Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Gene Med ; 20(10-11): e3050, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30129972

RESUMEN

BACKGROUND: Gene therapy approaches for the treatment of Fanconi anemia (FA) hold promise for patients without a suitably matched donor for an allogeneic bone marrow transplant. However, significant limitations include the collection of sufficient stem cell numbers from patients, the fragility of these cells during ex vivo manipulation, and clinically meaningful engraftment following transplantation. With these challenges in mind, we were interested in determining (i) whether gene-corrected cells at progressively lower numbers can successfully engraft in FA; (ii) whether low-dose conditioning facilitates this engraftment; and (iii) whether these cells can be selected for post-transplant. METHODS: Utilizing a well characterized mouse model of FA, we infused donor bone marrow from healthy heterozygote littermates that are unaffected carriers of the FANCA mutation to mimic a gene-corrected product, after administering low-dose conditioning. Once baseline engraftment was observed, we administered a second, very-low selective dose to determine whether gene-corrected cells could be selected for in vivo. RESULTS: We demonstrate that upfront low-dose conditioning greatly increases successful engraftment of hematopoietic corrected cells in a pre-clinical animal model of FA. Additionally, without conditioning, cells can still engraft and demonstrate a selective advantage in vivo over time following transplantation, and these corrected cells can be directly selected for in vivo after engraftment. CONCLUSIONS: Minimal conditioning prior to bone marrow transplant in Fanconi anemia promotes the multi-lineage engraftment of 10-fold fewer cells compared to nonconditioned controls. These data provide important insights into the potential of minimally toxic conditioning protocols for FA gene therapy applications.


Asunto(s)
Trasplante de Médula Ósea/métodos , Ciclofosfamida/administración & dosificación , Proteína del Grupo de Complementación A de la Anemia de Fanconi/metabolismo , Anemia de Fanconi/terapia , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Animales , Recuento de Células , Relación Dosis-Respuesta a Droga , Anemia de Fanconi/genética , Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Vectores Genéticos/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Inmunosupresores/administración & dosificación , Lentivirus/genética , Ratones Noqueados
2.
Blood ; 123(23): 3578-84, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24642749

RESUMEN

Current approaches to hematopoietic stem cell (HSC) gene therapy involve the collection and ex vivo manipulation of HSCs, a process associated with loss of stem cell multipotency and engraftment potential. An alternative approach for correcting blood-related diseases is the direct intravenous administration of viral vectors, so-called in vivo gene therapy. In this study, we evaluated the safety and efficacy of in vivo gene therapy using a foamy virus vector for the correction of canine X-linked severe combined immunodeficiency (SCID-X1). In newborn SCID-X1 dogs, injection of a foamy virus vector expressing the human IL2RG gene resulted in an expansion of lymphocytes expressing the common γ chain and the development of CD3(+) T lymphocytes. CD3(+) cells expressed CD4 and CD8 coreceptors, underwent antigen receptor gene rearrangement, and demonstrated functional maturity in response to T-cell mitogens. Retroviral integration site analysis in 4 animals revealed a polyclonal pattern of integration in all dogs with evidence for dominant clones. These results demonstrate that a foamy virus vector can be administered with therapeutic benefit in the SCID-X1 dog, a clinically relevant preclinical model for in vivo gene therapy.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Spumavirus , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Animales , Células Sanguíneas/metabolismo , Linaje de la Célula/genética , Modelos Animales de Enfermedad , Perros , Células HEK293 , Humanos , Inyecciones Intravenosas , Integración Viral/genética
3.
BMC Bioinformatics ; 16: 212, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26150117

RESUMEN

BACKGROUND: Analyzing the integration profile of retroviral vectors is a vital step in determining their potential genotoxic effects and developing safer vectors for therapeutic use. Identifying retroviral vector integration sites is also important for retroviral mutagenesis screens. RESULTS: We developed VISA, a vector integration site analysis server, to analyze next-generation sequencing data for retroviral vector integration sites. Sequence reads that contain a provirus are mapped to the human genome, sequence reads that cannot be localized to a unique location in the genome are filtered out, and then unique retroviral vector integration sites are determined based on the alignment scores of the remaining sequence reads. CONCLUSIONS: VISA offers a simple web interface to upload sequence files and results are returned in a concise tabular format to allow rapid analysis of retroviral vector integration sites.


Asunto(s)
Vectores Genéticos , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Internet , Retroviridae/genética , Programas Informáticos , Integración Viral/genética , Humanos
4.
Blood ; 122(2): 179-87, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23719296

RESUMEN

Despite continued progress in the development of novel antiretroviral therapies, it has become increasingly evident that drug-based treatments will not lead to a functional or sterilizing cure for HIV(+) patients. In 2009, an HIV(+) patient was effectively cured of HIV following allogeneic transplantation of hematopoietic stem cells (HSCs) from a CCR5(-/-) donor. The utility of this approach, however, is severely limited because of the difficulty in finding matched donors. Hence, we studied the potential of HIV-resistant stem cells in the autologous setting in a nonhuman primate AIDS model and incorporated a fusion inhibitor (mC46) as the means for developing infection-resistant cells. Pigtail macaques underwent identical transplants and Simian-Human Immunodeficiency Virus (SHIV) challenge procedures with the only variation between control and mC46 macaques being the inclusion of a fusion-inhibitor expression cassette. Following SHIV challenge, mC46 macaques, but not control macaques, showed a positive selection of gene-modified CD4(+) T cells in peripheral blood, gastrointestinal tract, and lymph nodes, accounting for >90% of the total CD4(+) T-cell population. mC46 macaques also maintained high frequencies of SHIV-specific, gene-modified CD4(+) T cells, an increase in nonmodified CD4(+) T cells, enhanced cytotoxic T lymphocyte function, and antibody responses. These data suggest that HSC protection may be a potential alternative to conventional antiretroviral therapy in patients with HIV/AIDS.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Proteínas Recombinantes de Fusión/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Antígenos Virales/inmunología , Linfocitos B/inmunología , Recuento de Linfocito CD4 , Relación CD4-CD8 , Linfocitos T CD4-Positivos/virología , Tratamiento Basado en Trasplante de Células y Tejidos , Expresión Génica , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Macaca nemestrina , Síndrome de Inmunodeficiencia Adquirida del Simio/terapia , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Carga Viral , Viremia/inmunología , Viremia/virología
5.
Blood ; 120(13): e35-44, 2012 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-22898598

RESUMEN

Induced pluripotent stem cell (iPSC) therapeutics are a promising treatment for genetic and infectious diseases. To assess engraftment, risk of neoplastic formation, and therapeutic benefit in an autologous setting, testing iPSC therapeutics in an appropriate model, such as the pigtail macaque (Macaca nemestrina; Mn), is crucial. Here, we developed a chemically defined, scalable, and reproducible specification protocol with bone morphogenetic protein 4, prostaglandin-E2 (PGE2), and StemRegenin 1 (SR1) for hematopoietic differentiation of Mn iPSCs. Sequential coculture with bone morphogenetic protein 4, PGE2, and SR1 led to robust Mn iPSC hematopoietic progenitor cell formation. The combination of PGE2 and SR1 increased CD34(+)CD38(-)Thy1(+)CD45RA(-)CD49f(+) cell yield by 6-fold. CD34(+)CD38(-)Thy1(+)CD45RA(-)CD49f(+) cells isolated on the basis of CD34 expression and cultured in SR1 expanded 3-fold and maintained this long-term repopulating HSC phenotype. Purified CD34(high) cells exhibited 4-fold greater hematopoietic colony-forming potential compared with unsorted hematopoietic progenitors and had bilineage differentiation potential. On the basis of these studies, we calculated the cell yields that must be achieved at each stage to meet a threshold CD34(+) cell dose that is required for engraftment in the pigtail macaque. Our protocol will support scale-up and testing of iPSC-derived CD34(high) cell therapies in a clinically relevant nonhuman primate model.


Asunto(s)
Antígenos CD34/metabolismo , Diferenciación Celular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Animales , Western Blotting , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Linaje de la Célula , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Dinoprostona/genética , Dinoprostona/metabolismo , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Linfocitos/citología , Linfocitos/metabolismo , Macaca , Células Mieloides/citología , Células Mieloides/metabolismo , Purinas/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Internalización del Virus , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
6.
Mol Ther ; 21(6): 1270-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23587923

RESUMEN

Umbilical cord blood (CB) transplantation is a promising therapeutic approach but continues to be associated with delayed engraftment and infections. Here, we explored in our macaque CB transplant model expansion and engraftment kinetics of cells expanded with the combination of HOXB4 and Delta-1. CB cells were divided into two equal fractions; one fraction was transduced with HOXB4 yellow fluorescent protein (YFP) and expanded on control OP9 cells, and the other was transduced with HOXB4 green fluorescent protein (GFP) and expanded on Delta-expressing OP9 cells (OP9-DL1). Both fractions were transplanted into myeloablated subjects. Neutrophil and platelet recovery occurred within 7 and 19 days respectively, which was significantly earlier than in our previous study using cells expanded with HOXB4 alone, which resulted in neutrophil recovery within 12 days (P = 0.05) and platelet recovery within 37 days (P = 0.02). Furthermore, two of three animals in the current study remained fully transfusion-independent after transplantation. By day 30, reconstitution of lymphocytes was significantly greater with the HOXB4/OP9-DL1 expanded cells in all animals (P = 0.05). In conclusion, our data show that the combination of OP9-DL1 and HOXB4 can result in increased numbers of repopulating cells, thus leading to rapid engraftment and transfusion independence in macaques transplanted with autologous, expanded CB cells.


Asunto(s)
Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Macaca nemestrina/genética , Proteínas de la Membrana/genética , Regiones Promotoras Genéticas , Factores de Transcripción/genética , Animales , Antígenos CD34/metabolismo , Plaquetas/metabolismo , Transfusión Sanguínea/métodos , Diferenciación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Femenino , Sangre Fetal/citología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Proteínas de Homeodominio/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macaca nemestrina/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Factores de Transcripción/metabolismo
7.
Nucleic Acids Res ; 40(2): e14, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22110042

RESUMEN

Two major limitations to achieve efficient homing endonuclease-stimulated gene correction using retroviral vectors are low frequency of gene targeting and random integration of the targeting vectors. To overcome these issues, we developed a reporter system for quick and facile testing of novel strategies to promote the selection of cells that undergo targeted gene repair and to minimize the persistence of random integrations and non-homologous end-joining events. In this system, the gene target has an I-SceI site upstream of an EGFP reporter; and the repair template includes a non-functional EGFP gene, the positive selection transgene MGMTP140K tagged with mCherry, and the inducible Caspase-9 suicide gene. Using this dual fluorescent reporter system it is possible to detect properly targeted integration. Furthermore, this reporter system provides an efficient approach to enrich for gene correction events and to deplete events produced by random integration. We have also developed a second reporter system containing MGMTP140K in the integrated target locus, which allows for selection of primary cells with the integrated gene target after transplantation. This system is particularly useful for testing repair strategies in primary hematopoietic stem cells. Thus, our reporter systems should allow for more efficient gene correction with less unwanted off target effects.


Asunto(s)
Endodesoxirribonucleasas/metabolismo , Marcación de Gen/métodos , Genes Reporteros , Línea Celular , Colorantes Fluorescentes/análisis , Genoma , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Proteína Metiltransferasas/análisis , Proteína Metiltransferasas/genética
8.
J Gene Med ; 15(6-7): 233-41, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23666780

RESUMEN

BACKGROUND: Nonmyeloablative allogeneic hematopoietic stem cell (HSC) transplantation can cure malignant and nonmalignant diseases affecting the hematopoietic system, such as severe combined immunodeficiencies, aplastic anemia and hemoglobinopathies. Although nonmyeloablative is favored over myeloablative transplantation for many patients, graft rejection remains problematic. One strategy for decreasing rejection is to protect donor activated T cells in the graft from methotrexate (MTX) by genetically modifying the cells to express MTX-resistant dihydrofolate reductase (Tyr22-DHFR), leaving the immunosuppressive effects of MTX to act solely on activated host T lymphocytes, shifting the balance to favor allogeneic engraftment. METHODS: To evaluate MTX resistance of Tyr22-DHFR(+) T lymphocytes in vivo, we transplanted dogs with autologous CD34(+) cells modified with yellow fluorescent protein (YFP) and DHFR-green fluorescent protein (GFP) lentivirus vectors. Dogs were then treated with a standard MTX regimen days 1, 3, 6 and 11) following immune activation with a foreign antigen as a surrogate assay to mimic early transplantation. RESULTS: DHFR-GFP(+) gene marking was maintained in CD3(+) CD25(+) and CD4(+) T lymphocytes after MTX treatment, whereas the level of T lymphocytes that expressed only a fluorescent reporter (YFP(+) ) decreased. These data show that Tyr22-DHFR expression protects T lymphocytes from MTX toxicity in dogs, highlighting a clinically relevant application for preserving donor T lymphocytes during post-transplantation immunosuppression. CONCLUSIONS: The findings of the present study have implications for the clinical translation of MTX-resistant T cells to facilitate engraftment of allogeneic cells following nonmyeloablative conditioning and to minimize the risk of rejection. In summary, Tyr22-DHFR expression in T lymphocytes provides chemoprotection from MTX-mediated elimination in the context of immune activation in vivo.


Asunto(s)
Resistencia a Medicamentos/genética , Metotrexato/farmacología , Linfocitos T/efectos de los fármacos , Linfocitos T/enzimología , Tetrahidrofolato Deshidrogenasa/genética , Tetrahidrofolato Deshidrogenasa/metabolismo , Animales , Perros , Inhibidores Enzimáticos/farmacología , Citometría de Flujo , Trasplante de Células Madre Hematopoyéticas , Reacción en Cadena de la Polimerasa , Ovinos , Linfocitos T/citología
9.
Mol Ther ; 19(7): 1287-94, 2011 07.
Artículo en Inglés | MEDLINE | ID: mdl-21326218

RESUMEN

We report long-term results from a large animal model of in vivo selection. Nine years ago, we transplanted two dogs (E900 and E958) with autologous marrow CD34(+) cells that had been transduced with a gammaretrovirus vector encoding a conditionally activatable derivative of the thrombopoietin receptor. Receptor activation through administration of a chemical inducer of dimerization (CID) (AP20187 or AP1903) confers a growth advantage. We previously reported responses to two 30-day intravenous (i.v.) courses of AP20187 administered within the first 8 months post-transplantation. We now report responses to 5-day subcutaneous (s.c.) courses of AP20187 or AP1903 at months 14, 90, and 93 (E900), or month 18 (E958), after transplantation. Long-term monitoring showed no rise in transduced cells in the absence of drug. Retroviral insertion site analysis showed that 4 of 6 (E958) and 5 of 12 (E900) transduced hematopoietic cell clones persisted lifelong. Both dogs were euthanized for reasons unrelated to the gene therapy treatment at 8 years 11 months (E958) and 11 years 1 month (E900) of age. Three clones from E900 remained detectable in each of two secondary recipients, one of which was treated with, and responded to, AP1903. Our results demonstrate the feasibility of safely regulating genetically engineered hematopoietic cells over many years.


Asunto(s)
Gammaretrovirus/genética , Vectores Genéticos/genética , Células Madre Hematopoyéticas/metabolismo , Animales , Células Cultivadas , Reactivos de Enlaces Cruzados/farmacología , Perros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Compuestos Orgánicos/farmacología , Tacrolimus/análogos & derivados , Tacrolimus/farmacología
10.
J Clin Invest ; 118(4): 1502-10, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18357342

RESUMEN

Retroviral vector-mediated HSC gene therapy has been used to treat individuals with a number of life-threatening diseases. However, some patients with SCID-X1 developed retroviral vector-mediated leukemia after treatment. The selective growth advantage of gene-modified cells in patients with SCID-X1 suggests that the transgene may have played a role in leukemogenesis. Here we report that 2 of 2 dogs and 1 of 2 macaques developed myeloid leukemia approximately 2 years after being transplanted with cells that overexpressed homeobox B4 (HOXB4) and cells transduced with a control gammaretroviral vector that did not express HOXB4. The leukemic cells had dysregulated expression of oncogenes, a block in myeloid differentiation, and overexpression of HOXB4. HOXB4 knockdown restored differentiation in leukemic cells, suggesting involvement of HOXB4. In contrast, leukemia did not arise from the cells carrying the control gammaretroviral vector. In addition, leukemia did not arise in 5 animals with high-level marking and polyclonal long-term repopulation following transplantation with cells transduced with an identical gammaretrovirus vector backbone expressing methylguanine methyltransferase. These findings, combined with the absence of leukemia in many other large animals transplanted with cells transduced with gammaretroviral vectors expressing genes other than HOXB4, show that HOXB4 overexpression poses a significant risk of leukemogenesis. Our data thus suggest the continued need for caution in genetic manipulation of repopulating cells, particularly when the transgene might impart an intrinsic growth advantage.


Asunto(s)
Gammaretrovirus/genética , Terapia Genética/efectos adversos , Vectores Genéticos/genética , Proteínas de Homeodominio/metabolismo , Leucemia Mieloide/etiología , Macaca nemestrina , Trasplante de Células Madre/efectos adversos , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Perros , Regulación de la Expresión Génica , Marcadores Genéticos/genética , Hematopoyesis , Proteínas de Homeodominio/genética , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Mutación/genética , Factores de Tiempo
11.
Blood ; 113(21): 5094-103, 2009 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-19336761

RESUMEN

Overexpression of methylguanine methyltransferase P140K (MGMTP140K) has been successfully used for in vivo selection and chemoprotection in mouse and large animal studies, and has promise for autologous and allogeneic gene therapy. We examined the long-term safety of MGMTP140K selection in a clinically relevant dog model. Based on the association of provirus integration and proto-oncogene activation leading to leukemia in the X-linked immunodeficiency trial, we focused our analysis on the distribution of retrovirus integration sites (RIS) relative to proto-oncogene transcription start sites (TSS). We analyzed RIS near proto-oncogene TSS before (n = 157) and after (n = 129) chemotherapy in dogs that received MGMTP140K gene-modified cells and identified no overall increase of RIS near proto-oncogene TSS after chemotherapy. We also wanted to determine whether in vivo selected cells retained fundamental characteristics of hematopoietic stem cells. To that end, we performed secondary transplantation of MGMTP140K gene-modified cells after in vivo selection in dog leukocyte antigen (DLA)-matched dogs. Gene-modified cells achieved multilineage repopulation, and we identified the same gene-modified clone in both dogs more than 800 and 900 days after transplantation. These data suggest that MGMTP140K selection is well tolerated and should allow clinically for selection of gene-corrected cells in genetic or infectious diseases or chemoprotection for treatment of malignancy.


Asunto(s)
Terapia Genética/métodos , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , O(6)-Metilguanina-ADN Metiltransferasa/genética , Animales , Linaje de la Célula , Perros , Vectores Genéticos , Retroviridae/genética
12.
Mol Ther ; 18(4): 725-33, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19997089

RESUMEN

Lentiviral vectors are established as efficient and convenient vehicles for gene transfer. They are almost always pseudotyped with the envelope glycoprotein of vesicular stomatitis virus (VSV-G) due to the high titers that can be achieved, their stability, and broad tropism. We generated a novel cocal vesiculovirus envelope glycoprotein plasmid and compared the properties of lentiviral vectors pseudotyped with cocal, VSV-G, and a modified feline endogenous retrovirus envelope glycoprotein (RD114/TR). Cocal-pseudotyped lentiviral vectors can be produced at titers as high as with VSV-G, have a broad tropism, and are stable, allowing for efficient concentration by centrifugation. Additionally, cocal vectors are more resistant to inactivation by human serum than VSV-G-pseudotyped vectors, and efficiently transduce human CD34(+) nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse-repopulating cells (SRCs), and long-term primate hematopoietic repopulating cells. These studies establish the potential of cocal-pseudotyped lentiviral vectors for a variety of scientific and therapeutic gene transfer applications, including in vivo gene delivery and hematopoietic stem cell (HSC) gene therapy.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos/inmunología , Células Madre Hematopoyéticas , Lentivirus/inmunología , Glicoproteínas de Membrana/inmunología , Células Receptoras Sensoriales , Animales , Antígenos CD34/genética , Gatos , Vectores Genéticos/genética , Humanos , Lentivirus/genética , Macaca , Glicoproteínas de Membrana/genética , Ratones , Ratones SCID , Transducción Genética , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
13.
Mol Ther Methods Clin Dev ; 22: 350-359, 2021 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-34514027

RESUMEN

Pyruvate kinase deficiency (PKD) is a rare autosomal recessive disorder caused by mutations in the PKLR gene. PKD is characterized by non-spherocytic hemolytic anemia of variable severity and may be fatal in some cases during early childhood. Although not considered the standard of care, allogeneic stem cell transplantation has been shown as a potentially curative treatment, limited by donor availability, toxicity, and incomplete engraftment. Preclinical studies were conducted to define conditions to enable consistent therapeutic reversal, which were based on our previous data on lentiviral gene therapy for PKD. Improvement of erythroid parameters was identified by the presence of 20%-30% healthy donor cells. A minimum vector copy number (VCN) of 0.2-0.3 was required to correct PKD when corrected cells were transplanted in a mouse model for PKD. Biodistribution and pharmacokinetics studies, with the aim of conducting a global gene therapy clinical trial for PKD patients (RP-L301-0119), demonstrated that genetically corrected cells do not confer additional side effects. Moreover, a clinically compatible transduction protocol with mobilized peripheral blood CD34+ cells was optimized, thus facilitating the efficient transduction on human cells capable of repopulating the hematopoiesis of immunodeficient mice. We established conditions for a curative lentiviral vector gene therapy protocol for PKD.

14.
Blood ; 111(12): 5537-43, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18388180

RESUMEN

Lentiviral vectors are attractive for hematopoietic stem cell (HSC) gene therapy because they do not require mitosis for nuclear entry, they efficiently transduce hematopoietic repopulating cells, and self-inactivating (SIN) designs can be produced at high titer. Experiments to evaluate HIV-derived lentiviral vectors in nonhuman primates prior to clinical trials have been hampered by low transduction frequencies due in part to host restriction by TRIM5alpha. We have established conditions for efficient transduction of pigtailed macaque (Macaca nemestrina) long-term repopulating cells using VSV-G-pseudotyped HIV-based lentiviral vectors. Stable, long-term, high-level gene marking was observed in 3 macaques using relatively low MOIs (5-10) in a 48-hour ex vivo transduction protocol. All animals studied had rapid neutrophil engraftment with a median of 10.3 days to a count greater than 0.5 x 10(9)/L (500/microL). Expression was detected in all lineages, with long-term marking levels in granulocytes at approximately 20% to 30%, and in lymphocytes at approximately 12% to 23%. All animals had polyclonal engraftment as determined by analysis of vector integration sites. These data suggest that lentiviral vectors should be highly effective for HSC gene therapy, particularly for diseases in which maintaining the engraftment potential of stem cells using short-term ex vivo transduction protocols is critical.


Asunto(s)
Vectores Genéticos , VIH-1/genética , Células Madre Hematopoyéticas/fisiología , Lentivirus/genética , Transducción Genética/métodos , Animales , Antígenos CD34/metabolismo , Linaje de la Célula/fisiología , Expresión Génica , Terapia Genética/métodos , Supervivencia de Injerto , Trasplante de Células Madre Hematopoyéticas , Técnicas In Vitro , Macaca nemestrina , Transgenes/genética
15.
Methods Mol Biol ; 506: 341-61, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19110637

RESUMEN

Large animal models have played a crucial role in the development of gene therapy protocols. A significant advantage of large animal models over rodent models includes the ability to more easily translate protocols developed in large animals to humans. For gene therapy applications, nonhuman primates and canines have been the main large animal models. Canines have the advantage that there are disease models available, e.g., hemolytic anemia (pyruvate kinase deficiency), leukocyte adhesion deficiency, severe combined immunodeficiency (XSCID), storage diseases, and others. In addition, all three major integrating virus systems, i.e., gammaretrovirus-, HIV-derived lenti- and foamy virus vectors are able to efficiently transduce canine hematopoietic cells. Here we describe protocols developed for efficient transduction of canine hematopoietic repopulating cells.


Asunto(s)
Hematopoyesis/genética , Modelos Animales , Animales , Antígenos CD34/inmunología , Células Cultivadas , Perros , Citometría de Flujo , Terapia Genética , Vectores Genéticos , Células Madre Hematopoyéticas/inmunología , Retroviridae/genética , Transducción Genética
16.
Cancer Res ; 67(18): 8783-90, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17875719

RESUMEN

Glioblastoma multiforme (GBM) is the most aggressive brain tumor, and patients rarely survive for more than 2 years. Gene therapy may offer new treatment options and improve the prognosis for patients with GBM. Adenovirus-mediated gene therapy strategies for brain tumors have been limited by inefficient gene transfer due to low expression of the adenovirus serotype 5 (Ad5) receptor. We have used an adenovirus vector that specifically replicates in tumor cells and uses an Ad5 capsid and the adenovirus serotype (Ad35) fiber for efficient infection of malignant tumor cells. This vector also expresses adenovirus E1A and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in a tumor-specific manner. Here, we show that this oncolytic vector (Ad5/Ad35.IR-E1A/TRAIL) efficiently infects the GBM tumor cell lines SF767, T98G, and U-87 MG. Tumor cell killing was markedly enhanced with Ad5/Ad35.IR-E1A/TRAIL compared with wild-type Ad5 and Ad35 virus or Ad5/Ad35.IR-E1A- vectors without TRAIL expression in vitro. In vivo experiments using s.c. xenografted U-87 MG cells in NOD/SCID mice showed a significant growth delay of tumors after i.t. injection of Ad5/Ad35.IR-E1A/TRAIL, whereas adenovirus wild-type injections showed only marginal or no effect. Our findings indicate that the use of a capsid-modified adenoviral vector, in combination with TRAIL expression, is a promising novel approach for gene therapy of glioblastoma.


Asunto(s)
Adenoviridae/fisiología , Cápside/metabolismo , Glioblastoma/terapia , Glioblastoma/virología , Viroterapia Oncolítica/métodos , Ligando Inductor de Apoptosis Relacionado con TNF/genética , Adenoviridae/genética , Adenoviridae/metabolismo , Adenoviridae/patogenicidad , Infecciones por Adenoviridae/metabolismo , Infecciones por Adenoviridae/patología , Infecciones por Adenoviridae/virología , Proteínas E1A de Adenovirus/genética , Animales , Apoptosis/fisiología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Terapia Genética/métodos , Vectores Genéticos/genética , Glioblastoma/genética , Glioblastoma/patología , Humanos , Proteína Cofactora de Membrana/biosíntesis , Proteína Cofactora de Membrana/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Receptores Virales/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/biosíntesis , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Hum Gene Ther ; 19(1): 103-7, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18092920

RESUMEN

In previous studies we showed that low-dose irradiation and immunosuppression with cyclosporine and mycophenolate mofetil prolonged in vivo persistence of gene-modified T cells but was unable to induce tolerance. We hypothesized that the lack of sustained antigen presentation because of the limited life span of the infused T cells might be responsible for the lack of tolerance induction. Thus, we examined whether tolerance could be induced by infusion of long-lived stem cells. Two baboons were transplanted with YFP/neo-transduced CD34+ cells. The transgene-marked cells disappeared completely within 5 weeks and CD8+ transgene-specific cytotoxic T lymphocytes were detected in both animals. Thus, this nonablative conditioning regimen did not provide sufficient immunosuppression for the induction of tolerance after infusion of gene-modified CD34+ cells.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Linfocitos T Citotóxicos/inmunología , Transgenes/inmunología , Acondicionamiento Pretrasplante , Animales , Antígenos CD34/análisis , Células Madre Hematopoyéticas/química , Papio
18.
Mol Ther ; 15(7): 1356-65, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17440443

RESUMEN

The potential for leukemia caused by retroviral vector integration has become a significant concern for hematopoietic stem cell gene therapy. We analyzed the distribution of vector integrants in pigtailed macaque and baboon repopulating cells for the two most commonly used retroviral vector systems, human immunodeficiency virus (HIV)-based lentiviral vectors and murine leukemia virus (MLV)-based gammaretroviral vectors, to help define their relative genotoxicity. All animals had polyclonal engraftment with no apparent adverse effects from transplantation with gene-modified cells. In all, 380 MLV and 235 HIV unique vector integration sites were analyzed and had distinct distribution patterns in relation to genes and CpG islands as observed in previous in vitro studies. Both vector types were found more frequently in and near proto-oncogenes in repopulating cells than in a random dataset. Analysis of functional classes of genes with integrants within 100 kilobases (kb) of their transcription start sites showed an over-representation of genes involved in growth or survival near both lentiviral and gammaretroviral integrants. Microarray analysis showed that both gammaretroviral and lentiviral vectors were found close to genes with high expression levels in primitive cells enriched for hematopoietic stem cells. These data help define the relative risk of insertional mutagenesis with MLV-, HIV-, and simian immunodeficiency virus (SIV)-based vectors in a highly relevant primate model.


Asunto(s)
Vectores Genéticos/genética , VIH/genética , Virus de la Leucemia Murina/genética , Macaca nemestrina/genética , Papio/genética , Integración Viral/genética , Animales , Células Sanguíneas/metabolismo , Islas de CpG , Expresión Génica , Genoma/genética , Proto-Oncogenes/genética , Transcripción Genética/genética
19.
Exp Hematol ; 35(7): 1132-9, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17588482

RESUMEN

OBJECTIVE: Recent studies in the mouse model have shown improved engraftment of repopulating cells when cells were administered by intramarrow (IM) vs intravenous (IV) injection. Here we wished to determine if IM injection was feasible and would result in improved engraftment in a clinically relevant large animal model. MATERIALS AND METHODS: We used a competitive repopulation assay to directly compare IM vs IV injection in four baboons. CD34+ autologous bone marrow cells were split into two equal fractions and transduced with either green fluorescent protein (GFP) or yellow fluorescent protein (YFP). Gene-marked cells were infused by IM or IV administration after myeloablative irradiation. RESULTS: Peripheral blood granulocyte marking peaked at 2 to 3 weeks after transplantation and decreased thereafter before stabilizing. In all animals, marking levels of IM-injected cells (GFP) were lower than those of IV-injected cells (YFP) early after transplantation. However, in two of the four monkeys, GFP marking steadily increased after 2 months resulting in higher marking levels from IM-injected cells. In one animal, this trend sustained up to the last follow-up at 1 year after transplantation, with marking levels of 63.4% and 9.7% from IM- and IV-injected cells, respectively. Transplantation of both IM- and IV-injected CD34+ cells resulted in polyclonal multilineage engraftment. CONCLUSION: Our data show efficient gene marking after IM injection and suggest a different engraftment profile for IM- vs IV-injected repopulating cells.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Animales , Marcadores Genéticos , Inyecciones Intravenosas , Ratones , Ratones SCID , Papio , Retroviridae/genética , Integración Viral
20.
Hum Gene Ther ; 18(5): 451-6, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17536975

RESUMEN

We have previously demonstrated successful in vivo selection, chemoprotection, and modulation of donor chimerism in dogs that received myeloablative allogeneic stem cell transplantation with cells expressing the P140K mutant of the DNA repair enzyme methylguanine methyltransferase (MGMTP140K). Here, we wished to investigate whether in vivo selection, chemoprotection, and modulation of donor chimerism could also be achieved after nonmyeloablative transplantation, which could allow for less toxic transplantation regimens for patients with malignant and genetic diseases. Three dogs received a nonmyeloablative conditioning regimen and infusion of allogeneic stem cells transduced with MGMTP140K. All three dogs had stable gene marking and donor chimerism before receiving a course of O(6) -benzylguanine (O(6) BG)/1,3-bis(2-chloroethyl)-1-nitrosourea (BCNU) between days 210 and 589 after transplantation. One to four doses led to a marked increase in gene marking in all dogs. Furthermore, the transduced cells conferred chemoprotection and prevented severe neutropenia. Our results suggest that drug resistance gene therapy is feasible and safe in the nonmyeloablative transplantation setting.


Asunto(s)
Resistencia a Medicamentos/genética , Terapia Genética/métodos , Trasplante de Células Madre , Animales , Secuencia de Bases , Carmustina/uso terapéutico , Cartilla de ADN/genética , Perros , Vectores Genéticos , Refuerzo Inmunológico de Injertos , Lentivirus/genética , Quimera por Trasplante , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA