Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Nat Commun ; 8(1): 389, 2017 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-28855514

RESUMEN

Therapies based on immune cells have been applied for diseases ranging from cancer to diabetes. However, the viral and electroporation methods used to create cytoreagents are complex and expensive. Consequently, we develop targeted mRNA nanocarriers that are simply mixed with cells to reprogram them via transient expression. Here, we describe three examples to establish that the approach is simple and generalizable. First, we demonstrate that nanocarriers delivering mRNA encoding a genome-editing agent can efficiently knock-out selected genes in anti-cancer T-cells. Second, we imprint a long-lived phenotype exhibiting improved antitumor activities into T-cells by transfecting them with mRNAs that encode a key transcription factor of memory formation. Third, we show how mRNA nanocarriers can program hematopoietic stem cells with improved self-renewal properties. The simplicity of the approach contrasts with the complex protocols currently used to program therapeutic cells, so our methods will likely facilitate manufacturing of cytoreagents.Current widely used viral and electroporation methods for creating therapeutic cell-based products are complex and expensive. Here, the authors develop targeted mRNA nanocarriers that can transiently program gene expression by simply mixing them with cells, to improve their therapeutic potential.


Asunto(s)
Técnicas de Reprogramación Celular , ARN Mensajero/química , Animales , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Femenino , Edición Génica/métodos , Técnicas de Inactivación de Genes , Impresión Genómica , Células Madre Hematopoyéticas/citología , Humanos , Células Jurkat , Células K562 , Leucocitos Mononucleares , Ratones Endogámicos NOD , Nanopartículas/uso terapéutico , Prueba de Estudio Conceptual , Linfocitos T/citología , Factores de Transcripción/genética , Transfección/métodos
2.
Bone Marrow Transplant ; 52(4): 570-573, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28067886

RESUMEN

We developed a haploidentical transplantation protocol with post-transplant cyclophosphamide (CY) for in vivo T-cell depletion (TCD) using a novel adapted-dosing schedule (25 mg/kg on days +3 and +4) for Fanconi anemia (FA). With median follow-up of 3 years (range, 37 days to 6.2 years), all six patients engrafted. Two patients with multiple pre-transplant comorbidities died, one from sepsis and one from sepsis with associated chronic GVHD. Four patients without preexisting comorbidities and early transplant referrals are alive with 100% donor chimerism and excellent performance status. We conclude that adjusted-dosing post-transplant CY is effective in in vivo TCD to promote full donor engraftment in patients with FA.


Asunto(s)
Ciclofosfamida/administración & dosificación , Anemia de Fanconi/terapia , Depleción Linfocítica/métodos , Trasplante Haploidéntico/métodos , Niño , Preescolar , Esquema de Medicación , Anemia de Fanconi/mortalidad , Femenino , Humanos , Inmunosupresores/administración & dosificación , Masculino , Linfocitos T
3.
Bone Marrow Transplant ; 51(12): 1573-1578, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27479689

RESUMEN

A total of 21 patients with severe aplastic anemia (SAA) underwent marrow transplantation from HLA-identical siblings following a standard conditioning regimen with cyclophosphamide (50 mg/kg/day × 4 days) and horse antithymocyte globulin (30 mg/kg/day × 3 days). Post-grafting immunosuppression consisted of a short course of methotrexate (MTX) combined with cyclosporine (CSP). The transplant protocol tested the hypothesis that the incidence of chronic GvHD could be reduced by limiting the marrow grafts to ⩽2.5 × 108 nucleated marrow cells/kg. None of the patients rejected the graft, all had sustained engraftment and all are surviving at a median of 4 (range 1-8) years after transplantation. Chronic GvHD developed in 16% of patients given ⩽2.5 × 108 nucleated marrow cells/kg. Post-grafting immunosuppression has been discontinued in 20 of the 21 patients. In conclusion, limiting the number of transplanted marrow cells may have resulted in minimal improvement in the incidence and severity of chronic GvHD.


Asunto(s)
Anemia Aplásica/terapia , Trasplante de Médula Ósea/métodos , Recuento de Células , Enfermedad Injerto contra Huésped/prevención & control , Adolescente , Adulto , Anemia Aplásica/complicaciones , Niño , Preescolar , Femenino , Supervivencia de Injerto , Prueba de Histocompatibilidad , Humanos , Terapia de Inmunosupresión/métodos , Masculino , Persona de Mediana Edad , Hermanos , Resultado del Tratamiento , Adulto Joven
4.
Gene Ther ; 22(12): 1007-12, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26355737

RESUMEN

Development of curative approaches for HIV-1 infected patients requires novel approaches aimed at eliminating viral reservoirs and replacing potential target cells with infection-resistant immune cell populations. We have previously shown that autologous transplantation of genetically modified hematopoietic stem cells (HSCs) with lentiviral vectors encoding the mC46-fusion inhibitor results in a significant reduction in viral pathogenesis following challenge with the highly pathogenic dual tropic, SHIV89.6P strain. In this study, we used a combinatorial approach in which following engraftment of genetically modified HSCs, pigtailed macaques were vaccinated with a previously developed vaccinia-based vaccine expressing SIV-Gag, Pol. Using this dual therapy approach, lower viremia was detected in both the acute and chronic phase of disease with levels reaching near the lower limits of detection. In comparison with macaques receiving HSCT only, the combination approach resulted in a further log decrease in plasma viremia. Similar to our previous studies, positive selection of all CD4(+) T-cell subsets was observed; however, higher gene-modified CD4(+) T-cell levels were observed during the chronic phase when vaccination was included suggesting that combining vaccination with HSCT may lower the necessary threshold for achieving viremic control.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Vacunas contra el SIDAS/farmacología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Animales , Linfocitos T CD4-Positivos/inmunología , Macaca nemestrina , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Subgrupos de Linfocitos T/inmunología , Vacunas de ADN/inmunología , Vacunas de ADN/farmacología , Carga Viral
5.
Gene Ther ; 22(7): 591-5, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25786870

RESUMEN

Foamy virus (FV) vectors are promising for hematopoietic stem cell (HSC) gene therapy but preclinical data on the clonal composition of FV vector-transduced human repopulating cells is needed. Human CD34(+) human cord blood cells were transduced with an FV vector encoding a methylguanine methyltransferase (MGMT)P140K transgene, transplanted into immunodeficient NOD/SCID IL2Rγ(null) mice, and selected in vivo for gene-modified cells. The retroviral insertion site profile of repopulating clones was examined using modified genomic sequencing PCR. We observed polyclonal repopulation with no evidence of clonal dominance even with the use of a strong internal spleen focus forming virus promoter known to be genotoxic. Our data supports the use of FV vectors with MGMTP140K for HSC gene therapy but also suggests additional safety features should be developed and evaluated.


Asunto(s)
Inmunodeficiencia Combinada Grave/genética , Spumavirus/genética , Integración Viral/genética , Animales , Pruebas Genéticas/métodos , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Humanos , Ratones Endogámicos NOD , Ratones SCID , Acondicionamiento Pretrasplante
6.
Gene Ther ; 20(7): 695-702, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23364313

RESUMEN

The 'Berlin Patient', who maintains suppressed levels of HIV viremia in the absence of antiretroviral therapy, continues to be a standard bearer in HIV eradication research. However, the unique circumstances surrounding his functional cure are not applicable to most HIV(+) patients. To achieve a functional or sterilizing cure in a greater number of infected individuals worldwide, combinatorial treatments, targeting multiple stages of the viral life cycle, will be essential. Several anti-HIV gene therapy approaches have been explored recently, including disruption of the C-C chemokine receptor 5 (CCR5) and CXC chemokine receptor 4 (CXCR4) coreceptor loci in CD4(+) T cells and CD34(+) hematopoietic stem cells. However, less is known about the efficacy of these strategies in patients and more relevant HIV model systems such as non-human primates (NHPs). Combinatorial approaches, including genetic disruption of integrated provirus, functional enhancement of endogenous restriction factors and/or the use of pharmacological adjuvants, could amplify the anti-HIV effects of CCR5/CXCR4 gene disruption. Importantly, delivering gene disruption molecules to genetic sites of interest will likely require optimization on a cell type-by-cell type basis. In this review, we highlight the most promising gene therapy approaches to combat HIV infection, methods to deliver these therapies to hematopoietic cells and emphasize the need to target viral replication pre- and post-entry to mount a suitably robust defense against spreading infection.


Asunto(s)
Terapia Antirretroviral Altamente Activa , Infecciones por VIH/genética , Infecciones por VIH/terapia , Replicación Viral/genética , Antígenos CD34/genética , Antígenos CD34/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Terapia Genética , Infecciones por VIH/virología , VIH-1/genética , VIH-1/patogenicidad , Células Madre Hematopoyéticas/citología , Humanos , Terapia Molecular Dirigida , Receptores CCR5/genética , Receptores CXCR4/genética
7.
Pediatr Hematol Oncol ; 29(6): 568-78, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22839094

RESUMEN

Allogeneic hematopoietic cell transplantation (HCT) is the only known cure for patients with Fanconi anemia (FA) who develop aplasia or leukemia. However, transplant regimens typically contain high-dose alkylators, which are poorly tolerated in FA patients. Furthermore, as many patients lack human leukocyte antigen (HLA)-matched family donors, alternative donors are used, which can increase the risk of both graft rejection and graft-versus-host disease (GVHD). To improve on these three concerns, we developed a multi-institutional clinical trial using a fludarabine (FLU)-based conditioning regimen with limited alkylators/low-dose radiation, HLA-haploidentical marrow, followed by reduced-dose cyclophosphamide (CY) to treat three FA patients with aplasia. All three patients engrafted with 100% donor CD3 chimerism at 1 month. One patient died early from disseminated toxoplasmosis infection. Of the two survivors, one had significant pretransplant co-morbidities and inadequate immunosuppression, and developed severe acute GVHD. The other patient had only mild acute and no chronic GVHD. With a follow-up of 2 and 3 years, respectively, both patients are doing well, are transfusion-independent, and maintain full donor chimerism. The patient with severe GVHD has resolving oral GVHD and good quality of life. We conclude that using low-intensity conditioning, HLA-haploidentical marrow, and reduced-dose CY for in vivo T-cell depletion can correct life-threatening aplasia in FA patients.


Asunto(s)
Anemia de Fanconi/terapia , Rechazo de Injerto/prevención & control , Enfermedad Injerto contra Huésped/prevención & control , Antígenos HLA/inmunología , Trasplante de Células Madre Hematopoyéticas , Depleción Linfocítica , Linfocitos T/inmunología , Vidarabina/análogos & derivados , Adolescente , Antineoplásicos/uso terapéutico , Niño , Terapia Combinada , Anemia de Fanconi/inmunología , Femenino , Estudios de Seguimiento , Humanos , Quimera por Trasplante/inmunología , Acondicionamiento Pretrasplante , Trasplante Homólogo , Vidarabina/uso terapéutico
8.
Cancer Gene Ther ; 19(8): 523-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22627392

RESUMEN

Chemotherapy with 1,3-bis (2-chloroethyl)-1-nitrosourea (BCNU) and temozolomide (TMZ) is commonly used for the treatment of glioblastoma multiforme (GBM) and other cancers. In preparation for a clinical gene therapy study in patients with glioblastoma, we wished to study whether these reagents could be used as a reduced-intensity conditioning regimen for autologous transplantation of gene-modified cells. We used an MGMT(P140K)-expressing lentivirus vector to modify dog CD34(+) cells and tested in four dogs whether these autologous cells engraft and provide chemoprotection after transplantation. Treatment with O(6)-benzylguanine (O6BG)/TMZ after transplantation resulted in gene marking levels up to 75%, without significant hematopoietic cytopenia, which is consistent with hematopoietic chemoprotection. Retrovirus integration analysis showed that multiple clones contribute to hematopoiesis. These studies demonstrate the ability to achieve stable engraftment of MGMT(P140K)-modified autologous hematopoietic stem cells (HSCs) after a novel reduced-intensity conditioning protocol using a combination of BCNU and TMZ. Furthermore, we show that MGMT(P140K)-HSC engraftment provides chemoprotection during TMZ dose escalation. Clinically, chemoconditioning with BCNU and TMZ should facilitate engraftment of MGMT(P140K)-modified cells while providing antitumor activity for patients with poor prognosis glioblastoma or alkylating agent-sensitive tumors, thereby supporting dose-intensified chemotherapy regimens.


Asunto(s)
Carmustina/uso terapéutico , Metilasas de Modificación del ADN , Enzimas Reparadoras del ADN , Dacarbazina/análogos & derivados , Glioblastoma/tratamiento farmacológico , Trasplante de Células Madre Hematopoyéticas , Proteínas Supresoras de Tumor , Animales , Metilasas de Modificación del ADN/genética , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Dacarbazina/uso terapéutico , Perros , Terapia Genética , Hematopoyesis , Humanos , Lentivirus , Temozolomida , Acondicionamiento Pretrasplante , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
9.
Vet Pathol ; 48(5): 999-1001, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20870955

RESUMEN

Gammaretroviral vectors are an efficient means to effect gene therapy. However, genotoxicity from insertion at nonrandom sites can confer a competitive advantage to transduced cells, resulting in clonal proliferation or neoplasia. Six pig-tailed macaques (Macaca nemestrina) underwent total body irradiation and reconstitution with autologous stem cells genetically modified by a gammaretroviral vector overexpressing HOXB4. Two animals were euthanized owing to irradiation- or transplantation-associated toxicity, whereas the other 4 had successful reconstitution. Of the 4 macaques with successful reconstitution, 1 has no long-term follow-up information; 1 was euthanized owing to infection with simian varicella virus infection 18 months post-total body irradiation; and the 2 others are described herein as case Nos. 1 and 2. After being stable for 3 years, case No. 1 developed pancytopenia and petechiation, and after 2 years of stability case No. 2 developed anemia and thrombocytopenia. Despite therapy, the animals deteriorated and were euthanized. Gross findings included emaciation; case No. 1 also had hemorrhage, peritonitis, and cholecystitis. Histologically, bone marrow was hypercellular with predominately blast cells of all hematopoietic lineages, though with myeloid predominance, and with maturation arrest and blast cell dysplasia (myelodysplasia). Myelodysplasia was likely from a combination of insertional mutagenesis by the retroviral vector and overexpression of HOXB4. Consequences of myelodysplasia included the blood dyscrasias and, in case No. 1, hemorrhage, bacterial cholecystitis, hepatitis, and peritonitis.


Asunto(s)
Proteínas de Homeodominio/genética , Macaca nemestrina , Enfermedades de los Monos/patología , Síndromes Mielodisplásicos/veterinaria , Factores de Transcripción/genética , Animales , Resultado Fatal , Terapia Genética/efectos adversos , Terapia Genética/métodos , Terapia Genética/veterinaria , Vectores Genéticos , Masculino , Enfermedades de los Monos/genética , Mutagénesis Insercional/métodos , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología
10.
Bone Marrow Transplant ; 46(4): 539-44, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20581880

RESUMEN

A nonmyeloablative conditioning regimen consisting of fludarabine (FLU) and 2 Gy TBI has been used extensively and with substantial engraftment success without promoting excessive nonrelapse mortality in medically infirm patients requiring hematopoietic cell transplantation. In this paper, we studied this same low-toxicity regimen as a means of promoting engraftment of unrelated donor hematopoietic cell transplantation in patients with Fanconi anemia (FA). All patients tolerated the regimen well with no mucositis or other severe toxicities. Of six patients transplanted, five achieved stable mixed or full donor chimerism. Acute and chronic GVHD occurred in four and three patients, respectively. Three patients are alive and well at a median of 45.9 (range, 20.9-68.1) months after transplant. In summary, this FLU-based regimen facilitates stable engraftment of unrelated PBSCs, but is associated with significant chronic GVHD.


Asunto(s)
Anemia de Fanconi/terapia , Trasplante de Células Madre Hematopoyéticas/métodos , Acondicionamiento Pretrasplante/efectos adversos , Vidarabina/análogos & derivados , Irradiación Corporal Total , Niño , Anemia de Fanconi/tratamiento farmacológico , Femenino , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Masculino , Tasa de Supervivencia , Donantes de Tejidos , Quimera por Trasplante , Acondicionamiento Pretrasplante/métodos , Resultado del Tratamiento , Vidarabina/administración & dosificación , Vidarabina/toxicidad , Irradiación Corporal Total/efectos adversos
11.
Gene Ther ; 17(10): 1244-52, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20485382

RESUMEN

One of the major hurdles for the development of gene therapy for Fanconi anemia (FA) is the increased sensitivity of FA stem cells to free radical-induced DNA damage during ex vivo culture and manipulation. To minimize this damage, we have developed a brief transduction procedure for lentivirus vector-mediated transduction of hematopoietic progenitor cells from patients with Fanconi anemia complementation group A (FANCA). The lentiviral vector FancA-sW contains the phosphoglycerate kinase promoter, the FANCA cDNA, and a synthetic, safety-modified woodchuck post transcriptional regulatory element (sW). Bone marrow mononuclear cells or purified CD34(+) cells from patients with FANCA were transduced in an overnight culture on recombinant fibronectin peptide CH-296, in low (5%) oxygen, with the reducing agent, N-acetyl-L-cysteine (NAC), and a combination of growth factors, granulocyte colony-stimulating factor (G-CSF), Flt3 ligand, stem cell factor, and thrombopoietin. Transduced cells plated in methylcellulose in hypoxia with NAC showed increased colony formation compared with 21% oxygen without NAC (P<0.03), showed increased resistance to mitomycin C compared with green fluorescent protein (GFP) vector-transduced controls (P<0.007), and increased survival. Thus, combining short transduction and reducing oxidative stress may enhance the viability and engraftment of gene-corrected cells in patients with FANCA.


Asunto(s)
Proteína del Grupo de Complementación A de la Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Terapia Genética/métodos , Lentivirus/genética , Acetilcisteína/metabolismo , Acetilcisteína/farmacología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Anemia de Fanconi/patología , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Mitomicina/farmacología , Transducción Genética
12.
Gene Ther ; 17(8): 939-48, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20428209

RESUMEN

Large animal models have been instrumental in advancing hematopoietic stem cell (HSC) gene therapy. Here we review the advantages of large animal models, their contributions to the field of HSC gene therapy and recent progress in this field. Several properties of human HSCs including their purification, their cell-cycle characteristics, their response to cytokines and the proliferative demands placed on them after transplantation are more similar in large animal models than in mice. Progress in the development and use of retroviral vectors and ex vivo transduction protocols over the last decade has led to efficient gene transfer in both dogs and nonhuman primates. Importantly, the approaches developed in these models have translated well to the clinic. Large animals continue to be useful to evaluate the efficacy and safety of gene therapy, and dogs with hematopoietic diseases have now been cured by HSC gene therapy. Nonhuman primates allow evaluation of aspects of transplantation as well as disease-specific approaches such as AIDS (acquired immunodeficiency syndrome) gene therapy that can not be modeled well in the dog. Finally, large animal models have been used to evaluate the genotoxicity of viral vectors by comparing integration sites in hematopoietic repopulating cells and monitoring clonality after transplantation.


Asunto(s)
Terapia Genética/tendencias , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Modelos Animales , Animales , Daño del ADN , Perros , Técnicas de Transferencia de Gen , Terapia Genética/efectos adversos , Infecciones por VIH/genética , Infecciones por VIH/terapia , Células Madre Hematopoyéticas/virología , Humanos , Ratones , Primates
13.
Gene Ther ; 17(1): 37-49, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19741733

RESUMEN

Highly active antiretroviral therapy has greatly reduced the morbidity and mortality from human immunodeficiency virus (HIV) infection, but AIDS continues to be a serious health problem worldwide. Despite enormous efforts to develop a vaccine, there is still no cure, and alternative approaches including gene therapy should be explored. In this study we developed and compared combinatorial foamy virus (FV) anti-HIV vectors that also express a mutant methylguanine methyltransferase (MGMTP140K) transgene to increase the percentage of gene-modified cells after transplantation. These FV vectors inhibit replication of HIV-1 and also the simian immunodeficiency virus/HIV-1 (SHIV) chimera that can be used in monkey AIDS gene therapy studies. We identified a combinatorial FV vector that expresses 3 anti-HIV transgenes and inhibits viral replication by over 4 logs in a viral challenge assay. This FV anti-HIV vector expresses an HIV fusion inhibitor and two short hairpin RNAs (shRNAs) targeted to HIV-1 tat and rev, and can be produced at high titer (3.8 x 10(7) transducing units ml(-1)) using improved helper plasmids suitable for clinical use. Using a competitive repopulation assay, we show that human CD34(+) cells transduced with this combinatorial FV vector efficiently engraft in a mouse xenotransplantation model, and that the percentage of transduced repopulating cells can be increased after transplantation.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos , VIH-1 , Virus de la Inmunodeficiencia de los Simios , Virus Espumoso de los Simios/genética , Animales , Metilasas de Modificación del ADN/deficiencia , Enzimas Reparadoras del ADN/deficiencia , Técnicas de Transferencia de Gen , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Transducción Genética , Transgenes , Proteínas Supresoras de Tumor/deficiencia , Replicación Viral
14.
Drugs Today (Barc) ; 46(11): 833-46, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21225022

RESUMEN

Management of central nervous system (CNS) malignancies continues to be a therapeutic challenge. Both primary and secondary (metastatic) CNS tumors are frequently resistant to commonly used chemotherapeutic agents. Surgery and radiotherapy provide palliation of symptoms but usually do not lead to curative outcomes. Alkylating agents have been used in the therapy of primary brain cancer for several decades. This group of medications has the ability to penetrate blood-brain barrier, achieving cytotoxic concentrations in cerebrospinal fluid and brain parenchyma. Temozolomide is a second-generation alkylating chemotherapeutic agent, introduced to therapy of primary brain tumors in the 1990s. It has since been approved for the therapy of recurrent and newly diagnosed malignant glioma. Temozolomide offers improved outcomes when used alone or in combination with irradiation. Its role in the therapy of other types of brain cancer, and specifically primary CNS lymphoma, continues to develop. This review will discuss the early stages of development of temozolomide, its introduction into the therapy of glioma, its role in the therapy of elderly patients, mechanisms of resistance and the evolution of its current and future applications.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Dacarbazina/análogos & derivados , Anciano , Animales , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/patología , Neoplasias del Sistema Nervioso Central/tratamiento farmacológico , Neoplasias del Sistema Nervioso Central/patología , Dacarbazina/farmacología , Dacarbazina/uso terapéutico , Resistencia a Antineoplásicos , Glioma/tratamiento farmacológico , Glioma/patología , Humanos , Temozolomida
15.
J Med Primatol ; 38(6): 425-32, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19638102

RESUMEN

PURPOSE: We used total body irradiation (TBI) as conditioning for cord blood transplantation studies in pigtailed macaques. In these studies, different doses of TBI were explored to obtain optimal myelosuppression with acceptable radiation-related side effects. METHODS: Four macaques received TBI ranging from 800 to 1320 cGy, followed by standard post-transplant care. Hematopoietic recovery was monitored by CBC and donor contribution by variable number of tandem repeats analysis. RESULTS: Animals receiving 800 or 1020 cGy TBI tolerated the irradiation well with autologous recovery of neutrophils within 24 days. At a dose of 1200 cGy, neither autologous recovery nor extramedullary toxicity was observed. A fourth animal received 1320 cGy of TBI and suffered significant toxicity necessitating termination of the study. CONCLUSIONS: We conclude that previously considered myeloablative doses of TBI allowed for autologous recovery in the pigtailed macaque, and that a dose of 1200 cGy may be most appropriate, providing both myeloablation and acceptable non-hematopoietic toxicities.


Asunto(s)
Médula Ósea/efectos de la radiación , Trasplante de Células Madre de Sangre del Cordón Umbilical , Irradiación Corporal Total , Animales , Plaquetas/efectos de la radiación , Médula Ósea/patología , Relación Dosis-Respuesta en la Radiación , Femenino , Hematopoyesis , Macaca nemestrina , Neutrófilos/efectos de la radiación , Recuento de Plaquetas , Dosis de Radiación , Traumatismos por Radiación/patología
16.
Gene Ther ; 15(21): 1460-2, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18563183

RESUMEN

We have previously compared the repopulation ability of gene-modified baboon CD34+ cells in an autologous transplantation versus a xenotransplant model in irradiated nonobese diabetic/severe combined immune deficiency (NOD/SCID) mice. Baboon CD34-selected marrow cells were transduced with a gammaretrovirus vector and infused into irradiated baboons and NOD/SCID mice. A limited integration-site analysis could only detect two common retrovirus integration sites in the NOD/SCID and monkey. Here, we performed locus-specific PCR on 30 clones recovered from NOD/SCID beta2-microglobulin mice reconstituted with transduced baboon CD34+ cells. We identified five common integrants in the baboon early after transplant (2-6 weeks) but none during the long-term follow-up (6 and 12 months). These results confirm that repopulating cells in the NOD/SCID mouse contribute only to short-term repopulation in a clinically relevant large animal model.


Asunto(s)
Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Papio , Animales , Antígenos CD34 , División Celular , Células Clonales , Cartilla de ADN/genética , Electroforesis en Gel de Poliacrilamida , Gammaretrovirus/genética , Terapia Genética/veterinaria , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Células Madre Hematopoyéticas/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Modelos Animales , Reacción en Cadena de la Polimerasa/métodos , Tiempo , Transducción Genética/métodos , Trasplante Autólogo , Trasplante Heterólogo
17.
Gene Ther ; 14(21): 1549-54, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17805303

RESUMEN

Several clinical studies of gene-modified T cells have shown limited in vivo function of the cells, immunogenicity of the transgene, and lack of a selective advantage for gene-modified T cells. To address these problems, we developed a lentiviral vector (LV) that provides a selectable, proliferative advantage and potentially decreases immunogenicity for transduced T cells. The bicistronic vector expressed two genes linked with an internal ribosomal entry site. One gene is a variant of the inosine monophosphate dehydrogenase 2, inosine monophosphate dehydrogenase (IMPDH(IY)), conferring resistance to the immunosuppressive drug mycophenolate mofetil (MMF). The other is a suicide gene, herpes simplex virus thymidine kinase (HSV-TK), rendering proliferating cells sensitive to ablation with ganciclovir, fused to the selectable transmembrane marker DeltaCD34 (DeltaCD34/TK). Cells transduced with LV-DeltaCD34/TK.IMPDH(IY) were efficiently enriched by immunomagnetic selection for CD34, proliferated in 0.5-5 microM MMF, and were killed by 0.5-25 microg ml(-1) ganciclovir. We demonstrate efficient selection and killing of gene-modified cells and suggest LV-DeltaCD34/TK.IMPDH(IY)-transduced T cells could be used to facilitate allogeneic hematopoietic cell engraftment. The expression of IMPDH(IY) would allow in vivo selection with MMF, and DeltaCD34/TK expression would allow rapid and safe elimination of transduced T cells if graft-versus-host disease developed.


Asunto(s)
Genes Transgénicos Suicidas , Terapia Genética/métodos , Vectores Genéticos/genética , Herpesvirus Humano 1/enzimología , IMP Deshidrogenasa/genética , Linfocitos T , Timidina Quinasa/genética , Antivirales/uso terapéutico , Proliferación Celular , Clonación Molecular , Resistencia a Medicamentos , Ganciclovir/uso terapéutico , Ingeniería Genética , Vectores Genéticos/administración & dosificación , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas , Humanos , IMP Deshidrogenasa/metabolismo , Separación Inmunomagnética/métodos , Inmunosupresores/farmacología , Células K562 , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/farmacología , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/virología , Transducción Genética/métodos , Trasplante Homólogo
18.
Gene Ther ; 13(4): 369-73, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16208421

RESUMEN

Efficient gene transfer to hematopoietic stem cells by Moloney murine leukemia virus-derived retroviral vectors benefits from ex vivo culture and cytokine support. Both also increase the risks of apoptosis and differentiation among cells targeted for transduction. In an effort to maximize the retention of stem cell properties in target cells, we developed a transduction protocol with a focus on minimizing graft manipulation, cytokine stimulation, and ex vivo exposure duration. Based on their wide host range and ability to transduce quiescent cells, human immunodeficiency virus (HIV)-derived lentivirus vectors are ideally suited for this purpose. Our present studies in a murine model show that whole bone marrow cells are readily transduced after a 1-hour vector exposure in the presence of stem cell factor and CH296 fibronectin fragment. Using this rapid transduction protocol, we achieved long-term, multilineage reconstitution of murine recipients with up to 25% GFP-expressing cells in primary and secondary recipients. Our results demonstrate the unique ability of HIV-derived vectors to transduce hematopoietic stem cells in the absence of enrichment, under minimal cytokine stimulation, and following brief exposures.


Asunto(s)
Médula Ósea/virología , Terapia Genética/métodos , Vectores Genéticos/farmacología , VIH-1/genética , Células Madre Hematopoyéticas/virología , Transducción Genética/métodos , Animales , Recuento de Células , Células Cultivadas , Medios de Cultivo , Fibronectinas/farmacología , Expresión Génica , Vectores Genéticos/genética , Proteínas Fluorescentes Verdes/genética , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/terapia , Ratones , Fragmentos de Péptidos/farmacología , Factor de Células Madre/farmacología , Factores de Tiempo
19.
Cytotherapy ; 7(4): 345-52, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16162456

RESUMEN

BACKGROUND: There is ongoing controversy about the transdifferentiation of hematopoietic stem cells (HSC) into different tissues such as mesenchymal cells. This transdifferentiation or 'plasticity' would be an appealing concept for many therapeutic strategies. While studies in the murine model show encouraging results, reports from clinical allogeneic stem cell transplantations do not support the concept of HSC plasticity. Our aim was to determine whether transplantation of transduced autologous marrow CD34+ cells leads to long-term engraftment of gene-marked cells with mesenchymal characteristics in the baboon. METHODS: We analyzed marrow of two baboons that had received green fluorescence protein (GFP)-marked CD34+ autologous marrow cells after myeloablative conditioning. Marrow was obtained 1 and 2.5 years after transplantation and adherent CD11a- (pan-leukocyte Ab) cells were cultured for 3 weeks. Cultures were then analyzed by flow cytometry and fluorescence microscopy for the presence of GFP+ cells. For further analysis fresh and cultured cells were also labeled with multiple Ab and functional analysis was performed. RESULTS: Both animals showed persistent and stable GFP marking by flow cytometry in peripheral blood leukocytes as well as in CD34+ marrow cells at 1 and 2.5 years after transplantation. There was no evidence of GFP+ mesenchymal cells by either flow cytometry or fluorescence microscopy, while functional and phenotypical analysis identified mesenchymal stem cells in these cultures. DISCUSSION: We conclude that genetically modified CD34+ cells do not contribute to the adherent marrow-derived mesenchymal cell population after autologous transplantation.


Asunto(s)
Antígenos CD34/biosíntesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Animales , Células Cultivadas , Gammaretrovirus , Genes Reporteros , Vectores Genéticos , Células Madre Hematopoyéticas/inmunología , Mesodermo/citología , Papio , Transducción Genética , Trasplante Autólogo
20.
Hum Gene Ther ; 16(6): 725-33, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15960603

RESUMEN

Green fluorescent proteins (GFPs) have been widely used to monitor gene transfer and expression after lentiviral and oncoretroviral transduction of hematopoietic cells. Studies have shown a complete disappearance of GFP-containing cells after transplantation of GFP-transduced repopulating cells in nonhuman primates that was further shown to be mediated by transgene-specific immune responses. We wished to evaluate whether cyclosporine could prevent immune responses to GFP. We first determined whether an immune response to GFP was responsible for the disappearance of gene-modified cells in dogs. We performed immune assays in two dogs transplanted with lentivirally transduced CD34+ cells. Blood samples were obtained twice per week for up to 800 days and the GFP transgene product was measured by flow cytometry in blood leukocytes. Peripheral blood leukocytes were stimulated in vitro for 5 days, using a panel of GFP peptides. Intracellular levels of tumor necrosis factor alpha (TNF-alpha), measured by flow cytometry, and T cell proliferation after GFP peptide stimulation were measured. Dogs that exhibited a decrease in GFP marking developed potent immune responses in vitro to the transgene product GFP as shown by an increase in GFP-specific TNF-alpha production (p < 0.05) when compared with nontransplanted controls. T cells from dogs with low GFP marking exhibited a significant increase in proliferation in response to GFP peptide stimulation in vitro (p < 0.05). To study whether cyclosporine could inhibit the development of GFP-specific immune responses, we treated five dogs with cyclosporine after transplantation of GFP-transduced hematopoietic cells. Dogs treated with cyclosporine after hematopoietic stem cell transplantation showed stable GFP marking in blood leukocytes over 800 days. Our data suggest that cyclosporine prevents immunoactivation against transgene products after transplantation of GFP-transduced hematopoietic stem cells as indicated by stable GFP marking.


Asunto(s)
Ciclosporina/farmacología , Proteínas Fluorescentes Verdes/genética , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/inmunología , Animales , Citocinas/metabolismo , Perros , Proteínas Fluorescentes Verdes/efectos de los fármacos , Proteínas Fluorescentes Verdes/inmunología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/fisiología , Inmunosupresores/farmacología , Leucocitos/efectos de los fármacos , Leucocitos/inmunología , Leucocitos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transducción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA