Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Ther Nucleic Acids ; 4: e236, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25872029

RESUMO

We described earlier a dual-combination anti-HIV type 1 (HIV-1) lentiviral vector (LVsh5/C46) that downregulates CCR5 expression of transduced cells via RNAi and inhibits HIV-1 fusion via cell surface expression of cell membrane-anchored C46 antiviral peptide. This combinatorial approach has two points of inhibition for R5-tropic HIV-1 and is also active against X4-tropic HIV-1. Here, we utilize the humanized bone marrow, liver, thymus (BLT) mouse model to characterize the in vivo efficacy of LVsh5/C46 (Cal-1) vector to engineer cellular resistance to HIV-1 pathogenesis. Human CD34+ hematopoietic stem/progenitor cells (HSPC) either nonmodified or transduced with LVsh5/C46 vector were transplanted to generate control and treatment groups, respectively. Control and experimental groups displayed similar engraftment and multilineage hematopoietic differentiation that included robust CD4+ T-cell development. Splenocytes isolated from the treatment group were resistant to both R5- and X4-tropic HIV-1 during ex vivo challenge experiments. Treatment group animals challenged with R5-tropic HIV-1 displayed significant protection of CD4+ T-cells and reduced viral load within peripheral blood and lymphoid tissues up to 14 weeks postinfection. Gene-marking and transgene expression were confirmed stable at 26 weeks post-transplantation. These data strongly support the use of LVsh5/C46 lentiviral vector in gene and cell therapeutic applications for inhibition of HIV-1 infection.

2.
Viruses ; 6(1): 54-68, 2013 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-24381033

RESUMO

Human immunodeficiency virus type 1 (HIV-1) infection of target cells requires CD4 and a co-receptor, predominantly the chemokine receptor CCR5. CCR5-delta32 homozygosity results in a truncated protein providing natural protection against HIV infection-this without detrimental effects to the host-and transplantation of CCR5-delta32 stem cells in a patient with HIV ("Berlin patient") achieved viral eradication. As a more feasible approach gene-modification strategies are being developed to engineer cellular resistance to HIV using autologous cells. We have developed a dual therapeutic anti-HIV lentiviral vector (LVsh5/C46) that down-regulates CCR5 and inhibits HIV-1 fusion via cell surface expression of the gp41-derived peptide, C46. This construct, effective against multiple strains of both R5- and X4-tropic HIV-1, is being tested in Phase I/II trials by engineering HIV-resistant hematopoietic cells.


Assuntos
Antagonistas dos Receptores CCR5 , Terapia Genética/métodos , Infecções por HIV/terapia , HIV-1/fisiologia , Receptores de HIV/antagonistas & inibidores , Proteínas Recombinantes de Fusão/biossíntese , Terapia Biológica/métodos , Ensaios Clínicos como Assunto , Infecções por HIV/virologia , HIV-1/imunologia , Transplante de Células-Tronco Hematopoéticas/métodos , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/fisiologia , Células-Tronco Hematopoéticas/virologia , Humanos , Receptores CCR5/biossíntese , Receptores de HIV/biossíntese , Proteínas Recombinantes de Fusão/genética
3.
Immunol Res ; 48(1-3): 84-98, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20737298

RESUMO

HIV/AIDS is a disease that impairs immune function, primarily by decreasing T-lymphocyte count. Its progression can be contained by highly active antiretroviral therapy (HAART), but there are side effects that can be severe, and the development of resistance often forces the physician to modify the HAART regimen. There are no vaccines available for HIV. An alternative approach that could provide a path to a curative therapy is the use of cell-delivered gene therapy in which an anti-HIV gene(s) is introduced into hematopoietic cells to produce a population that is protected from the effects of HIV. In this paper, we review the field and discuss an approach using a short hairpin RNA to CCR5, an important co-receptor for HIV.


Assuntos
Infecções por HIV/terapia , HIV/fisiologia , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , RNA Interferente Pequeno/uso terapêutico , Receptores CCR5/metabolismo , Receptores de HIV/metabolismo , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Genes Virais/genética , Terapia Genética/tendências , HIV/patogenicidade , Infecções por HIV/genética , Infecções por HIV/imunologia , Células-Tronco Hematopoéticas/patologia , Humanos , Camundongos , RNA Interferente Pequeno/genética , Receptores CCR5/genética , Receptores de HIV/genética , Replicação Viral/genética
4.
Virol J ; 6: 184, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19878571

RESUMO

BACKGROUND: Multiple short hairpin RNA (shRNA) gene therapy strategies are currently being investigated for treating viral diseases such as HIV-1. It is important to use several different shRNAs to prevent the emergence of treatment-resistant strains. However, there is evidence that repeated expression cassettes delivered via lentiviral vectors may be subject to recombination-mediated repeat deletion of 1 or more cassettes. RESULTS: The aim of this study was to determine the frequency of deletion for 2 to 6 repeated shRNA cassettes and mathematically model the outcomes of different frequencies of deletion in gene therapy scenarios. We created 500+ clonal cell lines and found deletion frequencies ranging from 2 to 36% for most combinations. While the central positions were the most frequently deleted, there was no obvious correlation between the frequency or extent of deletion and the number of cassettes per combination. We modeled the progression of infection using combinations of 6 shRNAs with varying degrees of deletion. Our in silico modeling indicated that if at least half of the transduced cells retained 4 or more shRNAs, the percentage of cells harboring multiple-shRNA resistant viral strains could be suppressed to < 0.1% after 13 years. This scenario afforded a similar protection to all transduced cells containing the full complement of 6 shRNAs. CONCLUSION: Deletion of repeated expression cassettes within lentiviral vectors of up to 6 shRNAs can be significant. However, our modeling showed that the deletion frequencies observed here for 6x shRNA combinations was low enough that the in vivo suppression of replication and escape mutants will likely still be effective.


Assuntos
Terapia Genética/métodos , Infecções por HIV/terapia , HIV-1/genética , Mutagênese Insercional/métodos , RNA Interferente Pequeno/genética , Deleção de Sequência , Simulação por Computador , Humanos
5.
J Gene Med ; 7(5): 552-64, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15655805

RESUMO

BACKGROUND: An anti-HIV-1 tat ribozyme, termed Rz2, has been shown to inhibit HIV-1 infection/replication and to decrease HIV-1-induced pathogenicity in T-lymphocyte cell lines and normal peripheral blood T-lymphocytes. We report here the results of a phase I gene transfer clinical trial using Rz2. METHODS: Apheresis was used to obtain a peripheral blood cell population from each of four HIV-negative donors. After enrichment for CD4+ T-lymphocytes, ex vivo expansion and genetic manipulation (approximately equal aliquots of the cells were transduced with the ribozyme-containing (RRz2) and the control (LNL6) retroviral vector), these cells were infused into the corresponding HIV-1-positive twin recipient. Marking was assessed over an initial 24-week period and in total over an approximate 4-year period. RESULTS: The gene transfer procedure was shown to be safe, and technically feasible. Both RRz2- and LNL6-gene-containing peripheral blood mononuclear cells (PBMC) were detected at all time points examined to 4 years. There was concomitant gene construct expression in the absence of the need for ex vivo peripheral blood cell stimulation and there was no evidence of immune elimination of the neoR T-lymphocytes nor of silencing of the Moloney murine leukemia virus long terminal repeat. CONCLUSIONS: The proof of principle results reported here demonstrate safety and feasibility of this type of gene transfer approach. While not specifically tested, T-lymphocytes containing an anti-HIV gene construct may impact on HIV-1 viral load and CD4+ T-lymphocyte count, potentially representing a new therapeutic modality for HIV-1 infection.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Doenças em Gêmeos/terapia , Terapia Genética , Infecções por HIV/terapia , HIV-1 , RNA Catalítico/farmacologia , Transdução Genética , Adulto , Contagem de Linfócito CD4 , Doenças em Gêmeos/imunologia , Expressão Gênica , Genes tat/fisiologia , Vetores Genéticos , Infecções por HIV/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , RNA Catalítico/genética , Retroviridae/genética , Taxa de Sobrevida , Fatores de Tempo , Gêmeos Monozigóticos
6.
Methods Mol Biol ; 252: 581-98, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15017082

RESUMO

Antiretroviral drug therapy can effectively reduce the viral load, and is associated with a degree of immune reconstitution in human immunodeficiency virus (HIV)-infected patients. However, the presence of a latent viral reservoir, the development of drug resistance, drug toxicity, and compliance problems are obstacles that impede full eradication of HIV through drug therapy. The cellular introduction of genetic elements that are capable of inhibiting HIV replication is conceptually appealing as a potential new treatment paradigm for acquired immunodeficiency syndrome (AIDS). In theory, this approach can lead to the development of regenerated hematopoiesis with cells that inhibit viral replication and are protected from the pathogenic effects of HIV. Ribozymes are catalytic RNA molecules that can efficiently and selectively cleave target RNA. By ex vivo retroviral transduction, we have introduced a HIV-1 tat gene-targeted ribozyme (RRz2) and a control construct (LNL6) into granulocyte-colony-stimulating factor (G-CSF) mobilized CD34+ hematopoietic progenitor cells (HPC). Transduced autologous CD34+ cells (an approximately equal mix of RRz2 and LNL6) were infused in 10 patients in this Phase I study. After a median follow-up of 2.5 yr, gene presence and expression were detected by a sensitive polymerase chain reaction (PCR) assay in a transduced-CD34+ cell dose-dependent manner. In this chapter, we describe general considerations related to HIV hematopoietic progenitor-cell gene therapy trial design, implementation, and safety, with an emphasis on the critical steps of this process, namely vector production and characterization, target-cell selection, transduction, final product release testing, and evaluation of vector presence.


Assuntos
Síndrome da Imunodeficiência Adquirida/tratamento farmacológico , Terapia Genética/métodos , Infecções por HIV/tratamento farmacológico , RNA Catalítico/uso terapêutico , Antígenos CD/sangue , Antígenos CD34/sangue , Sequência de Bases , Técnicas de Transferência de Genes , Humanos , Conformação de Ácido Nucleico , RNA Catalítico/química , RNA Catalítico/toxicidade , Transplante de Células-Tronco , Transplante Autólogo
7.
Methods Mol Biol ; 252: 599-616, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15017083

RESUMO

The implementation of a hematopoietic progenitor-cell gene-therapy program involves the performance of laboratory procedures and compliance with the current code of Good Manufacturing Practices. This chapter explains the multiple laboratory steps used in our recent Phase I gene transfer study for HIV. This study employed a retroviral vector to deliver an anti-HIV ribozyme to CD34+ hematopoietic progenitor cells.


Assuntos
Terapia Genética/métodos , Terapia Genética/normas , Vetores Genéticos , RNA Catalítico/uso terapêutico , Transplante de Células-Tronco/métodos , Mobilização de Células-Tronco Hematopoéticas/métodos , Humanos , Transplante Autólogo/métodos , Transplante Autólogo/normas
8.
Hum Gene Ther ; 15(3): 251-62, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15018734

RESUMO

A phase I gene transfer clinical study was undertaken to examine the ability to introduce a potential anti-human immunodeficiency virus (HIV) gene therapeutic into hematopoietic progenitor cells (HPC), thereby contributing to multilineage engraftment. The potential therapeutic effect of genetically modifying HPC with protective genes in HIV-infected adults depends in part on the presence of adult thymic activity and myeloid capacity in the setting of HIV replication. Herein we report the presence and expression of a retroviral vector encoding an anti-HIV-1 ribozyme in mature hematopoietic cells of different lineages, and de novo T-lymphocyte development ensuing from genetically engineered CD34(+) HPC. Sustained output of vector-containing mature myeloid and T-lymphoid cells was detected even in patients with multidrug-resistant infection. In addition, the study showed that the degree of persistence of gene-containing cells was dependent on transduced HPC dose. These novel findings support the concept of gene therapy as a modality to effect immune reconstitution with cells engineered to inhibit HIV replication and this report represents the first demonstration of long-term maintenance of a potential therapeutic transgene in HIV disease.


Assuntos
Fármacos Anti-HIV , Terapia Genética/métodos , Infecções por HIV/imunologia , HIV-1 , Transplante de Células-Tronco Hematopoéticas , RNA Catalítico/genética , Adulto , Antígenos CD34/análise , Linfócitos T CD4-Positivos/metabolismo , Feminino , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Infecções por HIV/terapia , HIV-1/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/enzimologia , Humanos , Linfócitos/citologia , Linfócitos/metabolismo , Masculino , Pessoa de Meia-Idade , Células Mieloides/citologia , Reação em Cadeia da Polimerase , Retroviridae/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA