Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Ther Methods Clin Dev ; 3: 16067, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27900346

RESUMO

Gene therapy for HIV-1 infection is a promising alternative to lifelong combination antiviral drug treatment. Chemokine receptor 5 (CCR5) is the coreceptor required for R5-tropic HIV-1 infection of human cells. Deletion of CCR5 renders cells resistant to R5-tropic HIV-1 infection, and the potential for cure has been shown through allogeneic stem cell transplantation with naturally occurring homozygous deletion of CCR5 in donor hematopoietic stem/progenitor cells (HSPC). The requirement for HLA-matched HSPC bearing homozygous CCR5 deletions prohibits widespread application of this approach. Thus, a strategy to disrupt CCR5 genomic sequences in HSPC using zinc finger nucleases was developed. Following discussions with regulatory agencies, we conducted IND-enabling preclinical in vitro and in vivo testing to demonstrate the feasibility and (preclinical) safety of zinc finger nucleases-based CCR5 disruption in HSPC. We report here the clinical-scale manufacturing process necessary to deliver CCR5-specific zinc finger nucleases mRNA to HSPC using electroporation and the preclinical safety data. Our results demonstrate effective biallelic CCR5 disruption in up to 72.9% of modified colony forming units from adult mobilized HSPC with maintenance of hematopoietic potential in vitro and in vivo. Tumorigenicity studies demonstrated initial product safety; further safety and feasibility studies are ongoing in subjects infected with HIV-1 (NCT02500849@clinicaltrials.gov).

2.
Mol Ther ; 20(4): 849-59, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22273578

RESUMO

HIV-1-infected individuals can harbor viral isolates that can use CCR5, as well as CXCR4, for viral entry. To genetically engineer HIV-1 resistance in CD4(+) T cells, we assessed whether transient, adenovirus delivered zinc-finger nuclease (ZFN) disruption of genomic cxcr4 or stable lentiviral expression of short hairpin RNAs (shRNAs) targeting CXCR4 mRNAs provides durable resistance to HIV-1 challenge. ZFN-modification of cxcr4 in CD4(+) T cells was found to be superior to cell integrated lentivirus-expressing CXCR4 targeting shRNAs when CD4(+) T cells were challenged with HIV-1s that utilizes CXCR4 for entry. Cxcr4 disruption in CD4(+) T cells was found to be stable, conferred resistance, and provided for continued cell enrichment during HIV-1 infection in tissue culture and, in vivo, in peripheral blood mononuclear cell transplanted NSG mice. Moreover, HIV-1-infected mice with engrafted cxcr4 ZFN-modified CD4(+) T cells demonstrated lower viral levels in contrast to mice engrafted with unmodified CD4(+) T cells. These findings provide evidence that ZFN-mediated disruption of cxcr4 provides a selective advantage to CD4(+) T cells during HIV-1 infection.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Endonucleases/metabolismo , HIV-1/imunologia , RNA Interferente Pequeno/genética , Receptores CXCR4/genética , Adenoviridae/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Células Cultivadas , Endonucleases/genética , Humanos , Camundongos , Dedos de Zinco/genética
3.
PLoS Pathog ; 7(4): e1002020, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21533216

RESUMO

HIV-1 entry requires the cell surface expression of CD4 and either the CCR5 or CXCR4 coreceptors on host cells. Individuals homozygous for the ccr5Δ32 polymorphism do not express CCR5 and are protected from infection by CCR5-tropic (R5) virus strains. As an approach to inactivating CCR5, we introduced CCR5-specific zinc-finger nucleases into human CD4+ T cells prior to adoptive transfer, but the need to protect cells from virus strains that use CXCR4 (X4) in place of or in addition to CCR5 (R5X4) remains. Here we describe engineering a pair of zinc finger nucleases that, when introduced into human T cells, efficiently disrupt cxcr4 by cleavage and error-prone non-homologous DNA end-joining. The resulting cells proliferated normally and were resistant to infection by X4-tropic HIV-1 strains. CXCR4 could also be inactivated in ccr5Δ32 CD4+ T cells, and we show that such cells were resistant to all strains of HIV-1 tested. Loss of CXCR4 also provided protection from X4 HIV-1 in a humanized mouse model, though this protection was lost over time due to the emergence of R5-tropic viral mutants. These data suggest that CXCR4-specific ZFNs may prove useful in establishing resistance to CXCR4-tropic HIV for autologous transplant in HIV-infected individuals.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Desoxirribonucleases/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Receptores CXCR4/imunologia , Animais , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/transplante , Proliferação de Células , Desoxirribonucleases/biossíntese , Desoxirribonucleases/genética , Modelos Animais de Doenças , Engenharia Genética , Infecções por HIV/genética , Infecções por HIV/metabolismo , Infecções por HIV/terapia , HIV-1/genética , HIV-1/metabolismo , Humanos , Macaca mulatta , Camundongos , Receptores CCR5/genética , Receptores CCR5/imunologia , Receptores CCR5/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transplante Autólogo , Transplante Heterólogo , Internalização do Vírus
4.
Nat Biotechnol ; 26(7): 808-16, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18587387

RESUMO

Homozygosity for the naturally occurring Delta32 deletion in the HIV co-receptor CCR5 confers resistance to HIV-1 infection. We generated an HIV-resistant genotype de novo using engineered zinc-finger nucleases (ZFNs) to disrupt endogenous CCR5. Transient expression of CCR5 ZFNs permanently and specifically disrupted approximately 50% of CCR5 alleles in a pool of primary human CD4(+) T cells. Genetic disruption of CCR5 provided robust, stable and heritable protection against HIV-1 infection in vitro and in vivo in a NOG model of HIV infection. HIV-1-infected mice engrafted with ZFN-modified CD4(+) T cells had lower viral loads and higher CD4(+) T-cell counts than mice engrafted with wild-type CD4(+) T cells, consistent with the potential to reconstitute immune function in individuals with HIV/AIDS by maintenance of an HIV-resistant CD4(+) T-cell population. Thus adoptive transfer of ex vivo expanded CCR5 ZFN-modified autologous CD4(+) T cells in HIV patients is an attractive approach for the treatment of HIV-1 infection.


Assuntos
Transferência Adotiva/métodos , Linfócitos T CD4-Positivos/enzimologia , Linfócitos T CD4-Positivos/transplante , Desoxirribonucleases/genética , Infecções por HIV/prevenção & controle , Infecções por HIV/cirurgia , Dedos de Zinco/genética , Animais , Células Cultivadas , Mapeamento Cromossômico/métodos , Engenharia Genética/métodos , Humanos , Imunidade Inata , Camundongos , Resultado do Tratamento
5.
Nat Biotechnol ; 25(11): 1298-306, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17965707

RESUMO

Achieving the full potential of zinc-finger nucleases (ZFNs) for genome engineering in human cells requires their efficient delivery to the relevant cell types. Here we exploited the infectivity of integrase-defective lentiviral vectors (IDLV) to express ZFNs and provide the template DNA for gene correction in different cell types. IDLV-mediated delivery supported high rates (13-39%) of editing at the IL-2 receptor common gamma-chain gene (IL2RG) across different cell types. IDLVs also mediated site-specific gene addition by a process that required ZFN cleavage and homologous template DNA, thus establishing a platform that can target the insertion of transgenes into a predetermined genomic site. Using IDLV delivery and ZFNs targeting distinct loci, we observed high levels of gene addition (up to 50%) in a panel of human cell lines, as well as human embryonic stem cells (5%), allowing rapid, selection-free isolation of clonogenic cells with the desired genetic modification.


Assuntos
Reparo do DNA , Desoxirribonucleases de Sítio Específico do Tipo II/metabolismo , Células-Tronco Embrionárias/enzimologia , Engenharia Genética/métodos , Lentivirus/genética , Dedos de Zinco , Desoxirribonucleases de Sítio Específico do Tipo II/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Humanos , Integrases/genética , Subunidade gama Comum de Receptores de Interleucina/genética , Lentivirus/enzimologia , Mutação Puntual , Moldes Genéticos , Transgenes , Integração Viral/genética
6.
Nat Biotechnol ; 25(7): 778-85, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17603475

RESUMO

Genome editing driven by zinc-finger nucleases (ZFNs) yields high gene-modification efficiencies (>10%) by introducing a recombinogenic double-strand break into the targeted gene. The cleavage event is induced using two custom-designed ZFNs that heterodimerize upon binding DNA to form a catalytically active nuclease complex. Using the current ZFN architecture, however, cleavage-competent homodimers may also form that can limit safety or efficacy via off-target cleavage. Here we develop an improved ZFN architecture that eliminates this problem. Using structure-based design, we engineer two variant ZFNs that efficiently cleave DNA only when paired as a heterodimer. These ZFNs modify a native endogenous locus as efficiently as the parental architecture, but with a >40-fold reduction in homodimer function and much lower levels of genome-wide cleavage. This architecture provides a general means for improving the specificity of ZFNs as gene modification reagents.


Assuntos
Biotecnologia/métodos , Dedos de Zinco , Sequência de Bases , Sítios de Ligação , Catálise , Desoxirribonucleases de Sítio Específico do Tipo II/química , Dimerização , Genoma , Proteínas de Fluorescência Verde/química , Humanos , Células K562 , Modelos Biológicos , Conformação Molecular , Dados de Sequência Molecular , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...