Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 127(5): 1689-1699, 2017 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-28346229

RESUMEN

BACKGROUND: Autologous hematopoietic stem cell transplantation (HSCT) of gene-modified cells is an alternative to enzyme replacement therapy (ERT) and allogeneic HSCT that has shown clinical benefit for adenosine deaminase-deficient (ADA-deficient) SCID when combined with reduced intensity conditioning (RIC) and ERT cessation. Clinical safety and therapeutic efficacy were evaluated in a phase II study. METHODS: Ten subjects with confirmed ADA-deficient SCID and no available matched sibling or family donor were enrolled between 2009 and 2012 and received transplantation with autologous hematopoietic CD34+ cells that were modified with the human ADA cDNA (MND-ADA) γ-retroviral vector after conditioning with busulfan (90 mg/m2) and ERT cessation. Subjects were followed from 33 to 84 months at the time of data analysis. Safety of the procedure was assessed by recording the number of adverse events. Efficacy was assessed by measuring engraftment of gene-modified hematopoietic stem/progenitor cells, ADA gene expression, and immune reconstitution. RESULTS: With the exception of the oldest subject (15 years old at enrollment), all subjects remained off ERT with normalized peripheral blood mononuclear cell (PBMC) ADA activity, improved lymphocyte numbers, and normal proliferative responses to mitogens. Three of nine subjects were able to discontinue intravenous immunoglobulin replacement therapy. The MND-ADA vector was persistently detected in PBMCs (vector copy number [VCN] = 0.1-2.6) and granulocytes (VCN = 0.01-0.3) through the most recent visits at the time of this writing. No patient has developed a leukoproliferative disorder or other vector-related clinical complication since transplant. CONCLUSION: These results demonstrate clinical therapeutic efficacy from gene therapy for ADA-deficient SCID, with an excellent clinical safety profile. TRIAL REGISTRATION: ClinicalTrials.gov NCT00794508. FUNDING: Food and Drug Administration Office of Orphan Product Development award, RO1 FD003005; NHLBI awards, PO1 HL73104 and Z01 HG000122; UCLA Clinical and Translational Science Institute awards, UL1RR033176 and UL1TR000124.


Asunto(s)
Adenosina Desaminasa/deficiencia , Agammaglobulinemia , Regulación Enzimológica de la Expresión Génica , Terapia Genética , Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave , Transducción Genética , Adenosina Desaminasa/biosíntesis , Adenosina Desaminasa/genética , Adolescente , Agammaglobulinemia/enzimología , Agammaglobulinemia/genética , Agammaglobulinemia/terapia , Autoinjertos , Niño , Preescolar , Femenino , Vectores Genéticos , Humanos , Lactante , Masculino , Retroviridae , Inmunodeficiencia Combinada Grave/enzimología , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/terapia
2.
Hum Gene Ther ; 28(1): 112-124, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27611239

RESUMEN

During B and T lymphocyte maturation, V(D)J recombination is initiated by creation of DNA double-strand breaks. Artemis is an exonuclease essential for their subsequent repair by nonhomologous end-joining. Mutations in DCLRE1C, the gene encoding Artemis, cause T-B-NK+ severe combined immunodeficiency (ART-SCID) and also confer heightened sensitivity to ionizing radiation and alkylating chemotherapy. Although allogeneic hematopoietic cell transplantation can treat ART-SCID, conditioning regimens are poorly tolerated, leading to early mortality and/or late complications, including short stature, endocrinopathies, and dental aplasia. However, without alkylating chemotherapy as preconditioning, patients usually have graft rejection or limited T cell and no B cell recovery. Thus, addition of normal DCLRE1C cDNA to autologous hematopoietic stem cells is an attractive strategy to treat ART-SCID. We designed a self-inactivating lentivirus vector containing human Artemis cDNA under transcriptional regulation of the human endogenous Artemis promoter (AProArt). Fibroblasts from ART-SCID patients transduced with AProArt lentivirus showed correction of radiosensitivity. Mobilized peripheral blood CD34+ cells from an ART-SCID patient as well as hematopoietic stem cells from Artemis-deficient mice demonstrated restored T and B cell development following AProArt transduction. Murine hematopoietic cells transduced with AProArt exhibited no increase in replating potential in an in vitro immortalization assay, and analysis of AProArt lentivirus insertions showed no predilection for sites that could activate oncogenes. These efficacy and safety findings support institution of a clinical trial of gene addition therapy for ART-SCID.


