Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Gene Ther ; 17(1): 50-60, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19759566

RESUMEN

Salivary glands are potentially useful target sites for multiple clinical applications of gene transfer. Previously, we have shown that serotype 2 adeno-associated viral (AAV2) vectors lead to stable gene transfer in the parotid glands of rhesus macaques. As AAV5 vectors result in considerably greater transgene expression in murine salivary glands than do AAV2 vectors, herein we have examined the use of AAV5 vectors in macaques at two different doses (n = 3 per group; 10(10) or 3 x 10(11) particles per gland). AAV5 vector delivery, as with AAV2 vectors, led to no untoward clinical, hematological or serum chemistry responses in macaques. The extent of AAV5-mediated expression of rhesus erythropoietin (RhEpo) was dose-dependent and similar to that seen with an AAV2 vector. However, unlike results with the AAV2 vector, AAV5 vector-mediated RhEpo expression was transient. Maximal expression peaked at day 56, was reduced by approximately 80% on day 84 and thereafter remained near background levels until day 182 (end of experiment). Quantitative PCR studies of high-dose vector biodistribution at this last time point showed much lower AAV5 copy numbers in the targeted parotid gland (approximately 1.7%) than found with the same AAV2 vector dose. Molecular analysis of the conformation of vector DNA indicated a markedly lower level of concatamerization for the AAV5 vector compared with that of a similar AAV2 vector. In addition, cellular immunological studies suggest that host response differences may occur with AAV2 and AAV5 vector delivery at this mucosal site. The aggregate data indicate that results with AAV5 vectors in murine salivary glands apparently do not extend to macaque glands.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Glándula Parótida/metabolismo , Animales , Terapia Genética/métodos , Macaca mulatta , Glándula Parótida/virología , Transducción Genética , Transgenes
2.
Gene Ther ; 17(6): 799-804, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20237508

RESUMEN

Adverse events linked to perturbations of cellular genes by vector insertion reported in gene therapy trials and animal models have prompted attempts to better understand the mechanisms directing viral vector integration. The integration profiles of vectors based on MLV, ASLV, SIV and HIV have all been shown to be non-random, and novel vectors with a safer integration pattern have been sought. Recently, we developed a producer cell line called CatPac that packages standard MoMLV vectors with feline leukemia virus (FeLV) gag, pol and env gene products. We now report the integration profile of this vector, asking if the FeLV integrase and capsid proteins could modify the MoMLV integration profile, potentially resulting in a less genotoxic pattern. We transduced rhesus macaque CD34+ hematopoietic progenitor cells with CatPac or standard MoMLV vectors, and determined their integration profile by LAM-PCR. We obtained 184 and 175 unique integration sites (ISs) respectively for CatPac and standard MoMLV vectors, and these were compared with 10 000 in silico-generated random IS. The integration profile for CatPac vector was similar to MoMLV and equally non-random, with a propensity for integration near transcription start sites and in highly dense gene regions. We found an IS for CatPac vector localized 715 nucleotides upstream of LMO-2, the gene involved in the acute lymphoblastic leukemia developed by X-SCID patients treated by gene therapy using MoMLV vectors. In conclusion, we found that replacement of MoMLV env, gag and pol gene products with FeLV did not alter the basic integration profile. Thus, there appears to be no safety advantage for this packaging system. However, considering the stability and efficacy of CatPac vectors, further development is warranted, using potentially safer vector backbones, for instance those with a SIN configuration.


Asunto(s)
Técnicas de Transferencia de Gen/efectos adversos , Vectores Genéticos/efectos adversos , Células Madre Hematopoyéticas/virología , Integrasas/genética , Virus de la Leucemia Felina/genética , Virus de la Leucemia Murina de Moloney/genética , Integración Viral , Animales , Cápside , Proteínas de la Cápside/genética , Virus de la Leucemia Felina/metabolismo , Macaca mulatta , Transducción Genética
3.
J Exp Med ; 182(6): 2037-43, 1995 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-7500049

RESUMEN

We have generated immunodeficient scid-/scid- (SCID)-transgenic mice expressing the genes for human interleukin 3, granulocyte/macrophage-colony stimulating factor, and stem cell factor. We have compared engraftment and differentiation of human hematopoietic cells in transgenic SCID mice with two strains of nontransgenic SCID mice. Human bone marrow cells carrying the CD34 antigen or human umbilical cord blood were injected into sublethally irradiated recipients. Human DNA was detected by polymerase chain reaction in peripheral blood and bone marrow of 14 of 28 transgenic SCID mice after transplantation, but in only 2 of 15 nontransgenic SCID littermates at a 10-fold lower level. Bone marrow cultures 8 wk after transplantation of cord blood gave rise to human burst-forming unit erythroid, colony-forming unit granulocyte/macrophage, or granulocyte/erythroid/macrophage/megakaryocyte colonies. Engraftment was observed for up to 6 mo in transgenic SCID mice, twice as long as nontransgenic littermates or previous studies in which transplanted SCID mice were given daily injections of growth factors. We conclude that the level and duration of engraftment of human cells in SCID mice can be improved by expression of human cytokine transgenes and that transgenic SCID mice are an efficient model system for the study of human hematopoiesis.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Interleucina-3/fisiología , Factor de Células Madre/fisiología , Animales , Secuencia de Bases , Cartilla de ADN/química , Sangre Fetal/citología , Humanos , Antígenos Comunes de Leucocito/análisis , Receptores de Lipopolisacáridos/análisis , Ratones , Ratones SCID , Ratones Transgénicos , Datos de Secuencia Molecular , Trasplante Heterólogo
4.
Br J Cancer ; 102(11): 1636-44, 2010 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-20424609

RESUMEN

BACKGROUND: Side population (SP) fraction cells, identified by efflux of Hoechst dye, are present in virtually all normal and malignant tissues. The relationship between SP cells, drug resistance and cancer stem cells is poorly understood. Small-cell lung cancer (SCLC) is a highly aggressive human tumour with a 5-year survival rate of <10%. These features suggest enrichment in cancer stem cells. METHODS AND RESULTS: We examined several SCLC cell lines and found that they contain a consistent SP fraction that comprises <1% of the bulk population. Side population cells have higher proliferative capacity in vitro, efficient self-renewal and reduced cell surface expression of neuronal differentiation markers, CD56 and CD90, as compared with non-SP cells. Previous reports indicated that several thousand SP cells from non-small-cell lung cancer are required to form tumours in mice. In contrast, as few as 50 SP cells from H146 and H526 SCLC cell lines rapidly reconstituted tumours. Whereas non-SP cells formed fewer and slower-growing tumours, SP cells over-expressed many genes associated with cancer stem cell and drug resistance: ABCG2, FGF1, IGF1, MYC, SOX1/2, WNT1, as well as genes involved in angiogenesis, Notch and Hedgehog pathways. CONCLUSIONS: Side population cells from SCLC are highly enriched in tumourigenic cells and are characterised by a specific stem cell-associated gene expression signature. This gene signature may be used for development of targeted therapies for this rapidly fatal tumour.


Asunto(s)
Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Células Madre Neoplásicas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/patología , Animales , Antígenos de Superficie/análisis , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Separación Celular , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/patología , Estudios de Validación como Asunto
5.
Oral Dis ; 16(3): 269-77, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20374510

RESUMEN

OBJECTIVES: Salivary glands are useful target organs for local and systemic gene therapeutics. For such applications, the regulation of transgene expression is important. Previous studies by us in murine submandibular glands showed that a rapamycin transcriptional regulation system in a single serotype 2, adeno-associated viral (AAV2) vector was effective for this purpose. This study evaluated if such a vector was similarly useful in rhesus macaque parotid glands. METHODS: A recombinant AAV2 vector (AAV-TF-RhEpo-2.3w), encoding rhesus erythropoietin (RhEpo) and a rapamycin-inducible promoter, was constructed. The vector was administered to macaques at either of two doses [1.5 x 10(11) (low dose) or 1.5 x 10(12) (high dose) vector genomes] via cannulation of Stensen's duct. Animals were followed up for 12-14 weeks and treated at intervals with rapamycin (0.1 or 0.5 mg kg(-1)) to induce gene expression. Serum chemistry, hematology, and RhEpo levels were measured at interval. RESULTS: AAV-TF-RhEpo-2.3w administration led to low levels of rapamycin-inducible RhEpo expression in the serum of most macaques. In five animals, no significant changes were seen in serum chemistry and hematology values over the study. One macaque, however, developed pneumonia, became anemic and subsequently required euthanasia. After the onset of anemia, a single administration of rapamycin led to significant RhEpo production in this animal. CONCLUSION: Administration of AAV-TF-RhEpo-2.3w to macaque parotid glands was generally safe, but led only to low levels of serum RhEpo in healthy animals following rapamycin treatment.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/administración & dosificación , Glándula Parótida/metabolismo , Sirolimus/farmacología , Transducción Genética , Adenoviridae/genética , Animales , Relación Dosis-Respuesta a Droga , Eritropoyetina/sangre , Eritropoyetina/genética , Eritropoyetina/metabolismo , Macaca mulatta , Masculino , Regiones Promotoras Genéticas , Proteínas Recombinantes , Transgenes
6.
Science ; 245(4925): 1493-6, 1989 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-2789432

RESUMEN

Autocrine growth due to dysregulated growth factor production may have a role in the development of neoplasia. Whether autocrine growth is stimulated by growth factor secretion in an autocrine loop or by intracellular binding of the growth factor to a receptor has been unclear. The carboxyl-terminus coding sequence for murine interleukin-3 (IL-3) was extended with an oligonucleotide encoding a four-amino acid endoplasmic reticulum retention signal. IL-3-dependent hematopoietic cells became growth factor-independent when the modified IL-3 gene was introduced by retroviral gene transfer, despite lack of secretion of the modified IL-3. Hence autocrine growth can occur as a result of the intracellular action of a growth factor and this mechanism may be important in neoplastic and normal cells.


Asunto(s)
División Celular , Interleucina-3/fisiología , Animales , División Celular/efectos de los fármacos , Transformación Celular Neoplásica , Células Cultivadas , Células Clonales , Interleucina-3/genética , Interleucina-3/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas Recombinantes de Fusión/farmacología
7.
J Clin Invest ; 86(2): 592-9, 1990 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-2384605

RESUMEN

Interleukin 6 (IL-6) is an important regulator of the acute phase response, T cell function, and terminal B cell differentiation. Excessive or inappropriate production of this cytokine may be involved in a variety of autoimmune and neoplastic disorders. To investigate the consequences of dysregulated synthesis of IL-6 in vivo, a high-titer recombinant retroviral vector produced in psi-2 packaging cells was used to introduce the coding sequences of murine IL-6 into mouse hematopoietic cells. Congenitally anemic W/Wv mice reconstituted with bone marrow cells transduced with the retroviral vector developed a syndrome characterized by anemia, transient granulocytosis, hypoalbuminemia, and polyclonal hypergammaglobulinemia, with marked splenomegaly and peripheral lymphadenopathy. Extensive plasma cell infiltration of lymph nodes, spleen, liver, and lung was noted. The similarity of these findings to those of multicentric Castleman's disease, taken together with the observation that lymph nodes from these patients elaborate large amounts of this cytokine, suggest that the inappropriate synthesis of IL-6 has a primary role in the pathogenesis of this systemic lymphoproliferative disorder.


Asunto(s)
Enfermedad de Castleman/fisiopatología , Interleucina-6/genética , Animales , Southern Blotting , Regulación de la Expresión Génica , Reordenamiento Génico de Linfocito B , Genes de Inmunoglobulinas , Vectores Genéticos , Células Madre Hematopoyéticas/fisiología , Riñón/patología , Recuento de Leucocitos , Ratones , Ratones Endogámicos , ARN Mensajero/genética , Retroviridae/genética , Transfección
8.
J Clin Invest ; 108(3): 447-55, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11489938

RESUMEN

Recent reports suggest that cells in active cell cycle have an engraftment defect compared with quiescent cells. We used nonhuman primates to investigate this finding, which has direct implications for clinical transplantation and gene therapy applications. Transfer of rhesus CD34(+) cells to culture in stem cell factor (SCF) on the CH-296 fibronectin fragment (FN) after 4 days of culture in stimulatory cytokines maintained cell viability but decreased cycling. Using retroviral marking with two different gene transfer vectors, we compared the engraftment potential of cytokine-stimulated cells versus those transferred to nonstimulatory conditions (SCF on FN alone) before reinfusion. In vivo competitive repopulation studies showed that the level of marking originating from the cells continued in culture for 2 days with SCF on FN following a 4-day stimulatory transduction was significantly higher than the level of marking coming from cells transduced for 4 days and reinfused without the 2-day culture under nonstimulatory conditions. We observed stable in vivo overall gene marking levels of up to 29%. This approach may allow more efficient engraftment of transduced or ex vivo expanded cells by avoiding active cell cycling at the time of reinfusion.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Animales , Antígenos CD34/metabolismo , Secuencia de Bases , Ciclo Celular/efectos de los fármacos , Transformación Celular Viral , Células Cultivadas , Citocinas/farmacología , Cartilla de ADN/genética , Fibronectinas/farmacología , Terapia Genética , Vectores Genéticos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/inmunología , Humanos , Macaca mulatta , Fragmentos de Péptidos/farmacología , Retroviridae/genética , Factor de Células Madre/farmacología , Transducción Genética
9.
Mol Cell Biol ; 9(2): 798-808, 1989 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-2565534

RESUMEN

A high-titer, recombinant retroviral vector produced in psi 2 packaging cells has been used to introduce the murine interleukin-3 (IL-3) gene into mouse hematopoietic cells. Integration and expression of the IL-3 gene was observed in spleen foci from which could be derived factor-independent, continuously proliferating cell lines. Irradiated or genetically anemic W/Wv recipients of infected hematopoietic cells developed a myeloproliferative syndrome characterized by a marked elevation in leukocyte count, bone marrow hyperplasia, and enlargement of the liver and spleen. The syndrome reflected proliferation of one or more stem cell clones, the progeny of which were capable of repopulating secondary recipients. One animal developed the syndrome primarily by a paracrine mechanism. Endogenous IL-3 production caused amplification of hematopoietic cells but did not appear to alter the maturational or self-renewal potential of these cells.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Interleucina-3/genética , Trastornos Mieloproliferativos/etiología , Animales , Regulación de la Expresión Génica , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Interleucina-3/biosíntesis , Leucocitosis/etiología , Ratones , Ratones Endogámicos , Trastornos Mieloproliferativos/sangre , Trastornos Mieloproliferativos/patología , Retroviridae/genética , Síndrome , Transfección
10.
Bone Marrow Transplant ; 37(4): 353-8, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16400343

RESUMEN

Systemic mastocytosis (SM) is a disease characterized by tissue infiltration of neoplastic mast cells originating from hematopoietic stem cells. Patients with advanced SM have a poor prognosis, and there is no mast cell ablative therapy available for most patients who carry an activating point mutation in the c-kit gene. We report results of a prospective study evaluating the safety, engraftment, and possibility of inducing a graft-versus-mast cell (GvMC) effect after allogeneic nonmyeloablative hematopoietic cell transplantation (HCT) from an HLA-identical sibling. Three patients with advanced SM were transplanted. All achieved complete donor T cell chimerism followed by clinical evidence for GvMC effect. However, all patients experienced disease progression with the longest response duration of 39 months. The GvMC effect can be observed after nonmyeloablative HCT with limited efficacy. Effective cytoreductive therapy prior to HCT may be required for long-term disease control and cure.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Mastocitosis Sistémica/terapia , Adulto , Progresión de la Enfermedad , Femenino , Supervivencia de Injerto , Enfermedad Injerto contra Huésped/etiología , Enfermedad Injerto contra Huésped/terapia , Antígenos HLA/análisis , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Masculino , Mastocitosis Sistémica/inmunología , Mastocitosis Sistémica/patología , Persona de Mediana Edad , Proyectos Piloto , Estudios Prospectivos , Recurrencia , Hermanos , Tasa de Supervivencia , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo , Resultado del Tratamiento
11.
Exp Hematol ; 21(4): 585-91, 1993 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7681785

RESUMEN

Retroviral-mediated gene transfer has been shown to be a feasible method for the introduction of new genes into bone marrow hematopoietic stem cells. We have investigated the application of this technology to primitive CD34-enriched human peripheral blood cells as a potential alternative stem cell source. Bone marrow (BM) and peripheral blood (PB) CD34-enriched cells from normal volunteers and patients with multiple myeloma were exposed to retroviral vectors containing the neomycin-resistance gene and gene transfer efficiency into colony-forming unit colonies (CFU-C) and CD34+ cells was assessed by polymerase chain reaction (PCR). Peripheral blood was a target equally efficient to BM, and PB cells mobilized with chemotherapy and growth factors were also shown to take up retroviral vectors readily. Conditions favoring gene transfer were investigated, and exposure of cells to interleukin-3 (IL-3), interleukin-6 (IL-6), and stem cell factor (SCF) during a 72-hour transduction was found to be most effective. The use of PB stem cells as targets for gene transfer could allow repeated collections and transductions, with obvious advantages over a single BM collection.


Asunto(s)
Antígenos CD/análisis , Células Sanguíneas , Células Madre Hematopoyéticas/citología , Retroviridae/genética , Antígenos CD34 , Secuencia de Bases , Separación Celular , Vectores Genéticos , Humanos , Técnicas In Vitro , Datos de Secuencia Molecular , Mieloma Múltiple/patología , Oligodesoxirribonucleótidos/química , Reacción en Cadena de la Polimerasa , Transfección
12.
Exp Hematol ; 27(5): 895-903, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10340406

RESUMEN

Several lines of evidence suggest that overexpression of interferon gamma (IFN-gamma) in the marrow microenvironment may play a role in the pathogenesis of marrow suppression in aplastic anemia. We previously showed that overexpression of IFN-gamma by marrow stromal cells inhibits human long-term culture initiating cell activity assayed in vitro to a much greater degree than the addition of soluble IFN-gamma. The effect of IFN-gamma on true repopulating stem cells assayed in vivo has not been studied previously. We compared the effect of co-culture of murine marrow cells in the presence of stromal cells transduced with a retroviral vector expressing murine IFN-gamma vs stromal cells transduced with a control neo vector. Using a murine congenic competitive repopulation assay, there was significantly less long-term repopulating stem cell activity remaining after culture on mIFN-gamma-expressing stroma as compared to control stroma. We also investigated the effect of directly transducing murine bone marrow cells with the mIFN-gamma or control vector. Marrow cells transduced with either vector were transplanted into W/Wv recipient mice. The percentage of vector-containing cells in the mIFN-gamma mice was significantly lower than in the control mice, suggesting that mIFN-gamma-transduced primitive cells may not have survived culture, or that mIFN-gamma directly decreases gene transfer into repopulating cells. Despite no significant differences in white or red blood cells in the mice transplanted with the mIFN-gamma-transduced cells, the number of bone marrow colony-forming unit-C 16 weeks after transplantation was significantly lower in the IFN-gamma group. These data indicate that ectopic or overexpression of mIFN-gamma, especially by marrow microenvironmental elements, may have a marked effect on primitive hematopoiesis as assayed in vivo.


Asunto(s)
División Celular/genética , Células Madre Hematopoyéticas/citología , Interferón gamma/genética , Células del Estroma/metabolismo , Animales , Secuencia de Bases , Técnicas de Cocultivo , Cartilla de ADN , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Recombinantes/genética , Retroviridae/genética , Transducción Genética
13.
Exp Hematol ; 21(9): 1245-54, 1993 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-8330649

RESUMEN

Myeloid leukemias have been shown to secrete as well as respond to cytokines such as interleukin-3 (IL-3) with an increased growth rate and may therefore become self-stimulatory through an external autocrine mechanism. In vitro evidence that IL-3 is functional within the intracellular compartment has been obtained through modification of the murine IL-3 gene to encode for the amino acids SEKDEL on the carboxyl terminus of the protein, resulting in preferential intracellular retention. The ability of bone marrow-derived hematopoietic progenitor cells to increase their proliferative capacity through intracellular mechanisms was investigated in vivo using retroviruses containing the wild-type or SEKDEL-modified IL-3 gene, transcriptionally regulated by the retroviral long terminal repeat (LTR) or by the SV40 early promoter, in lethally irradiated, bone marrow-reconstituted mice. Bone marrow cells exposed to the N2KDEL virus containing the SEKDEL-modified IL-3 gene were shown by bioassay to retain large amounts of IL-3 intracellularly, and the presence of an integrated provirus containing the SEKDEL sequences was demonstrated by polymerase chain reaction (PCR) in the spleen and bone marrow of these animals. Transduction with all four types of IL-3 viruses resulted in dramatic increases in the circulating white blood cell (WBC) count; this myeloproliferative state occurred within several weeks following bone marrow transplantation (BMT), when viruses expressing the IL-3 or modified gene were under transcriptional regulation of the viral LTR, and approximately 2 months post-BMT, when they were under control of the SV40 internal promoter. Serum levels of IL-3 were measured in transplanted animals and found to be markedly increased in each case in which WBC elevation was observed, including mice receiving marrow transduced with constructs containing the IL-3 gene modified for intracellular retention. No animals were observed in which myeloproliferation occurred without secretion. From these experiments, it seems unlikely that exclusively intracellular mechanisms are a major contributor to the development of the myeloproliferative syndrome observed in these animals.


Asunto(s)
Interleucina-3/sangre , Interleucina-3/genética , Trastornos Mieloproliferativos/genética , Retroviridae/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células de la Médula Ósea , Trasplante de Médula Ósea/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Fluorouracilo/farmacología , Líquido Intracelular/química , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Trastornos Mieloproliferativos/patología , Retroviridae/genética , Transducción Genética/efectos de los fármacos
14.
Hum Gene Ther ; 12(6): 607-17, 2001 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-11426461

RESUMEN

Transfer of genes into hematopoietic stem cells or primary lymphocytes has been a primary focus of the gene therapy field for more than a decade because of the wide variety of congenital and acquired diseases that potentially could be cured by successful gene transfer into these cell populations. However, despite success in murine models and in vitro, progress has been slow, and early clinical trials were disappointing due to inefficient gene transfer into long-term repopulating cells. The unique predictive value of nonhuman primate or other large animal models has become more apparent, and major advances in gene transfer efficiency have been made by utilizing these powerful but expensive and complex systems. This review summarizes more recent findings from nonhuman primate investigations focusing on hematopoietic stem cells or lymphocytes as target populations, and highlights specific preclinical issues, including safety. Results from studies using standard retroviral vectors, lentiviral vectors, adenoviral vectors, and adeno-associated viral vectors are discussed. Judicious application of these models should continue to be a priority, and advances should now be tested in proof-of-concept clinical trials.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Células Madre Hematopoyéticas/fisiología , Macaca/fisiología , Modelos Animales , Animales , Evaluación Preclínica de Medicamentos , Vectores Genéticos , Células Madre Hematopoyéticas/virología , Humanos , Linfocitos T/fisiología , Linfocitos T/virología
15.
Hum Gene Ther ; 8(11): 1313-9, 1997 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-9295126

RESUMEN

Green fluorescent protein (GFP) has been used as a reporter molecule for gene expression because it fluoresces green after blue-light excitation. Inclusion of this gene in a vector could allow rapid, nontoxic selection of successfully transduced cells. However, many attempts by our laboratory to isolate stable retroviral producer cell clones secreting biologically active vectors containing either the highly fluorescent S65T-GFP mutant or humanized GFP have failed. Vector plasmids containing various forms of GFP and the neomycin resistance gene were transfected into three different packaging cell lines and fluorescence was observed for several days, but stable clones selected with G418 no longer fluoresced. Using confocal microscopy, the brightest cells were observed to contract and die within a matter of days. RNA slot-blot analysis of retroviral producer supernatants showed no viral production from the GFP plasmid-transfected clones, although all clones derived after transfection with an identical retroviral construct not containing GFP produced virus. Genomic Southern analysis of the GFP-transduced clones showed a much higher probability of rearrangement of the priviral sequences than in the control non-GFP clones. Overall, 18/34 S65T-GFP clones and 17/33 humanized-GFP clones had rearrangements, whereas 2/15 control non-GFP clones had rearrangements. Hence, producer cells expressing high levels of these GFP genes seem to be selected against, with stable clones undergoing major rearrangements or other mutations that both abrogate GFP expression and prevent vector production. These observations indicate that GFP may not be an appropriate reporter gene for gene transfer applications in our vector/packaging system.


Asunto(s)
Genes Reporteros/genética , Vectores Genéticos/metabolismo , Proteínas Luminiscentes/genética , Retroviridae/genética , Animales , Southern Blotting , Línea Celular , Técnicas de Transferencia de Gen , Genes Virales/genética , Marcadores Genéticos , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente , Plásmidos , ARN Viral/análisis , Retroviridae/fisiología , Replicación Viral
16.
Hum Gene Ther ; 7(1): 33-8, 1996 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-8825866

RESUMEN

The use of defined or serum-free culture conditions during retroviral transduction of hematopoietic cells would be desirable for standardization and safety reasons, as well as potentially allowing greater expansion of progenitor cells. Retroviral vector supernatants were concentrated and purified via tangential flow filtration polyethylene glycol (PEG)-precipitation, and ultracentrifugation, allowing serum-free transductions at standard multiplicities of infection (moi). Protein content of transductions using these concentrated vectors was 5-6 logs lower than in standard transductions. Transduction efficiencies of these concentrated vector preparations added back to serum-free or serum-containing media were equivalent to standard retroviral supernatant transductions of CD34-enriched progenitors. Absolute progenitor (CFU-C) numbers at the end of transduction were higher in serum-free + concentrated virus transductions, as opposed to transductions in standard vector supernatants containing fetal calf serum.


Asunto(s)
Antígenos CD34/genética , Expresión Génica , Vectores Genéticos/genética , Células Madre Hematopoyéticas/inmunología , Secuencia de Bases , Medio de Cultivo Libre de Suero , Cartilla de ADN , Células Madre Hematopoyéticas/citología , Humanos , Datos de Secuencia Molecular , Retroviridae/genética , Transfección
17.
Hum Gene Ther ; 9(8): 1157-64, 1998 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-9625254

RESUMEN

The neomycin phosphotransferase (neo) gene is one of the most common marker genes used in gene transfer experimentation, but potential effects of neo gene expression in vivo have not been systematically investigated. Several early clinical retroviral gene transfer studies have suggested that neo gene expression could have deleterious effects on hematopoiesis, owing to a discrepancy between the level of neo-marked transduced marrow progenitor cells compared with mature circulating progeny cells posttransplantation (Brenner et al., 1993; Kohn et al., 1995; Brenner, 1996b). We examined the long-term in vivo repopulating ability of bone marrow from transgenic mice expressing neo from a strong constitutive promoter using a competitive repopulation assay. Different ratios of neo transgenic and wild-type congenic marrow cells were cotransplanted into W/Wv recipient mice. The percentages of blood cells containing the neo transgene in each group of recipient mice monitored for 4 months posttransplantation closely matched the input ratios of neo transgenic to congenic control marrow cells. Similar concordances of engraftment with input ratios of neo transgenic cells were also found in spleen, thymus, and whole marrow of recipient mice at 4 months posttransplantation. Analysis of the beta-hemoglobin phenotype (beta(single) for the neo transgenic and C57 control cells and beta(diffuse) for the congenic competitor HW80 cells) in recipients confirmed erythroid repopulation from neo transgenic marrow cells at levels matching the input ratios. We conclude that hematopoietic cells expressing neo had no engraftment or maturation defects detectable in vivo. These results suggest that the low-level contribution of vector-marked cells to circulating populations in clinical trials is not due to direct deleterious effects of neo gene expression on hematopoiesis.


Asunto(s)
Antibacterianos/farmacología , Trasplante de Médula Ósea , Regulación de la Expresión Génica/fisiología , Células Madre Hematopoyéticas/metabolismo , Kanamicina Quinasa/genética , Neomicina/farmacología , Animales , Farmacorresistencia Microbiana/genética , Estudios de Evaluación como Asunto , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Regiones Promotoras Genéticas
18.
Hum Gene Ther ; 10(4): 633-40, 1999 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-10094206

RESUMEN

Reports of 1- to 2-log higher gene transfer levels in purified CD34+ cells or marrow CFU compared with levels in mature circulating blood cells after transplantation of retrovirally transduced primitive human hematopoietic cells have resulted in concern that transduced progenitors do not contribute proportionally to ongoing hematopoiesis (Kohn et al., 1995; Brenner, 1996). To study the issue in a relevant large animal, we analyzed samples of mature blood cells, marrow CD34-enriched cells and marrow CD34-depleted cells, and marrow CFU from a cohort of 11 rhesus transplanted with retrovirally transduced cells and followed for up to 5.5 years. They were transplanted with CD34-enriched bone marrow (BM) or G-CSF/SCF-mobilized peripheral blood (PB) cells transduced with vectors containing either neo, human glucocerebrosidase, or murine adenosine deaminase genes. There were no significant differences between the levels of vector sequences found in BM CD34+ cells, BM CD34- cells, PB granulocytes, or PB mononuclear cells (MNCs) in any animal. In four animals transplanted with SCF/G-CSF-primed BM cells and analyzed 3-6 months posttransplantation, the percentage of CFU containing the neo vector appeared to be 1 log higher than the representation of marked cells in the PB of these animals, but this discrepancy did not persist at time points greater than 6 months posttransplantation. The level of CFU marking was no higher than PB granulocyte or MNC marking at any time points in the other animals. Low levels of mature gene-modified cells probably reflect poor transduction of repopulating stem cells, not a block in differentiation or specific immune rejection of mature cells. This study represents the longest follow-up of primates transplanted with transduced hematopoietic cells, and it is encouraging that the levels of vector-containing cells appear stable for up to 5 years.


Asunto(s)
Antígenos CD34/inmunología , Células Sanguíneas/metabolismo , Técnicas de Transferencia de Gen/normas , Células Madre Hematopoyéticas/metabolismo , Animales , Secuencia de Bases , Cartilla de ADN , Resistencia a Medicamentos/genética , Marcadores Genéticos , Humanos , Macaca mulatta , Neomicina/farmacología , Transducción Genética
19.
Hum Gene Ther ; 10(11): 1783-90, 1999 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-10446918

RESUMEN

The ability to engraft significant numbers of genetically modified hematopoietic stem and progenitor cells without the requirement for fully myeloablative conditioning therapy is a highly desirable goal for the treatment of many nonmalignant hematologic disorders. The aims of this study were to examine, in nonhuman primates (rhesus), (1) the effects of pretreatment of host animals with cytokines (G-CSF and SCF), i.e., before nonmyeloablative irradiation, on the degree and duration of neo gene marking of circulating leukocytes after autologous cell reinfusion and (2) to compare transduction of primitive hematopoietic target cells in the presence of our standard transduction cytokine combination of IL-3, IL-6, and stem cell factor (SCF) and in the presence of an alternative combination containing SCF, G-CSF, and the thrombopoietin analog MGDF. Cytokine-mobilized rhesus peripheral blood progenitor/stem cells (PBSCs) were enriched for CD34+ cells and transduced with neo vectors (either G1Na or LNL6) for 96 hr in cultures containing rhIL-3, rhIL-6, and rhSCF or MGDF, rhSCF, and rhG-CSF and cryopreserved. Four animals underwent minimal myeloablative conditioning with 500 cGy irradiation with or without pretreatment with SCF and G-CSF, followed by reinfusion of the cryopreserved cells on the subsequent day. Neutrophil nadirs (< or =500/mm3) were 0-3 days in duration; there were no significant periods of severe thrombocytopenia. Marking of circulating granulocytes and mononuclear cells was extensive and durable in all animals (exceeding 12% in the mononuclear cells of one animal) and persisted beyond the final sampling time in all animals (up to 33 weeks). No difference in extent or duration of marking was attributable to either cytokine presensitization of recipients prior to irradiation, or to the substitution of MDGF and G-CSF for IL-3 and IL-6 during transduction.


Asunto(s)
Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/efectos de la radiación , Trasplante de Células Madre Hematopoyéticas , Retroviridae/genética , Acondicionamiento Pretrasplante , Animales , Antibacterianos/farmacología , Antígenos CD34/metabolismo , Resistencia a Medicamentos/genética , Técnicas de Transferencia de Gen , Marcadores Genéticos , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos/farmacología , Granulocitos/inmunología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de la radiación , Células Madre Hematopoyéticas/virología , Leucocitos Mononucleares/inmunología , Macaca mulatta , Neomicina/farmacología , Reacción en Cadena de la Polimerasa , Dosis de Radiación , Factor de Células Madre/farmacología , Transducción Genética
20.
Hum Gene Ther ; 12(13): 1663-72, 2001 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-11535169

RESUMEN

Many nonmalignant hematologic disorders could potentially be treated by genetic correction of as few as 5-10% of target lineage cells. However, immune system clearance of cells expressing gene products perceived as foreign could be limiting. There is evidence that tolerance to foreign proteins can result when myeloablative conditioning is used, but this limits the overall applicability of such techniques. Therefore, we sought to evaluate the engraftment of hematopoietic stem cells carrying a foreign transgene after low-dose irradiation by comparing in vivo survival of murine long-term repopulating cells (LTRC) transduced with either a retroviral vector expressing the bacterial neomycin phosphotransferase gene (neo) or a vector containing neo gene sequences but modified to prevent protein expression (nonexpression). First, marrow cells from congenic donors were transduced with either vector and transplanted into recipients treated with standard dose irradiation of 800 rads. High-level engraftment and gene marking resulted, without differences in the marking levels or pattern of persistence of the cells between cells transduced with either vector. Low-dose irradiation at 100 rads was tested using higher cell doses. Marking levels as high as 10% overall were obtained, again with no differences between mice receiving cells transduced with the neo versus the nonexpression vectors. To investigate a potentially more immunogenic protein, marrow cells were transduced with a vector containing the green fluorescent protein (GFP) gene, and their persistence was studied in recipient mice receiving 100 rads. Stable GFP expression in 5-10% of circulating cells was observed long term. We conclude that even with very low dose conditioning, engraftment by genetically modified LTRC cells at clinically significant levels can be achieved without evidence for clearance of cells known to be expressing immunogenic proteins.


Asunto(s)
Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Mieloides/efectos de la radiación , Retroviridae/genética , Transducción Genética , Transgenes/genética , Animales , Animales Congénicos , Línea Celular , Supervivencia Celular , Femenino , Citometría de Flujo , Expresión Génica , Vectores Genéticos/genética , Supervivencia de Injerto/genética , Supervivencia de Injerto/inmunología , Proteínas Fluorescentes Verdes , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/efectos de la radiación , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/inmunología , Ratones , Ratones Endogámicos C57BL , Quimera por Radiación/genética , Quimera por Radiación/inmunología , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda