult T cell leukemia (ATL). Worldwide, more than 10 million people are infected and approximately 5% of these individuals will develop ATL, a highly aggressive cancer that is currently incurable. In the last years, genome editing tools have emerged as promising antiviral agents. In this proof-of-concept study, we use substrate-linked directed evolution (SLiDE) to engineer Cre-derived site-specific recombinases to excise the HTLV-1 proviral genome from infected cells. We identified a conserved loxP-like sequence (loxHTLV) present in the long terminal repeats of the majority of virus isolates. After 181 cycles of SLiDE, we isolated a designer-recombinase (designated RecHTLV), which efficiently recombines the loxHTLV sequence in bacteria and human cells with high specificity. Expression of RecHTLV in human Jurkat T cells resulted in antiviral activity when challenged with an HTLV-1 infection. Moreover, expression of RecHTLV in chronically infected SP cells led to the excision of HTLV-1 proviral DNA. Our data suggest that recombinase-mediated excision of the HTLV-1 provirus represents a promising approach to reduce proviral load in HTLV-1-infected individuals, potentially preventing the development of HTLV-1-associated diseases.


Subject(s)
Human T-lymphotropic virus 1 , Paraparesis, Tropical Spastic , Adult , Humans , Human T-lymphotropic virus 1/genetics , Paraparesis, Tropical Spastic/drug therapy , Paraparesis, Tropical Spastic/genetics , Proviruses/genetics , Antiviral Agents
3.
PLoS One ; 16(12): e0261269, 2021.
Article in English | MEDLINE | ID: mdl-34928979

ABSTRACT

Adeno-associated viruses (AAV) are considered non-pathogenic in humans, and thus have been developed into powerful vector platforms for in vivo gene therapy. Although the various AAV serotypes display broad tropism, frequently infecting multiple tissues and cell types, vectors for specific and efficient targeting of human CD4+ T lymphocytes are largely missing. In fact, a substantial translational bottleneck exists in the field of therapeutic gene transfer that would require in vivo delivery into peripheral disease-related lymphocytes for subsequent genome editing. To solve this issue, capsid modification for retargeting AAV tropism, and in turn improving vector potency, is considered a promising strategy. Here, we genetically modified the minor AAV2 capsid proteins, VP1 and VP2, with a set of novel nanobodies with high-affinity for the human CD4 receptor. These novel vector variants demonstrated improved targeting of human CD4+ cells, including primary human peripheral blood mononuclear cells (PBMC) and purified human CD4+ T lymphocytes. Thus, the technical approach presented here provides a promising strategy for developing specific gene therapy vectors, particularly targeting disease-related peripheral blood CD4+ leukocytes.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Capsid Proteins/genetics , Dependovirus/genetics , Genetic Vectors/administration & dosage , Leukocytes, Mononuclear/metabolism , Single-Domain Antibodies/chemistry , Transduction, Genetic , CD4-Positive T-Lymphocytes/immunology , Gene Transfer Techniques , HEK293 Cells , HeLa Cells , Humans , Leukocytes, Mononuclear/immunology
4.
Hum Gene Ther Methods ; 29(2): 104-113, 2018 04.
Article in English | MEDLINE | ID: mdl-29631437

ABSTRACT

The delivery of therapeutic genes for treatment of inherited or infectious diseases frequently requires lentiviral transduction of CD34+ hematopoietic stem and progenitor cells (HSC). Optimized transduction protocols with a therapeutic goal aim to maximize the number of transduction-positive cells while limiting the vector copy number that reach each individual cell. Importantly, the transduced HSC should maintain their "stem-like" properties. Here, we analyzed LentiBOOST™ reagent, a membrane-sealing poloxamer, with respect to enhancing lentiviral transduction of CD34+ peripheral blood stem cells. We demonstrate that inclusion of LentiBOOST™ in a standard HSC transduction protocol yields high transduction efficiencies while preserving the ability of the transduced HSC to differentiate into various hematopoietic lineages. Thus, LentiBOOST™ reagent can significantly improve lentiviral CD34+ HSC transduction protocols with the potential to improve production of gene-modified cell products.


Subject(s)
Genetic Vectors/immunology , HIV-1/genetics , Hematopoietic Stem Cells/virology , Real-Time Polymerase Chain Reaction/methods , T-Lymphocytes/immunology , Transduction, Genetic/methods , Antigens, CD34/genetics , Antigens, CD34/immunology , Cell Differentiation , Cell Line , Cell Lineage/immunology , DNA Copy Number Variations , Genes, Reporter , Genetic Therapy/methods , Genetic Vectors/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , HEK293 Cells , HIV-1/immunology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Plasmids/chemistry , Plasmids/metabolism , Poloxamer/chemistry , Primary Cell Culture , Protamines/chemistry , Real-Time Polymerase Chain Reaction/instrumentation , Real-Time Polymerase Chain Reaction/standards , T-Lymphocytes/cytology , T-Lymphocytes/virology , Transduction, Genetic/instrumentation , Transgenes
5.
Nat Biotechnol ; 34(4): 401-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26900663

ABSTRACT

Current combination antiretroviral therapies (cART) efficiently suppress HIV-1 reproduction in humans, but the virus persists as integrated proviral reservoirs in small numbers of cells. To generate an antiviral agent capable of eradicating the provirus from infected cells, we employed 145 cycles of substrate-linked directed evolution to evolve a recombinase (Brec1) that site-specifically recognizes a 34-bp sequence present in the long terminal repeats (LTRs) of the majority of the clinically relevant HIV-1 strains and subtypes. Brec1 efficiently, precisely and safely removes the integrated provirus from infected cells and is efficacious on clinical HIV-1 isolates in vitro and in vivo, including in mice humanized with patient-derived cells. Our data suggest that Brec1 has potential for clinical application as a curative HIV-1 therapy.


Subject(s)
Antiviral Agents/pharmacology , Directed Molecular Evolution/methods , HIV Infections/virology , HIV-1/drug effects , Proviruses/drug effects , Recombinases/pharmacology , Virus Integration/drug effects , Animals , Antiviral Agents/metabolism , Base Sequence , Cells, Cultured , HIV-1/genetics , Humans , Mice , Molecular Sequence Data , Proviruses/genetics , Recombinases/metabolism , Virus Integration/genetics
6.
Sci Rep ; 5: 13734, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26334116

ABSTRACT

Current antiviral therapies cannot cure hepatitis B virus (HBV) infection; successful HBV eradication would require inactivation of the viral genome, which primarily persists in host cells as episomal covalently closed circular DNA (cccDNA) and, to a lesser extent, as chromosomally integrated sequences. However, novel designer enzymes, such as the CRISPR/Cas9 RNA-guided nuclease system, provide technologies for developing advanced therapy strategies that could directly attack the HBV genome. For therapeutic application in humans, such designer nucleases should recognize various HBV genotypes and cause minimal off-target effects. Here, we identified cross-genotype conserved HBV sequences in the S and X region of the HBV genome that were targeted for specific and effective cleavage by a Cas9 nickase. This approach disrupted not only episomal cccDNA and chromosomally integrated HBV target sites in reporter cell lines, but also HBV replication in chronically and de novo infected hepatoma cell lines. Our data demonstrate the feasibility of using the CRISPR/Cas9 nickase system for novel therapy strategies aiming to cure HBV infection.


Subject(s)
Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Deoxyribonuclease I/genetics , Genome, Viral/genetics , Hepatitis B virus/genetics , Hepatitis B/genetics , Open Reading Frames/genetics , Gene Silencing
7.
RNA Biol ; 10(2): 216-27, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23235494

ABSTRACT

Aptamers are oligonucleotides that bind targets with high specificity and affinity. They have become important tools for biosensing, target detection, drug delivery and therapy. We selected the quadruplex-forming 16-mer DNA aptamer AID-1 [d(GGGT) 4] with affinity for the interleukin-6 receptor (IL-6R) and identified single nucleotide variants that showed no significant loss of binding ability. The RNA counterpart of AID-1 [r(GGGU) 4] also bound IL-6R as quadruplex structure. AID-1 is identical to the well-known HIV inhibitor T30923, which inhibits both HIV infection and HIV-1 integrase. We also demonstrated that IL-6R specific RNA aptamers not only bind HIV-1 integrase and inhibit its 3' processing activity in vitro, but also are capable of preventing HIV de novo infection with the same efficacy as the established inhibitor T30175. All these aptamer target interactions are highly dependent on formation of quadruplex structure.


Subject(s)
Aptamers, Nucleotide/pharmacology , HIV Integrase Inhibitors/pharmacology , HIV-1/drug effects , Receptors, Interleukin-6/metabolism , Circular Dichroism , Drug Evaluation, Preclinical , G-Quadruplexes/drug effects , HIV Envelope Protein gp120/genetics , HIV Envelope Protein gp120/metabolism , HIV Infections/pathology , HIV Infections/virology , HIV Integrase/genetics , HIV Integrase/metabolism , HIV-1/enzymology , HIV-1/pathogenicity , HeLa Cells , Humans , Oligonucleotides/pharmacology , Virus Attachment/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...