Asunto(s)
Endonucleasas/genética , Terapia Genética , Vectores Genéticos/administración & dosificación , Lentivirus/genética , Proteínas Nucleares/genética , Inmunodeficiencia Combinada Grave/terapia , Animales , Linfocitos B/citología , Linfocitos B/metabolismo , Linfocitos B/efectos de la radiación , Células Cultivadas , Terapia Combinada , Reparación del ADN/efectos de la radiación , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Endonucleasas/deficiencia , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Rayos gamma , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de la radiación , Humanos , Ratones , Ratones Noqueados , Ratones SCID , Proteínas Nucleares/deficiencia , Tolerancia a Radiación/genética , Inmunodeficiencia Combinada Grave/genética , Linfocitos T/citología , Linfocitos T/metabolismo , Linfocitos T/efectos de la radiación
3.
Mol Ther ; 22(3): 607-622, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24256635

RESUMEN

Gene transfer into autologous hematopoietic stem cells by γ-retroviral vectors (gRV) is an effective treatment for adenosine deaminase (ADA)-deficient severe combined immunodeficiency (SCID). However, current gRV have significant potential for insertional mutagenesis as reported in clinical trials for other primary immunodeficiencies. To improve the efficacy and safety of ADA-SCID gene therapy (GT), we generated a self-inactivating lentiviral vector (LV) with a codon-optimized human cADA gene under the control of the short form elongation factor-1α promoter (LV EFS ADA). In ADA(-/-) mice, LV EFS ADA displayed high-efficiency gene transfer and sufficient ADA expression to rescue ADA(-/-) mice from their lethal phenotype with good thymic and peripheral T- and B-cell reconstitution. Human ADA-deficient CD34(+) cells transduced with 1-5 × 10(7) TU/ml had 1-3 vector copies/cell and expressed 1-2x of normal endogenous levels of ADA, as assayed in vitro and by transplantation into immune-deficient mice. Importantly, in vitro immortalization assays demonstrated that LV EFS ADA had significantly less transformation potential compared to gRV vectors, and vector integration-site analysis by nrLAM-PCR of transduced human cells grown in immune-deficient mice showed no evidence of clonal skewing. These data demonstrated that the LV EFS ADA vector can effectively transfer the human ADA cDNA and promote immune and metabolic recovery, while reducing the potential for vector-mediated insertional mutagenesis.


Asunto(s)
Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Agammaglobulinemia/inmunología , Agammaglobulinemia/terapia , Vectores Genéticos/efectos adversos , Lentivirus/genética , Factor 1 de Elongación Peptídica/genética , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/terapia , Adenosina Desaminasa/inmunología , Adenosina Desaminasa/metabolismo , Agammaglobulinemia/genética , Agammaglobulinemia/patología , Animales , Linfocitos B/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Células HEK293 , Células HT29 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/patología , Linfocitos T/inmunología , Transducción Genética , Integración Viral
4.
Blood ; 120(18): 3635-46, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22968453

RESUMEN

We conducted a gene therapy trial in 10 patients with adenosine deaminase (ADA)-deficient severe combined immunodeficiency using 2 slightly different retroviral vectors for the transduction of patients' bone marrow CD34(+) cells. Four subjects were treated without pretransplantation cytoreduction and remained on ADA enzyme-replacement therapy (ERT) throughout the procedure. Only transient (months), low-level (< 0.01%) gene marking was observed in PBMCs of 2 older subjects (15 and 20 years of age), whereas some gene marking of PBMC has persisted for the past 9 years in 2 younger subjects (4 and 6 years). Six additional subjects were treated using the same gene transfer protocol, but after withdrawal of ERT and administration of low-dose busulfan (65-90 mg/m(2)). Three of these remain well, off ERT (5, 4, and 3 years postprocedure), with gene marking in PBMC of 1%-10%, and ADA enzyme expression in PBMC near or in the normal range. Two subjects were restarted on ERT because of poor gene marking and immune recovery, and one had a subsequent allogeneic hematopoietic stem cell transplantation. These studies directly demonstrate the importance of providing nonmyeloablative pretransplantation conditioning to achieve therapeutic benefits with gene therapy for ADA-deficient severe combined immunodeficiency.


Asunto(s)
Agammaglobulinemia/terapia , Trasplante de Médula Ósea/métodos , Terapia Genética/métodos , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas/métodos , Inmunodeficiencia Combinada Grave/terapia , Adenosina Desaminasa/deficiencia , Adolescente , Antígenos CD34/metabolismo , Niño , Preescolar , Femenino , Humanos , Lactante , Masculino , Retroviridae/genética , Transducción Genética , Acondicionamiento Pretrasplante , Adulto Joven
5.
Blood ; 120(18): 3677-87, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22833548

RESUMEN

Gene therapy (GT) for adenosine deaminase-deficient severe combined immune deficiency (ADA-SCID) can provide significant long-term benefit when patients are given nonmyeloablative conditioning and ADA enzyme-replacement therapy (ERT) is withheld before autologous transplantation of γ-retroviral vector-transduced BM CD34+ cells. To determine the contributions of conditioning and discontinuation of ERT to the therapeutic effects, we analyzed these factors in Ada gene knockout mice (Ada(-/-)). Mice were transplanted with ADA-deficient marrow transduced with an ADA-expressing γ-retroviral vector without preconditioning or after 200 cGy or 900 cGy total-body irradiation and evaluated after 4 months. In all tissues analyzed, vector copy numbers (VCNs) were 100- to 1000-fold greater in mice receiving 900 cGy compared with 200 cGy (P < .05). In mice receiving 200 cGy, VCN was similar whether ERT was stopped or given for 1 or 4 months after GT. In unconditioned mice, there was decreased survival with and without ERT, and VCN was very low to undetectable. When recipients were conditioned with 200 cGy and received transduced lineage-depleted marrow, only recipients receiving ERT (1 or 4 months) had detectable vector sequences in thymocytes. In conclusion, cytoreduction is important for the engraftment of gene-transduced HSC, and short-term ERT after GT did not diminish the capacity of gene-corrected cells to engraft and persist.


Asunto(s)
Adenosina Desaminasa/uso terapéutico , Agammaglobulinemia/terapia , Trasplante de Médula Ósea/métodos , Terapia Genética/métodos , Inmunodeficiencia Combinada Grave/terapia , Acondicionamiento Pretrasplante/métodos , Adenosina Desaminasa/deficiencia , Animales , Modelos Animales de Enfermedad , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas/métodos , Ratones , Ratones Noqueados , Retroviridae , Transducción Genética
6.
Blood ; 111(12): 5745-54, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18356486

RESUMEN

Adenosine deaminase (ADA)-deficient severe combined immune deficiency (SCID) may be treated by allogeneic hematopoietic stem cell transplantation without prior cytoreductive conditioning, although the mechanism of immune reconstitution is unclear. We studied this process in a murine gene knockout model of ADA-deficient SCID. Newborn ADA-deficient pups received transplants of intravenous infusion of normal congenic bone marrow, without prior cytoreductive conditioning, which resulted in long-term survival, multisystem correction, and nearly normal lymphocyte numbers and mitogenic proliferative responses. Only 1% to 3% of lymphocytes and myeloid cells were of donor origin without a selective expansion of donor-derived lymphocytes; immune reconstitution was by endogenous, host-derived ADA-deficient lymphocytes. Preconditioning of neonates with 100 to 400 cGy of total body irradiation before normal donor marrow transplant increased the levels of engrafted donor cells in a radiation dose-dependent manner, but the chimerism levels were similar for lymphoid and myeloid cells. The absence of selective reconstitution by donor T lymphocytes in the ADA-deficient mice indicates that restoration of immune function occurred by rescue of endogenous ADA-deficient lymphocytes through cross-correction from the engrafted ADA-replete donor cells. Thus, ADA-deficient SCID is unique in its responses to nonmyeloablative bone marrow transplantation, which has implications for clinical bone marrow transplantation or gene therapy.


Asunto(s)
Adenosina Desaminasa/genética , Trasplante de Médula Ósea , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/terapia , Subgrupos de Linfocitos T/inmunología , Adenosina Desaminasa/metabolismo , Animales , Animales Recién Nacidos , Activación Enzimática , Supervivencia de Injerto/inmunología , Subunidad gamma Común de Receptores de Interleucina/genética , Ratones , Ratones Endogámicos , Ratones Noqueados , Ratones SCID , Recuperación de la Función/inmunología , Inmunodeficiencia Combinada Grave/genética , Tasa de Supervivencia , Subgrupos de Linfocitos T/citología , Quimera por Trasplante , Acondicionamiento Pretrasplante
7.
Blood ; 109(2): 503-6, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16973956

RESUMEN

A patient with adenosine deaminase-deficient severe combined immune deficiency (ADA-SCID) was enrolled in a study of retroviral-mediated ADA gene transfer to bone marrow hematopoietic stem cells. After the discontinuation of ADA enzyme replacement, busulfan (75 mg/m2) was administered for bone marrow cytoreduction, followed by infusion of autologous, gene-modified CD34+ cells. The expected myelosuppression developed after busulfan but then persisted, necessitating the administration of untransduced autologous bone marrow back-up at day 40. Because of sustained pancytopenia and negligible gene marking, diagnostic bone marrow biopsy and aspirate were performed at day 88. Analyses revealed hypocellular marrow and, unexpectedly, evidence of trisomy 8 in 21.6% of cells. Trisomy 8 mosaicism (T8M) was subsequently diagnosed by retrospective analysis of a pretreatment marrow sample that might have caused the lack of hematopoietic reconstitution. The confounding effects of this preexisting marrow cytogenetic abnormality on the response to gene transfer highlights another challenge of gene therapy with the use of autologous hematopoietic stem cells.


Asunto(s)
Adenosina Desaminasa/deficiencia , Cromosomas Humanos Par 8/genética , Terapia Genética , Mosaicismo , Pancitopenia/terapia , Inmunodeficiencia Combinada Grave/terapia , Trisomía , Adenosina Desaminasa/genética , Preescolar , Análisis Citogenético , Femenino , Humanos , Pancitopenia/etiología , Estudios Retrospectivos , Inmunodeficiencia Combinada Grave/genética
8.
Mol Ther ; 13(6): 1110-20, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16651028

RESUMEN

Using a mouse model of adenosine deaminase-deficient severe combined immune deficiency syndrome (ADA-deficient SCID), we have developed a noninvasive method of gene transfer for the sustained systemic expression of human ADA as enzyme replacement therapy. The method of delivery is a human immunodeficiency virus 1-based lentiviral vector given systemically by intravenous injection on day 1 to 2 of life. In this article we characterize the biodistribution of the integrated vector, the expression levels of ADA enzyme activity in various tissues, as well as the efficacy of systemic ADA expression to correct the ADA-deficient phenotype in this mouse model. The long-term expression of enzymatically active ADA achieved by this method, primarily from transduction of liver and lung, restored immunologic function and significantly extended survival. These studies illustrate the potential for sustained in vivo production of enzymatically active ADA, as an alternative to therapy by frequent injection of exogenous ADA protein.


Asunto(s)
Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Vectores Genéticos/administración & dosificación , VIH-1/genética , Inmunodeficiencia Combinada Grave/terapia , Adenosina Desaminasa/metabolismo , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Terapia Genética/métodos , Vectores Genéticos/farmacocinética , Células Germinativas/fisiología , Humanos , Inyecciones Intravenosas , Masculino , Ratones , Ratones Noqueados , Inmunodeficiencia Combinada Grave/inmunología , Distribución Tisular , Transducción Genética
9.
Mol Ther ; 12(1): 77-86, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15963923

RESUMEN

Two HIV-1-infected children on antiretroviral therapy were enrolled into a clinical study of retroviral-mediated transfer of a gene that inhibits replication of HIV-1, targeting bone marrow CD34+ hematopoietic stem/progenitor cells. Two retroviral vectors were used, one encoding a "humanized" dominant-negative REV protein (huM10) that is a potent inhibitor of HIV-1 replication and one encoding a nontranslated marker gene (FX) to serve as an internal control for the level of gene marking. Peripheral blood mononuclear cells (PBMC) containing the huM10 gene or FX gene were detected by quantitative PCR at frequencies of approximately 1/10,000 in both subjects for the first 1-3 months following re-infusion of the gene-transduced bone marrow, but then were at or below the limits of detection (<1/1,000,000) at most times over 2 years. In one patient, a reappearance of PBMC containing the huM10 gene, but not the FX gene, occurred concomitant with a rise in the HIV-1 viral load during a period of nonadherence to the antiretroviral regimen. Unique clones of gene-marked PBMC were detected by LAM-PCR during the time of elevated HIV-1 levels. These findings indicate that there was a selective survival advantage for PBMC containing the huM10 gene during the time of increased HIV-1 load.


Asunto(s)
Células de la Médula Ósea/metabolismo , Terapia Genética , Infecciones por VIH/tratamiento farmacológico , Linfocitos/metabolismo , Adolescente , Antígenos CD34/inmunología , Células de la Médula Ósea/inmunología , Supervivencia Celular/efectos de los fármacos , Niño , Preescolar , ADN/farmacología , Femenino , Marcadores Genéticos , VIH-1/efectos de los fármacos , Humanos , Reacción en Cadena de la Polimerasa , Transducción Genética
10.
Mol Ther ; 11(5): 776-89, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15851016

RESUMEN

Mucopolysaccharidosis type I (MPS I) is a lysosomal glycosaminoglycan (GAG) storage disorder caused by deficiency of alpha-l-iduronidase (IDUA). In this study, we evaluated the potential to perform gene therapy for MPS I by direct in vivo injection of a lentiviral vector, using an IDUA gene knockout murine model. We compared the efficacy in newborn versus young adult MPS I mice of a single intravenous injection of the lentiviral vector. The extent of transduction was dose-dependent, with the liver receiving the highest level of vector, but other somatic organs reaching almost the same level. The phenotypic manifestations of disease were partially improved in the mice treated as young adults, but were nearly normalized at every end-point measured in the mice treated as neonates. In the neonatally treated mice, the expressed IDUA activity resulted in decreased GAG storage, prevention of skeletal abnormalities, a more normal gross appearance, and improved survival. Most strikingly, significant levels of IDUA enzyme were produced in the brain of mice treated as neonates, with transduction of neurons at high levels. The sustained expression of enzymatically active IDUA in multiple organs had a significant beneficial effect on the phenotypic abnormalities of MPS I, which may be translated to clinical gene therapy of patients with Hurler disease.


Asunto(s)
Terapia Genética , Vectores Genéticos/genética , Lentivirus/genética , Mucopolisacaridosis I/genética , Mucopolisacaridosis I/terapia , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Huesos/anomalías , Huesos/metabolismo , Línea Celular Tumoral , Sistema Nervioso Central/metabolismo , ADN Complementario/genética , Expresión Génica , Vectores Genéticos/administración & dosificación , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Humanos , Iduronidasa/deficiencia , Iduronidasa/genética , Iduronidasa/metabolismo , Inyecciones Intravenosas , Lisosomas/genética , Lisosomas/metabolismo , Ratones , Ratones Noqueados , Mucopolisacaridosis I/enzimología , Neuronas/metabolismo , Azufre/química , Tasa de Supervivencia
11.
Ann N Y Acad Sci ; 996: 112-21, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12799289

RESUMEN

The identification of unknown genomic flanking DNA sequences can be used for the molecular monitoring of retro-, lenti- and foamyviral integration, transgenes in early embryogenesis, insertional mutagenesis, cell fate, and stem cell plasticity. Most existing methods reflect shortcomings in sensitivity and or specificity, thus limiting genomic sequencing of unknown flanking DNA to clonal preparations. The application of linear amplification-mediated PCR (LAM-PCR), a recently developed direct sequencing technique for flanking DNA, should circumvent current limitations in different research fields. This technique combines preamplification of target DNA with a unique succession of enzymatic reactions on solid-phase. Using LAM-PCR, we show the previously unfeasible in vivo retro-, lenti- and foamyvirus integration site analysis in primate peripheral blood hematopoietic cells and human xenograft hematopoiesis. In light of two severe adverse events that occurred in a clinical SCID-X1 gene therapy trial, in vivo monitoring of the reinfused transduced cell pool by integration site analysis will be an important component of each gene transfer and therapy study aimed at clinical use.


Asunto(s)
Células Madre Hematopoyéticas/clasificación , Células Madre Hematopoyéticas/citología , Animales , Secuencia de Bases , Elementos Transponibles de ADN/genética , Vectores Genéticos , Células HeLa , Humanos , Macaca mulatta , Papio , Reacción en Cadena de la Polimerasa , Retroviridae/genética , Transducción Genética
12.
Nat Med ; 9(4): 463-8, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12640448

RESUMEN

A clinical trial of retroviral-mediated transfer of the adenosine deaminase (ADA) gene into umbilical cord blood CD34(+) cells was started in 1993. ADA-containing peripheral blood mononuclear cells (PBMCs) have persisted in patients from this trial, with T lymphocytes showing the highest prevalence of gene marking. To gain a greater understanding of the nature and number of the transduced cells that were engrafted, we used linear amplification-mediated PCR (LAM-PCR) to identify clonal vector proviral integrants. In one patient, a single vector integrant was predominant in T lymphocytes at a stable level over most of the eight-year time span analyzed and was also detected in some myeloid samples. T-cell clones with the predominant integrant, isolated after eight years, showed multiple patterns of T-cell receptor (TCR) gene rearrangement, indicating that a single pre-thymic stem or progenitor cell served as the source of the majority of the gene-marked cells over an extended period of time. It is important to distinguish the stable pattern of monoclonal gene marking that we observed here from the progressive increase of a T-cell clone with monoclonal gene marking that results from leukemic transformation, as observed in two subjects in a clinical trial of gene therapy for X-linked severe combined immunodeficiency (SCID).


Asunto(s)
Adenosina Desaminasa/genética , Antígenos CD34 , Sangre Fetal , Técnicas de Transferencia de Gen , Inmunodeficiencia Combinada Grave/terapia , Transducción Genética , Células Clonales , Vectores Genéticos , Humanos , Recién Nacido , Reacción en Cadena de la Polimerasa , Retroviridae/genética , Linfocitos T/metabolismo
13.
Mol Ther ; 6(5): 645-52, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12409263

RESUMEN

Vectors based on the feline immunodeficiency virus (FIV) have been developed as an alternative to those based on another lentivirus, human immunodeficiency virus-1 (HIV-1), because of theoretical safety advantages. We compared the efficiency of gene transfer and expression in human and feline hematopoietic progenitors using second-generation HIV-1 and FIV-based vectors. Vector pairs were tested using either human cytomegalovirus or murine phospho-glycerate kinase (PGK) internal promoters and were pseudotyped with the vesicular stomatitis virus G protein (VSV-G). Vector proviral copy numbers were similar in human and feline hematopoietic primary cells and cell lines transduced by HIV-1 or FIV vectors, demonstrating that both vectors are able to transfer genes efficiently to these cell types. HIV-1 vectors were well expressed in human primary hematopoietic cells and cell lines. However, transgene expression from FIV vectors was almost undetectable in human hematopoietic cells. In contrast, the FIV vector was expressed well in primary hematopoietic feline cells and human non-hematopoietic cells, demonstrating that low transgene expression from the FIV vector is a phenomenon specific to human hematopoietic cells. Northern blot analysis demonstrated decreased vector transcript levels in human CEM cells transduced with FIV relative to cells transduced with HIV-1, despite high vector copy numbers. No evidence of vector transcript instability was seen in studies of transduced CEM cells treated with actinomycin D. We conclude that FIV vectors can transfer genes into human hematopoietic cells as effectively as HIV-1 vectors, but that unknown elements in the current FIV backbone inhibit expression from FIV vectors in human hematopoietic cells.


Asunto(s)
Técnicas de Transferencia de Gen , Vectores Genéticos , Células Madre Hematopoyéticas/metabolismo , Virus de la Inmunodeficiencia Felina/genética , Antígenos CD34/biosíntesis , Northern Blotting , Células de la Médula Ósea/citología , Citomegalovirus/genética , Sangre Fetal/metabolismo , Terapia Genética/métodos , Proteínas Fluorescentes Verdes , VIH-1/genética , Humanos , Lentivirus/genética , Proteínas Luminiscentes/metabolismo , Fosfoglicerato Quinasa/genética , ARN/metabolismo , Factores de Tiempo , Transgenes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...