Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Transl Med ; 13: 240, 2015 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-26198406

RESUMEN

BACKGROUND: Reactivation of latent viruses such as human cytomegalovirus (HCMV) after allogeneic hematopoietic stem cell transplantation (HSCT) results in high morbidity and mortality. Effective immunization against HCMV shortly after allo-HSCT is an unmet clinical need due to delayed adaptive T cell development. Donor-derived dendritic cells (DCs) have a critical participation in stimulation of naïve T cells and immune reconstitution, and therefore adoptive DC therapy could be used to protect patients after HSCT. However, previous methods for ex vivo generation of adoptive donor-derived DCs were complex and inconsistent, particularly regarding cell viability and potency after thawing. We have previously demonstrated in humanized mouse models of HSCT the proof-of-concept of a novel modality of lentivirus-induced DCs ("SmyleDCpp65") that accelerated antigen-specific T cell development. METHODS: Here we demonstrate the feasibility of good manufacturing practices (GMP) for production of donor-derived DCs consisting of monocytes from peripheral blood transduced with an integrase-defective lentiviral vector (IDLV, co-expressing GM-CSF, IFN-α and the cytomegalovirus antigen pp65) that were cryopreserved and thawed. RESULTS: Upscaling and standardized production of one lot of IDLV and three lots of SmyleDCpp65 under GMP-compliant conditions were feasible. Analytical parameters for quality control of SmyleDCpp65 identity after thawing and potency after culture were defined. Cell recovery, uniformity, efficacy of gene transfer, purity and viability were high and consistent. SmyleDCpp65 showed only residual and polyclonal IDLV integration, unbiased to proto-oncogenic hot-spots. Stimulation of autologous T cells by GMP-grade SmyleDCpp65 was validated. CONCLUSION: These results underscore further developments of this individualized donor-derived cell vaccine to accelerate immune reconstitution against HCMV after HSCT in clinical trials.


Asunto(s)
Infecciones por Citomegalovirus/inmunología , Células Dendríticas/citología , Lentivirus , Trasplante de Células Madre/métodos , Animales , Técnicas de Cultivo de Célula , Supervivencia Celular , Criopreservación , Citomegalovirus , Infecciones por Citomegalovirus/prevención & control , Células Dendríticas/virología , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Células HEK293 , Trasplante de Células Madre Hematopoyéticas , Humanos , Interferón-alfa/metabolismo , Leucocitos Mononucleares/citología , Fosfoproteínas/metabolismo , Plásmidos/metabolismo , Transgenes , Proteínas de la Matriz Viral/metabolismo
2.
Mol Ther ; 15(5): 1024-1033, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-28182893

RESUMEN

Drug toxicity and viral resistance limit the long-term efficacy of antiviral drug treatment for human immunodeficiency virus (HIV) infection. Thus, alternative therapies need to be explored. We tested the infusion of T lymphocytes transduced with a retroviral vector (M87o) that expresses an HIV entry-inhibitory peptide (maC46). Gene-modified autologous T cells were infused into ten HIV-infected patients with advanced disease and multidrug-resistant virus during anti-retroviral combination therapy. T-cell infusions were tolerated well, with no severe side effects. A significant increase of CD4 counts was observed after infusion. At the end of the 1-year follow-up, the CD4 counts of all patients were still around or above baseline. Gene-modified cells could be detected in peripheral blood, lymph nodes, and bone marrow throughout the 1-year follow-up, and marking levels correlated with the cell dose. No significant changes of viral load were observed during the first 4 months. Four of the seven patients who changed their antiviral drug regimen thereafter responded with a significant decline in plasma viral load. In conclusion, the transfer of gene-modified cells was safe, led to sustained levels of gene marking, and may improve immune competence in HIV-infected patients with advanced disease and multidrug-resistant virus.

3.
Exp Hematol ; 30(2): 150-7, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11823050

RESUMEN

OBJECTIVE: Hematopoietic progenitor cells are a promising source for generation of genetically modified dendritic cells. A prerequisite for using these cells in therapeutic approaches is stable vector-mediated transgene expression during and after cell maturation. We investigated the expression of enhanced green fluorescence protein (EGFP) mediated by retroviral vectors in dendritic cells and other hematopoietic cells differentiated in vitro. MATERIAL AND METHODS: CD34(+) cells were efficiently transduced with retroviral vector constructs known to mediate different expression levels due to distinct cis-acting elements. EGFP(+) cells were purified by cell sorting and differentiated to monocytes, granulocytes, dendritic cells, and erythrocytes. Coexpression of EGFP and cell type-specific markers was analyzed by flow cytometry. RESULTS: Transgene expression from various retroviral vectors was silenced exclusively in dendritic cells, but not in other mature myeloid cells. Loss of EGFP was most pronounced in cells initially displaying low expression levels. This was confirmed by using a retroviral vector coding for a variant of EGFP with significantly reduced half-life. In contrast, a majority of dendritic cells showed stable expression when a self-inactivating retroviral construct using an internal cytomegalovirus promotor was used. CONCLUSIONS: We suggest that expression from the retroviral long terminal repeat is silenced during dendritic cell differentiation in vitro. High levels of stable transgene product in progenitor cells may mask a loss of expression. An improvement of retroviral vectors mediating stable transgenic expression is necessary for therapeutic approaches using gene-modified dendritic cells.


Asunto(s)
Linaje de la Célula/genética , Células Dendríticas/fisiología , Vectores Genéticos , Células Madre Hematopoyéticas/fisiología , Retroviridae , Transducción Genética , Células Cultivadas , Células Dendríticas/citología , Regulación hacia Abajo/genética , Células Madre Hematopoyéticas/citología , Humanos , Monocitos/citología , Monocitos/fisiología
4.
Stem Cells Dev ; 13(1): 71-81, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15068695

RESUMEN

In a clinical trial that we recently reported, a suicide gene transfer in human primary T cells required 12 days of ex vivo culture, including activation of peripheral blood mononuclear cells (PBMC) with CD3 monoclonal antibody (CD3 mAb), retrovirus-mediated transduction, and selection of gene-modified cells (GMC) by G418. The aim of the present study was to determine the impact of the initial T cell activation and of the transduction/selection on T cell receptor beta variable chain (TCRBV) repertoire of GMC by using the spectratyping method. The TCRBV repertoires of nontransduced, nonselected control (Co) cells and of GMC generated after an initial stimulation with CD3 mAb, CD3/CD28 beads, or allogeneic PBMC or Epstein-Barr virus-transformed B (B-EBV) cells were compared to the ones of their corresponding PBMC. The TCRBV repertoires were skewed in Co cells generated after CD3 mAb or after allogeneic stimulation, and even more so in their corresponding GMC, demonstrating that both culture-dependent and transduction/selection-dependent events led to TCRBV repertoire alterations. However, TCRBV repertoires were not altered, or to a lesser extent, in Co cells or GMC produced after CD3/CD28 bead activation, demonstrating a protective effect on both culture-dependent and transduction/selection-dependent repertoire alterations. Thus, we suggest to replace the initial CD3 mAb stimulation by CD3/CD28 beads for the production of clinical-grade GMC in the setting of future gene therapy trials.


Asunto(s)
Técnicas de Transferencia de Gen , Retroviridae/genética , Linfocitos T/inmunología , Anticuerpos Monoclonales/química , Linfocitos B/metabolismo , Linfocitos B/virología , Antígenos CD28/inmunología , Antígenos CD28/metabolismo , Complejo CD3/inmunología , Complejo CD3/metabolismo , Antígenos CD4/biosíntesis , Antígenos CD8/biosíntesis , Células Cultivadas , ADN Complementario/metabolismo , Terapia Genética , Humanos , Leucocitos Mononucleares/citología , Activación de Linfocitos , Prueba de Cultivo Mixto de Linfocitos , Oligonucleótidos/química , Receptores de Antígenos de Linfocitos T/química , Receptores de Antígenos de Linfocitos T alfa-beta/química , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/metabolismo , Factores de Tiempo , Transgenes
5.
Hum Gene Ther Clin Dev ; 25(4): 218-28, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25381930

RESUMEN

The clinical application of self-inactivating (SIN) retroviral vectors requires an efficient vector production technology. To enable production of γ-retroviral SIN vectors from stable producer cells, new targetable HEK293-based producer clones were selected, providing amphotropic, GALV, or RD114 pseudotyping. Viral vector expression constructs can reliably be inserted at a predefined genomic locus via Flp-recombinase-mediated cassette exchange. Introduction of a clean-up step, mediated by Cre-recombinase, allows the removal of residual sequences that were required for targeting and selection, but were dispensable for the final producer clones and eliminated homology-driven recombination between the tagging and the therapeutic vector. The system was used to establish GALV and RD114 pseudotyping producer cells (HG- and HR820) for a clinically relevant long terminal repeat-driven therapeutic vector, designed for the transfer of a recombinant TCR that delivered titers in the range of 2×10(7) infectious particles (IP)/ml. Production capacity of the amphotropic producer cell (HA820) was challenged by a therapeutic SIN vector transferring the large COL7A1 cDNA. The final producer clone delivered a titer of 4×10(6) IP/ml and the vector containing supernatant was used directly to functionally restore primary fibroblasts and keratinocytes isolated from recessive dystrophic epidermolysis bullosa patients. Thus, the combinatorial approach (fc-technology) to generate producer cells for therapeutic γ-retroviral (SIN) vectors is feasible, is highly efficient, and allows their safe production and application in clinical trials.


Asunto(s)
Colágeno Tipo VII/genética , ADN Recombinante/genética , Gammaretrovirus/genética , Ingeniería Genética/métodos , Vectores Genéticos/genética , Colágeno Tipo VII/metabolismo , ADN Recombinante/aislamiento & purificación , Gammaretrovirus/metabolismo , Marcación de Gen/métodos , Vectores Genéticos/aislamiento & purificación , Células HEK293 , Humanos
6.
Mol Ther ; 15(5): 1024-33, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17356541

RESUMEN

Drug toxicity and viral resistance limit the long-term efficacy of antiviral drug treatment for human immunodeficiency virus (HIV) infection. Thus, alternative therapies need to be explored. We tested the infusion of T lymphocytes transduced with a retroviral vector (M87o) that expresses an HIV entry-inhibitory peptide (maC46). Gene-modified autologous T cells were infused into ten HIV-infected patients with advanced disease and multidrug-resistant virus during anti-retroviral combination therapy. T-cell infusions were tolerated well, with no severe side effects. A significant increase of CD4 counts was observed after infusion. At the end of the 1-year follow-up, the CD4 counts of all patients were still around or above baseline. Gene-modified cells could be detected in peripheral blood, lymph nodes, and bone marrow throughout the 1-year follow-up, and marking levels correlated with the cell dose. No significant changes of viral load were observed during the first 4 months. Four of the seven patients who changed their antiviral drug regimen thereafter responded with a significant decline in plasma viral load. In conclusion, the transfer of gene-modified cells was safe, led to sustained levels of gene marking, and may improve immune competence in HIV-infected patients with advanced disease and multidrug-resistant virus.


Asunto(s)
Traslado Adoptivo/métodos , Infecciones por VIH/terapia , VIH/inmunología , Linfocitos T/inmunología , Adulto , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Farmacorresistencia Viral/inmunología , Citometría de Flujo , Vectores Genéticos/genética , Infecciones por VIH/inmunología , Humanos , Masculino , Persona de Mediana Edad , Mutación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Retroviridae/genética , Linfocitos T/citología , Linfocitos T/metabolismo , Resultado del Tratamiento , Carga Viral
7.
Blood ; 105(11): 4235-46, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15713797

RESUMEN

Previous studies have demonstrated leukemic complications in mice after high-copy retroviral gene transfer of the multidrug resistance 1 (MDR1) cDNA, encoding a membrane-located efflux pump expressed in hematopoietic stem cells. In contrast, no such complications or MDR1-associated alterations of hematopoiesis were observed in numerous other studies exploring MDR1 gene transfer into cell lines, mice, dogs, nonhuman primates, and human subjects. Here, we show that leukemias associated with retroviral expression of MDR1 depend on high vector dose, and involve the selection of clones with combinatorial insertional mutagenesis of proto-oncogenes or other signaling genes. Compared with insertion patterns in normal long-term repopulating hematopoietic cells, such hits were overrepresented in leukemic clones, pointing to a causal role. A similar constellation of insertion sites was also observed in a leukemia arising after high-copy retroviral gene transfer of a fluorescent protein. Spectral karyotyping demonstrated additional chromosomal translocations in a subset of cases, indicative of secondary genetic instability. We also show that insertional mutants can be amplified in vitro prior to transplantation. On the basis of these findings, we suggest the use of preclinical dose-escalation studies to define a therapeutic index for retroviral transgene delivery.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/administración & dosificación , Técnicas de Transferencia de Gen/efectos adversos , Leucemia/etiología , Mutagénesis Insercional , Retroviridae/genética , Animales , Dosificación de Gen , Genes MDR/genética , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Ratones , Ratones Endogámicos C57BL , Translocación Genética
8.
Mol Ther ; 9(5): 738-46, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15120335

RESUMEN

Retroviral producer cells are generated by the introduction of a viral genome into "helper" cell lines containing all the necessary components for viral packaging and the release of infectious particles. The selection of high-titer vector producer cells is most efficient if the vector genome encodes a selectable marker, while it is extremely tedious to select high-titer producer clones if the transgene cannot be detected and selected directly. Here we describe the development of a screening system that uses reversible integration of lox-P-flanked eGFP as a qualitative and quantitative marker gene in two different vector systems, greatly facilitating the selection of viral producer cells. After selection and titration of high-titer viral producer cells based on eGFP expression, the eGFP gene could be removed from the provirus by transient introduction of Cre-recombinase into the producer cells, thus allowing the production of therapeutic relevant vectors expressing solely the gene of interest. However, after removal of the marker gene a slight but consistent increase in viral titers compared to the respective control vectors was found, independent of the transgene or backbone used. The single lox-P site retained in the vector backbone does not affect gene expression level or fidelity of RNA processing.


Asunto(s)
Vectores Genéticos/genética , Integrasas/metabolismo , Retroviridae/genética , Proteínas Virales/metabolismo , Animales , Antígenos CD/genética , Antígenos CD34/genética , Técnicas de Cultivo de Célula , Expresión Génica , Reordenamiento Génico/genética , Marcadores Genéticos , Vectores Genéticos/uso terapéutico , Humanos , Integrasas/genética , Proteína Cofactora de Membrana , Glicoproteínas de Membrana/genética , Ratones , Provirus/genética , Provirus/crecimiento & desarrollo , Recombinación Genética/genética , Retroviridae/crecimiento & desarrollo , Secuencias Repetidas Terminales/genética , Proteínas Virales/genética
9.
Stem Cells ; 22(4): 570-9, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15277702

RESUMEN

Methods to analyze the clonality of an adverse event in preclinical or clinical retroviral stem cell gene therapy protocols are needed. We analyzed the progeny of retrovirally transduced human peripheral blood progenitor cells (PBPCs) after transplantation and engraftment in immune-deficient mice. The integration site of the provirus serves as a unique tag of the individual transduced PBPC. A plasmid library of junctions between proviral and human genomic DNA was generated. We were able to detect individual transduced cell clones that amounted to 0.14%-0.0001% of chimeric bone marrow cells. This is the first report in which the contribution of individual marrow-repopulating cells to human hematopoiesis is directly quantified.


Asunto(s)
Antígenos CD34/sangre , Células de la Médula Ósea/citología , Trasplante de Células Madre/métodos , Trasplante Heterólogo/inmunología , Animales , Terapia Genética/métodos , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID
10.
Blood ; 101(6): 2191-8, 2003 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-12424203

RESUMEN

Increasing use of hematopoietic stem cells for retroviral vector-mediated gene therapy and recent reports on insertional mutagenesis in mice and humans have created intense interest to characterize vector integrations on a genomic level. We studied retrovirally transduced human peripheral blood progenitor cells with bone marrow-repopulating ability in immune-deficient mice. By using a highly sensitive and specific ligation-mediated polymerase chain reaction (PCR) followed by sequencing of vector integration sites, we found a multitude of simultaneously active human stem cell clones 8 weeks after transplantation. Vector integrations occurred with significantly increased frequency into chromosomes 17 and 19 and into specific regions of chromosomes 6, 13, and 16, although most of the chromosomes were targeted. Preferred genomic target sites have previously only been reported for wild-type retroviruses. Our findings reveal for the first time that retroviral vector integration into human marrow-repopulating cells can be nonrandom (P =.000 37).


Asunto(s)
Células de la Médula Ósea/citología , Vectores Genéticos , Retroviridae/genética , Animales , Cromosomas Humanos Par 13 , Cromosomas Humanos Par 16 , Cromosomas Humanos Par 17 , Cromosomas Humanos Par 19 , Cromosomas Humanos Par 6 , Clonación Molecular , Femenino , Hematopoyesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Hibridación Fluorescente in Situ , Ratones , Ratones Endogámicos NOD , Ratones SCID , Reacción en Cadena de la Polimerasa/métodos , Transfección , Integración Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA