Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Hum Gene Ther ; 9(15): 2243-51, 1998 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-9794208

RESUMEN

We previously demonstrated that severe graft-versus-host disease (GvHD), associated with the therapeutic infusion of donor lymphocytes after allogeneic marrow transplantation (BMT), can be efficiently controlled by the SFCMM-2-mediated expression of the herpes simplex virus thymidine kinase (HSV-tk) suicide gene into the allogeneic lymphocytes. This was achieved by selective elimination of transduced lymphocytes by ganciclovir (GCV) infusion. Despite the positive results of the pilot clinical trial, two vector-related limitations were observed. The induction of a strong immune response against genetically modified cells was observed in two patients. In addition, the only patient who developed chronic GvHD showed only partial response to ganciclovir treatment. In an attempt to overcome these limitations, we developed a new generation of vectors. The neomycin resistance (neoR) gene was removed from the SFCMM-3 and SFCMM-4 retroviral vectors. These vectors are less immunogenic and able to confer higher ganciclovir sensitivity to transduced human lymphocytes. All the vectors carry a modified form of the low-affinity nerve growth factor receptor cDNA, as cell surface selectable marker (deltaLNGFR). The vectors were compared in terms of gene transfer efficiency, and ability to confer high and specific sensitivity to ganciclovir. The SFCMM-3 vector, carrying the entire HSV-tk gene driven by the LTR promoter, allows efficient transduction of human T lymphocytes and confers the highest GCV sensitivity to transduced lymphocytes with a high and a low proliferation index. The expression of the deltaLNGFR marker allows an easier in vitro manipulation and a faster immune selection of transduced cells compared with neoR selection. Finally, the elimination of the neoR gene removes a potent immunogen from transduced lymphocytes.


Asunto(s)
Terapia Genética , Vectores Genéticos , Enfermedad Injerto contra Huésped/terapia , Efecto Injerto vs Tumor , Simplexvirus/enzimología , Timidina Quinasa/genética , Antibacterianos , Antivirales/farmacología , Southern Blotting , Farmacorresistencia Microbiana , Citometría de Flujo , Ganciclovir/farmacología , Técnicas de Transferencia de Gen , Humanos , Activación de Linfocitos , Linfocitos/efectos de los fármacos , Linfocitos/virología , Neomicina/farmacología , Provirus , Retroviridae/efectos de los fármacos , Retroviridae/genética , Simplexvirus/genética
2.
Science ; 270(5235): 470-5, 1995 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-7570000

RESUMEN

Adenosine deaminase (ADA) deficiency results in severe combined immunodeficiency, the first genetic disorder treated by gene therapy. Two different retroviral vectors were used to transfer ex vivo the human ADA minigene into bone marrow cells and peripheral blood lymphocytes from two patients undergoing exogenous enzyme replacement therapy. After 2 years of treatment, long-term survival of T and B lymphocytes, marrow cells, and granulocytes expressing the transferred ADA gene was demonstrated and resulted in normalization of the immune repertoire and restoration of cellular and humoral immunity. After discontinuation of treatment, T lymphocytes, derived from transduced peripheral blood lymphocytes, were progressively replaced by marrow-derived T cells in both patients. These results indicate successful gene transfer into long-lasting progenitor cells, producing a functional multilineage progeny.


Asunto(s)
Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Técnicas de Transferencia de Gen , Terapia Genética , Células Madre Hematopoyéticas , Linfocitos , Inmunodeficiencia Combinada Grave/terapia , Adenosina Desaminasa/administración & dosificación , Adenosina Desaminasa/sangre , Adenosina Desaminasa/uso terapéutico , Formación de Anticuerpos , Secuencia de Bases , Células de la Médula Ósea , Células Cultivadas , Preescolar , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/enzimología , Humanos , Inmunidad Celular , Transfusión de Linfocitos , Linfocitos/enzimología , Linfocitos/inmunología , Datos de Secuencia Molecular , Inmunodeficiencia Combinada Grave/enzimología , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Linfocitos T/enzimología , Linfocitos T/inmunología
3.
Hum Gene Ther ; 6(6): 813-9, 1995 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-7548281

RESUMEN

The infusion of donor lymphocytes after allogeneic bone marrow transplantation is a promising therapeutic tool for achieving a graft versus leukemia (GvL) effect in case of leukemic relapse (1-7), and for the treatment of other complications related to the severe immunosuppressive status of transplanted patients, such as Epstein Barr virus-induced lymphoproliferative disorders (EBV-BLPD) (8) or reactivation of CMV infection (9). Although the delay in the administration of T lymphocytes is expected to reduce the risk of severe GvHD, this risk is still present at higher doses of donor T-cells. The transfer of a suicide gene into donor lymphocytes could allow the in vivo selective elimination of cells responsible for severe GvHD. Additionally, under appropriate conditions, it may allow in vivo modulation of donor anti-tumor responses, and to separate GvL from GvHD. Finally, crucial questions concerning survival and function of donor lymphocytes could be answered by their gene marking. Previous studies documented that T lymphocytes are suitable targets for gene transfer through retroviral vectors (10, 11). This protocol has been designed to evaluate in the contest of allogeneic BMT: 1--the safety of increasing doses of donor lymphocytes transduced with a suicide retroviral vector; 2--the efficacy in terms of survival and immunologic potential of donor lymphocytes after in vitro activation, gene transduction, and immunoselection; 3--the possibility of in vivo down regulation of GvHD by the administration of ganciclovir to patients treated by tk-transduced donor lymphocytes.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Trasplante de Médula Ósea/inmunología , Técnicas de Transferencia de Gen , Terapia Genética , Leucemia/terapia , Simplexvirus/genética , Linfocitos T , Timidina Quinasa/genética , Protocolos Clínicos , Ganciclovir/uso terapéutico , Vectores Genéticos , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/inmunología , Infecciones por Herpesviridae/etiología , Infecciones por Herpesviridae/terapia , Herpesvirus Humano 4 , Humanos , Huésped Inmunocomprometido , Leucemia/inmunología , Selección de Paciente , Complicaciones Posoperatorias/terapia , Retroviridae/genética , Simplexvirus/enzimología , Linfocitos T/inmunología , Linfocitos T/trasplante , Trasplante Homólogo , Infecciones Tumorales por Virus/etiología , Infecciones Tumorales por Virus/terapia
4.
Blood ; 83(7): 1988-97, 1994 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-8142665

RESUMEN

Peripheral blood lymphocytes (PBLs) are key target cells for gene therapy of a number of inherited and acquired blood disorders. We have systematically compared four retroviral vectors, designed according to different strategies, for their efficiency in transfer and expression in human PBLs of the same reporter gene. The receptor gene used in the study codes for the human low-affinity nerve growth factor receptor (LNGFR), and is not expressed on the majority of human hematopoietic cells, thus allowing quantitative analysis of the transduced gene expression by immunofluorescence, with single cell resolution. Peripheral blood mononuclear cells (PBMCs), as well as human hematopoietic cell lines of myeloid and lymphoid origin, were transduced with the four vectors and analyzed for efficiency of gene transfer, integration and stability of vector proviruses, and LNGFR expression at both RNA and protein level. Fluorescence-activated cell sorter analysis of coexpression of LNGFR and lineage-specific cell surface markers was performed in transduced cell lines, PBLs, and T-cell clones to study gene expression on specific cell subpopulations. Although crucial differences were observed among different constructs, all retroviral vectors could transduce, under appropriate infection conditions, T-cell populations representative of the normal immune repertoire. Gene transfer and expression could be demonstrated also in circulating progenitors of mature T cells. Expression of the transduced gene was heterogeneous among cell populations infected with the different vectors, with optimal results obtained by two of the four constructs. Finally, we have devised a simple protocol based on vector-mediated gene transfer and positive immunoselection of the transduced cells that produces virtually 100% gene-modified cells. This may represent a crucial improvement in the way of designing efficacious protocols involving the use of gene-modified T lymphocytes in clinical studies.


Asunto(s)
Técnicas de Transferencia de Gen , Linfocitos/metabolismo , Retroviridae/genética , Línea Celular , Expresión Génica , Terapia Genética , Vectores Genéticos , Humanos , Linfocitos/microbiología , Receptores de Factor de Crecimiento Nervioso/biosíntesis , Receptores de Factor de Crecimiento Nervioso/genética , Secuencias Repetitivas de Ácidos Nucleicos , Transducción Genética , Integración Viral
5.
Blood ; 80(5): 1120-4, 1992 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-1325209

RESUMEN

Peripheral blood lymphocytes obtained from a patient affected by adenosine deaminase (ADA) deficiency and severe combined immunodeficiency were infected with a retroviral vector containing two copies of a human ADA minigene, and injected into bg/nu/xid (BNX) immunodeficient mice. Six to 10 weeks after injection, human T cells were cloned from the spleens of recipient animals and analyzed for proliferative potential, T-cell surface markers, expression of ADA activity, integration of retroviral sequences, T-cell receptor (TCR) beta gene rearrangement, and specificity of antigen recognition. Efficient gene transfer and expression restored proliferative potential in vitro and long-term survival in vivo. All clonable human T lymphocytes obtained from the spleen of recipient animals had high levels of vector-derived ADA enzyme activity and showed predominantly the CD4+ phenotype. Retroviral integrations and TCR-beta gene rearrangements demonstrated the presence of a variety of different clones in the spleens of recipient mice. Furthermore, the combined analyses of vector integration and TCR rearrangement provided evidence that a circulating progenitor cell was transduced by the retroviral vector, giving rise to different and functional TCRs. Evaluation of antigen-specificity demonstrated both alloreactive and foreign antigen specific immune responses. These results suggest that restoration of enzyme activity in human ADA-deficient peripheral blood T cells by retroviral-mediated ADA gene transfer allows in vivo survival and reconstitution of specific immune functions. Therefore, retroviral vector-mediated gene transfer into circulating mononuclear cells could be successful not only in maintaining the metabolic homeostasis, but also for the development of a functional immune repertoire. This is a fundamental prerequisite for the usage of genetically engineered peripheral blood lymphocytes for somatic cell gene therapy of ADA deficiency.


Asunto(s)
Adenosina Desaminasa/genética , Terapia Genética , Linfocitos T/enzimología , Transfección , Adenosina Desaminasa/deficiencia , Animales , Preescolar , Reordenamiento Génico de la Cadena beta de los Receptores de Antígenos de los Linfocitos T , Humanos , Ratones
6.
Science ; 251(4999): 1363-6, 1991 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-1848369

RESUMEN

Deficiency of adenosine deaminase (ADA) results in severe combined immunodeficiency (SCID), a candidate genetic disorder for somatic cell gene therapy. Peripheral blood lymphocytes from patients affected by ADA- SCID were transduced with a retroviral vector for human ADA and injected into immunodeficient mice. Long-term survival of vector-transduced human cells was demonstrated in recipient animals. Expression of vector-derived ADA restored immune functions, as indicated by the presence in reconstituted animals of human immunoglobulin and antigen-specific T cells. Retroviral vector gene transfer, therefore, is necessary and sufficient for development of specific immune functions in vivo and has therapeutic potential to correct this lethal immunodeficiency.


Asunto(s)
Adenosina Desaminasa/deficiencia , Síndromes de Inmunodeficiencia/genética , Adenosina Desaminasa/genética , Animales , Secuencia de Bases , Terapia Genética , Vectores Genéticos , Humanos , Síndromes de Inmunodeficiencia/terapia , Kanamicina Quinasa , Transfusión de Linfocitos , Linfocitos/fisiología , Ratones , Ratones Mutantes , Oligonucleótidos/química , Fosfotransferasas/genética , Reacción en Cadena de la Polimerasa , Retroviridae/genética
7.
J Immunol ; 144(12): 4878-82, 1990 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-2161881

RESUMEN

Tumor-associated macrophages (TAM) have peculiar membrane phenotype and functional properties. In an effort to unravel possible molecular determinants associated with the reprogramming of mononuclear phagocytes that infiltrate tumors, we have investigated the expression of immediate-early genes in TAM from murine sarcomas. c-fos is a prototypic immediate oncogene, with transregulatory function on gene transcription, expressed by myelomonocytic cells and induced by certain activation signals. TAM from three murine sarcomas expressed basal levels of c-fos transcripts considerably higher than those of peritoneal exudate macrophages (PEM). Activation of myelomonocytic cells by bacterial LPS is associated with an early transient increase in c-fos transcription. Unlike PEM, TAM did not show any increase in c-fos expression after exposure to LPS. A similar unresponsiveness to LPS stimulation was observed in macrophages isolated from peritoneal ascitic tumors. c-fos expression in macrophages can be induced via protein kinase C activation or via an increase in cAMP levels. Unlike PEM, TAM did not respond to the protein kinase C activator PMA and to cholera toxin. After culture for 18 to 20 h, c-fos expression in TAM declined, and concomitantly, TAM completely recovered responsiveness to LPS in terms of augmented oncogene mRNA levels. These results demonstrate that TAM from murine sarcomas have an altered expression of the c-fos proto-oncogene, with high basal levels and unresponsiveness to augmenting signals, reversible upon in vitro culture. The altered c-fos expression in TAM may reflect exposure to cytokines present in the tumor microenvironment and may underlie at least some of the peculiar properties of TAM.


Asunto(s)
Macrófagos/fisiología , Proteínas Proto-Oncogénicas/genética , Sarcoma Experimental/genética , Animales , Northern Blotting , Toxina del Cólera/farmacología , Expresión Génica , Lipopolisacáridos/farmacología , Ratones , Colagenasa Microbiana/farmacología , Proteínas Proto-Oncogénicas c-fos , ARN Mensajero/genética
9.
J Immunol ; 144(6): 2409-12, 1990 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-2138198

RESUMEN

Tumor-associated macrophages (TAM) isolated from two murine sarcomas (mFS6 and MN/MCA1) had high levels of proliferative activity (7 to 11% of cells in S phase) compared to peritoneal macrophages (1 to 2% of cells in S phase). In an effort to elucidate the mechanisms responsible for the proliferative activity of TAM, expression of c-fms and macrophage (M)-CSF was investigated in TAM and sarcoma cells. TAM had high levels of mRNA transcripts of the c-fms protooncogene, which encodes a tyrosine kinase probably identical to the M-CSF receptor, but did not express M-CSF transcripts whereas sarcoma cells had high levels of M-CSF mRNA. Sarcoma cell conditioned medium had M-CSF activity on bone marrow cells and induced proliferation of peritoneal exudate and bone marrow-derived macrophages. These activities were blocked by anti-M-CSF antibodies. These findings outline a paracrine circuit in the regulation of TAM proliferation, involving M-CSF, secreted by sarcoma cells and acting on c-fms expressing TAM. Inasmuch as TAM from these murine sarcomas have tumor growth promoting activity, a "ping pong" reciprocal feeding interaction may occur between macrophages and neoplastic cells in these tumors.


Asunto(s)
Macrófagos/inmunología , Proteínas Proto-Oncogénicas/genética , Sarcoma Experimental/inmunología , Animales , Ciclo Celular , Factores Estimulantes de Colonias/genética , Factores Estimulantes de Colonias/farmacología , Expresión Génica , Factor Estimulante de Colonias de Macrófagos , Macrófagos/citología , Ratones , Cavidad Peritoneal/citología , ARN Mensajero/genética , Receptor de Factor Estimulante de Colonias de Macrófagos , Sarcoma Experimental/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA