Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
4.
Gene Ther ; 21(2): 148-57, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24257347

RESUMEN

Reduced intensity conditioning (RIC) is desirable for hematopoietic stem cell (HSC) targeted gene therapy; however, RIC may be insufficient for efficient engraftment and inducing immunological tolerance to transgenes. We previously established long-term gene marking in our rhesus macaque autologous HSC transplantation model following 10 Gy total body irradiation (TBI). In this study, we evaluated RIC transplantation with 4 Gy TBI in two rhesus macaques that received equal parts of CD34(+) cells transduced with green fluorescent protein (GFP)-expressing lentiviral vector and empty vector not expressing transgenes. In both animals, equivalently low gene marking between GFP and empty vectors was observed 6 months post-transplantation, even with efficient transduction of CD34(+) cells in vitro. Autologous lymphocyte infusion with GFP marking resulted in an increase of gene marking in lymphocytes in a control animal with GFP tolerance, but not in the two RIC-transplanted animals. In vitro assays revealed strong cellular and humoral immune responses to GFP protein in the two RIC-transplanted animals, but this was not observed in controls. In summary, 4 Gy TBI is insufficient to permit engraftment of genetically modified HSCs and induce immunological tolerance to transgenes. Our findings should help in the design of conditioning regimens in gene therapy trials.


Asunto(s)
Antígenos CD34/metabolismo , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Supervivencia de Injerto/inmunología , Supervivencia de Injerto/efectos de la radiación , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/inmunología , Irradiación Corporal Total/métodos , Animales , Células Cultivadas , Terapia Combinada , Relación Dosis-Respuesta en la Radiación , Células Madre Hematopoyéticas/efectos de la radiación , Lentivirus/genética , Macaca mulatta , Modelos Animales , Transducción Genética , Transgenes , Acondicionamiento Pretrasplante
5.
Bone Marrow Transplant ; 48(10): 1335-41, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23604009

RESUMEN

Because of the toxicity associated with myeloablative conditioning, nonmyeloablative regimens are increasingly being used in vulnerable patient populations. For patients with sickle cell disease, stable mixed chimerism has proven sufficient to reverse the phenotype. Because the vast majority of patients do not have an HLA-matched sibling, a safe nonmyeloablative regimen that could be applied to the haploidentical setting would be ideal. We employed a mismatched mouse model using BALB/c donors and C57BL/6 recipients. Recipient mice were conditioned with 200 cGy TBI and sirolimus or CSA with or without post transplant Cy (PT-Cy). Our data show that when sirolimus or PT-Cy alone is given to C57BL/6 recipients, donor cells are not detected. However, when sirolimus is administered for 15 or 31 days starting 1 day before or up to 6 days after transplant with PT-Cy, all mice maintain stable mixed chimerism. In contrast, conventional therapy employing CSA with or without PT-Cy does not result in stable mixed chimerism. Lastly, mice with stable mixed chimerism after sirolimus display decreased reactivity to donor Ag both in vitro and in vivo. These data identify a novel strategy for inducing mixed chimerism for the treatment of nonmalignant hematologic diseases.


Asunto(s)
Ciclofosfamida/farmacología , Inmunosupresores/farmacología , Sirolimus/farmacología , Trasplante de Piel/métodos , Animales , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Quimera por Trasplante
6.
Leukemia ; 27(12): 2311-21, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23619564

RESUMEN

Chronic lymphocytic leukemia (CLL) cells depend on microenvironmental factors for proliferation and survival. In particular, the B-cell receptor (BCR) and nuclear factor- κB (NF-κB) pathways are activated in the lymph node (LN) microenvironment. Thus, model systems mimicking tumor-host interactions are important tools to study CLL biology and pathogenesis. We investigated whether the recently established NOD/scid/γc(null) (NSG) mouse xenograft model can recapitulate the effects of the human microenvironment. We assessed, therefore, tumor characteristics previously defined in LN-resident CLL cells, including proliferation, and activation of the BCR and NF-κB pathways. We found that the murine spleen (SP) microenvironment supported CLL cell proliferation and activation to a similar degree than the human LN, including induction of BCR and NF-κB signaling in the xenografted cells. Next, we used this model to study ibrutinib, a Bruton's tyrosine kinase inhibitor in clinical development. Ibrutinib inhibited BCR and NF-κB signaling induced by the microenvironment, decreased proliferation, induced apoptosis and reduced the tumor burden in vivo. Thus, our data demonstrate that the SP of xenografted NSG mice can, in part, recapitulate the role of the human LN for CLL cells. In addition, we show that ibrutinib effectively disrupts tumor-host interactions essential for CLL cell proliferation and survival in vivo.


Asunto(s)
Leucemia Linfocítica Crónica de Células B/patología , Modelos Biológicos , Ensayos Antitumor por Modelo de Xenoinjerto , Adenina/análogos & derivados , Anciano , Animales , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Inmunofenotipificación , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/terapia , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Piperidinas , Pirazoles/farmacología , Pirimidinas/farmacología , Microambiente Tumoral
7.
Gene Ther ; 18(11): 1078-86, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21544097

RESUMEN

Cytokines are required for γ-retroviral transduction of human CD34+ cells. However, cytokines may reduce engraftment of CD34+ cells and may not be necessary for their lentiviral transduction. We sought to optimize transduction and engraftment of human CD34+ cells using lentiviral vectors. Single 24 h transduction of human CD34+ cells with human immunodeficiency virus type 1 (HIV1)-based lentiviral vectors in media containing stem cell factor (SCF), FMS-like tyrosine kinase 3 (FLT3) ligand, thrombopoietin (each 100 ng ml⁻¹) and 10% fetal bovine serum was compared with various cytokine conditions during ex vivo culture and assayed using humanized xenograft mice for 6 months after transplantation. Serum-free media improved transduction efficiency of human CD34+ cells. Interleukin-3 (20 ng ml⁻¹) had little effect on transduction efficiency or engraftment. Threefold higher cytokine mixture (each 300 ng ml⁻¹) reduced engraftment of CD34+ cells. SCF alone (100 ng ml⁻¹) proved insufficient for maintaining engraftment ability and reduced transduction efficiency. Short-term prestimulation had little effect on transduction efficiency or engraftment, yet 24 h prestimulation showed higher transduction efficiency, higher gene expression levels and lower engraftment. In summary, 24 h prestimulation followed by single 24-h lentiviral transduction in serum-free media with SCF, FLT3 ligand and thrombopoietin yields high transduction efficiency to engrafting human CD34+ cells, and is applicable in human clinical gene therapy trials.


Asunto(s)
Antígenos CD34/metabolismo , Trasplante de Células Madre Hematopoyéticas/métodos , Interleucina-3/farmacología , Lentivirus/genética , Transducción Genética , Animales , Medio de Cultivo Libre de Suero , Vectores Genéticos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Modelos Animales , Factor de Células Madre/inmunología , Trombopoyetina/inmunología , Trasplante Heterólogo , Tirosina Quinasa 3 Similar a fms/inmunología
8.
J Clin Endocrinol Metab ; 95(10): E234-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20660050

RESUMEN

AIMS: Diabetes mellitus results from an absolute or relative deficiency of insulin-producing pancreatic ß-cells. The turnover rate of adult human ß-cells remains unknown. We employed two techniques to examine adult human islet ß-cell turnover and longevity in vivo. METHODS: Subjects enrolled in National Institutes of Health clinical trials received thymidine analogs [iododeoxyuridine (IdU) or bromodeoxyuridine (BrdU)] 8 d to 4 yr prior to death. Archival autopsy samples from 10 patients (aged 17-74 yr) were employed to assess ß-cell turnover by scoring nuclear analog labeling within insulin-staining cells. Human adult ß-cell longevity was determined by estimating the cells' genomic DNA integration of atmospheric (14)C. DNA was purified from pancreatic islets isolated from cadaveric donors; whole islet prep DNA was obtained from a 15-yr-old donor, and purified ß-cell DNA was obtained from two donors (ages 48 and 80 yr). (14)C levels were then determined using accelerator mass spectrometry. Cellular "birth date" was determined by comparing the subject's DNA (14)C content relative to a well-established (14)C atmospheric prevalence curve. RESULTS: In the two subjects less than 20 yr of age, 1-2% of the ß-cell nuclei costained for BrdU/IdU. No ß-cell nuclei costained in the eight patients more than 30 yr old. Consistent with the BrdU/IdU turnover data, ß-cell DNA (14)C content indicated that the "birth date" of cells occurred within the subject's first 30 yr of life. CONCLUSIONS: Under typical circumstances, human ß-cells and their cellular precursors are established by young adulthood.


Asunto(s)
Envejecimiento/fisiología , Bromodesoxiuridina/farmacocinética , Proliferación Celular , Células Secretoras de Insulina/fisiología , Datación Radiométrica , Adolescente , Adulto , Anciano , Envejecimiento/metabolismo , Femenino , Humanos , Células Secretoras de Insulina/metabolismo , Masculino , Persona de Mediana Edad , Datación Radiométrica/métodos , Coloración y Etiquetado/métodos , Timidina/análogos & derivados , Timidina/farmacocinética , Donantes de Tejidos , Adulto Joven
9.
Rheumatology (Oxford) ; 46(6): 952-6, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17317716

RESUMEN

OBJECTIVES: To determine the safety and efficacy of a short course of fludarabine combined with cyclophoshamide in lupus nephritis. METHODS: A phase I/II open label pilot study. Thirteen patients with active proliferative lupus nephritis received monthly oral boluses of low-dose cyclophoshamide (0.5 gm/m(2) on day 1) and subcutaneous fludarabine (30 mg/m(2) on days 1-3) for 3-6 cycles. Concomitant prednisone was aggressively tapered from 0.5 mg/kg/day to a low-dose, alternate-day schedule. Patients were followed for at least 24 months after therapy. The primary outcome was the number of patients achieving renal remission defined as stable creatinine, proteinuria <1 gm/day and inactive urine sediment for at least 6 months. RESULTS: The study was terminated early because of bone marrow toxicity. Eleven patients who received at least three cycles were evaluated for efficacy. Ten patients improved markedly with seven patients achieving complete remission and three patients achieving partial remission. There were three serious haematological adverse events during the treatment with one death due to transfusion-associated graft vs host disease. Profound and prolonged CD4 (mean CD4: 98/microl at 7 months and 251/microl at 12 months) and CD20 lymphocytopenia was noted in most patients. Three patients developed Herpes zoster infections. CONCLUSIONS: A short course of low-dose fludarabine and cyclophoshamide can induce long-lasting remissions in patients with proliferative lupus nephritis, but severe myelosuppression limits its widespread use.


Asunto(s)
Ciclofosfamida/uso terapéutico , Inmunosupresores/uso terapéutico , Nefritis Lúpica/tratamiento farmacológico , Vidarabina/análogos & derivados , Adulto , Anciano , Recuento de Linfocito CD4 , Ciclofosfamida/efectos adversos , Esquema de Medicación , Quimioterapia Combinada , Femenino , Humanos , Inmunosupresores/efectos adversos , Linfopenia/inducido químicamente , Masculino , Persona de Mediana Edad , Neutropenia/inducido químicamente , Proyectos Piloto , Proteinuria/tratamiento farmacológico , Resultado del Tratamiento , Vidarabina/efectos adversos , Vidarabina/uso terapéutico
11.
Mol Ther ; 3(6): 911-9, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11407905

RESUMEN

The hematopoietic stem cell has long been considered an ideal target for the introduction of therapeutic genes to treat human disorders such as Fanconi anemia (FA). Although recent progress in large animal models is encouraging, application to nonmalignant conditions is limited by the perceived necessity of myeloablative conditioning. We and others have shown that very low irradiation doses are sufficient to allow significant hematopoietic engraftment in murine hosts even after the introduction of xenogeneic genes. To determine the degree of engraftment of genetically modified cells attainable with very low irradiation doses in larger animals, we employed the rhesus macaque competitive repopulation model. Four animals underwent mobilization with stem cell factor (SCF) and granulocyte colony-stimulating factor (G-CSF) followed by apheresis. The apheresis product was enriched for the CD34-positive fraction by immunomagnetic selection and split equally for transduction with either G1FC26, a retroviral vector carrying the Fanconi anemia complementation group C gene, or PLII, a nonexpression control retroviral vector carrying both neomycin and beta-galactosidase gene sequences modified to prevent translation. Transductions were performed daily in the presence of fresh IL-3, IL-6, SCF, and Flt-3 ligand on fibronectin-coated plates over 96 h. Animals were conditioned with a single dose of either 100 (n = 2) or 200 (n = 2) cGy and received the combined products of transduction on the following day. None of the animals experienced clinically significant neutropenia nor required the use of central line placement, transfusional support with blood products, or intravenous antibiotics. Using real-time PCR, circulating levels of genetically modified cells as high as 1% were initially detected. Stable, albeit, significantly lower levels from both vector-transduced aliquots (<0.1%) persisted beyond 12 months posttransplant in all four animals. Although not sufficient to correct the phenotype in many human disorders, stable low-level engraftment by genetically modified cells following low-intensity conditioning may prove adequate in disorders such as FA due to the selective advantage conferred upon corrected cells.


Asunto(s)
Proteínas de Ciclo Celular , Proteínas de Unión al ADN , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/efectos de la radiación , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/efectos de la radiación , Macaca mulatta/sangre , Proteínas Nucleares , Proteínas/genética , Retroviridae/genética , Acondicionamiento Pretrasplante , Animales , Antígenos CD34/metabolismo , Ensayo de Unidades Formadoras de Colonias , Cartilla de ADN/química , Proteína del Grupo de Complementación C de la Anemia de Fanconi , Proteínas del Grupo de Complementación de la Anemia de Fanconi , Técnicas de Transferencia de Gen , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/virología , Interleucina-3/farmacología , Interleucina-6/farmacología , Proteínas de la Membrana/farmacología , Reacción en Cadena de la Polimerasa , Protectores contra Radiación/farmacología , Transducción Genética , Irradiación Corporal Total
12.
Exp Hematol ; 29(6): 779-85, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11378274

RESUMEN

OBJECTIVE: Previous studies have shown improved engraftment in a murine model when granulocyte colony-stimulating factor (G-CSF) and stem cell factor (SCF) were administered for 5 days prior to irradiation, with significant levels of engraftment in the growth factor-preconditioned group even at very low radiation doses. We sought to explore the mechanisms behind this effect. METHODS: The radiation sensitivity of mice with or without 5 days of prestimulation with G-CSF (200 microg/kg/d) and SCF (50 microg/kg/d) was compared. To further evaluate whether growth factor prestimulation enhances engraftment by mobilization of hematopoietic progenitors into peripheral blood, thus creating less endogenous competition within the marrow compartment, female mice were pretreated with 5 days of G-CSF/SCF or control diluent. Engraftment of 40 x 10(6) peripheral blood stem cells (PBSCs) harvested from G-CSF/SCF-mobilized male mice was compared in the two recipient groups. RESULTS: There was no difference in survival between the pretreated and control mice at the radiation doses tested. Additionally, there was no significant difference in the recovery of blood counts, bone marrow cellularity, colony-forming unit (CFU) content, or stem cell numbers assessed 4 months later in a competitive repopulation model. Engraftment levels of male cells did not differ between G-CSF/SCF-pretreated and control recipients, and could be detected in 30% of recipients at 20-24 weeks (4/12 in each group) at overall levels of 0.1-1%. CONCLUSIONS: The enhanced engraftment in cytokine pretreated recipients is unlikely to be due to increased endogenous stem-cell killing or to the creation of endogenous marrow "space" by egress of endogenous stem cells after cytokine prestimulation.


Asunto(s)
Células de la Médula Ósea/citología , Factor Estimulante de Colonias de Granulocitos/farmacología , Células Madre Hematopoyéticas/citología , Factor de Células Madre/farmacología , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Femenino , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de la radiación , Humanos , Recuento de Leucocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratas , Proteínas Recombinantes/farmacología , Irradiación Corporal Total
13.
Blood ; 97(6): 1888-91, 2001 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11238136

RESUMEN

Transduction of murine stem cells with a multidrug-resistance 1 gene (MDR1) retrovirus results in dramatic ex vivo and in vivo expansion of repopulating cells accompanied by a myeloproliferative disorder. Given the use of MDR1-containing vectors in human trials, investigations have been extended to nonhuman primates. Peripheral blood stem cells from 2 rhesus monkeys were collected, CD34-enriched, split into 2 portions, and transduced with either MDR1 vectors or neo vectors and continued in culture for a total of 10 days before reinfusion. At engraftment, the copy number in granulocytes was extremely high from both MDR vectors and neo vectors, but the copy number fell to 0.01 to 0.05 for both. There were no perturbations of the leukocyte count or differential noted. After 3 cycles of stem cell factor/granulocyte colony-stimulating factor, there were no changes in the levels of MDR1 vector- or neo vector-containing cells. There was no evidence for expansion of MDR1 vector-transduced cells. Long-term engraftment with MDR1 vector- and neo vector-transduced cells occurred despite prolonged culture.


Asunto(s)
Farmacorresistencia Microbiana/genética , Genes MDR/genética , Terapia Genética/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Animales , Técnicas de Cultivo de Célula , División Celular/efectos de los fármacos , Dosificación de Gen , Terapia Genética/normas , Vectores Genéticos/efectos adversos , Vectores Genéticos/normas , Trasplante de Células Madre Hematopoyéticas/normas , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Macaca mulatta , Modelos Animales , Neomicina , Transducción Genética/métodos , Transducción Genética/normas
14.
Lancet ; 356(9241): 1554-9, 2000 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-11075769

RESUMEN

BACKGROUND: High-dose cyclophosphamide has been proposed as an alternative immunosuppressive agent for treatment of severe aplastic anaemia, with a response rate similar to that with regimens containing antithymocyte globulin (ATG) but neither relapse nor clonal haematological complications. We undertook a phase III, prospective, randomised trial to compare response rates to immunosuppression with either high-dose cyclophosphamide plus cyclosporin or conventional immunosuppression with ATG plus cyclosporin in previously untreated patients. METHODS: Between June, 1997, and March, 2000, 31 patients were enrolled. 15 were assigned cyclophosphamide (1 h intravenous infusion of 50 mg/kg daily for 4 days) and 16 were assigned ATG (40 mg/kg daily for 4 days); both groups received cyclosporin, initially at 12 mg/kg daily with adjustment to maintain concentrations at 200-400 microg/L, for 6 months. The primary endpoint was haematological response (no longer meeting criteria for severe aplastic anaemia). The trial was terminated prematurely after three early deaths in the cyclophosphamide group. Analyses were by intention to treat. FINDINGS: Median follow-up was 21.9 months (range 1-33). There was excess morbidity in the cyclophosphamide group (invasive fungal infections, four cyclophosphamide vs no ATG patients; p=0.043) as well as excess early mortality (three deaths within the first 3 months cyclophosphamide vs no ATG patients; p=0.101). There was no significant difference at 6 months after treatment in the overall response rates among evaluable patients (six of 13 [46%] cyclophosphamide vs nine of 12 [75%] ATG). INTERPRETATION: A longer period of observation will be necessary to assess the secondary endpoints of relapse and late clonal complications as well as disease-free and overall survival. However, cyclophosphamide seems a dangerous choice for treatment of this disorder, given the good results achievable with standard therapy.


Asunto(s)
Anemia Aplásica/tratamiento farmacológico , Suero Antilinfocítico/uso terapéutico , Ciclofosfamida/efectos adversos , Ciclosporina/uso terapéutico , Inmunosupresores/efectos adversos , Adulto , Anciano , Anemia Aplásica/mortalidad , Suero Antilinfocítico/administración & dosificación , Ciclofosfamida/administración & dosificación , Ciclofosfamida/uso terapéutico , Ciclosporina/administración & dosificación , Esquema de Medicación , Humanos , Inmunosupresores/administración & dosificación , Inmunosupresores/uso terapéutico , Infusiones Intravenosas , Persona de Mediana Edad , Micosis/etiología , Factores de Riesgo
15.
Mol Ther ; 1(3): 285-93, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10933944

RESUMEN

Low-level retroviral transduction and engraftment of hematopoietic long-term repopulating cells in large animals and humans remain primary obstacles to the successful application of hematopoietic stem cell (HSC) gene transfer in humans. Recent studies have reported improved efficiency by including stromal cells (STR), or the fibronectin fragment CH-296 (FN), and various cytokines such as flt3 ligand (FLT) during ex vivo culture and transduction in nonhuman primates. In this work, we extend our studies using the rhesus competitive repopulation model to further explore optimal and clinically feasible peripheral blood (PB) progenitor cell transduction methods. First, we compared transduction in the presence of either preformed autologous STR or immobilized FN. Long-term clinically relevant gene marking levels in multiple hematopoietic lineages from both conditions were demonstrated in vivo by semiquantitative PCR, colony PCR, and genomic Southern blotting, suggesting that FN could replace STR in ex vivo transduction protocols. Second, we compared transduction on FN in the presence of IL-3, IL-6, stem cell factor (SCF), and FLT (our best cytokine combination in prior studies) with a combination of megakaryocyte growth and development factor (MGDF), SCF, and FLT. Gene marking levels were equivalent in these animals, with no significant effect on retroviral gene transfer efficiency assessed in vivo by the replacement of IL-3 and IL-6 with MGDF. Our results indicate that SCF/G-CSF-mobilized PB CD34+ cells are transduced with equivalent efficiency in the presence of either STR or FN, with stable long-term marking of multiple lineages at levels of 10-15% and transient marking as high as 54%. These results represent an advance in the field of HSC gene transfer using methods easily applied in the clinical setting.


Asunto(s)
Antígenos CD34/genética , Técnicas de Transferencia de Gen , Células Madre Hematopoyéticas/metabolismo , Retroviridae/genética , Animales , Southern Blotting , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Fibronectinas/metabolismo , Factor Estimulante de Colonias de Granulocitos/farmacología , Interleucina-3/farmacología , Interleucina-6/farmacología , Macaca mulatta , Proteínas de la Membrana/farmacología , Reacción en Cadena de la Polimerasa , Factor de Células Madre/farmacología , Células del Estroma/metabolismo , Trombopoyetina/farmacología
16.
Mol Ther ; 1(6): 533-44, 2000 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10933978

RESUMEN

Host immune responses against foreign transgenes may be a major obstacle to successful gene therapy. To clarify the impact of an immune response to foreign transgene products on the survival of genetically modified cells, we studied the in vivo persistence of cells transduced with a vector expressing a foreign transgene compared to cells transduced with a nonexpressing vector in the clinically predictive rhesus macaque model. We constructed retroviral vectors containing the neomycin phosphotransferase gene (neo) sequences modified to prevent protein expression (nonexpressing vectors). Rhesus monkey lymphocytes or hematopoietic stem cells (HSCs) were transduced with nonexpressing and neo-expressing vectors followed by reinfusion, and their in vivo persistence was studied. While lymphocytes transduced with a nonexpressing vector could be detected for more than 1 year, lymphocytes transduced with a neo-expressing vector were no longer detectable within several weeks of infusion. However, five of six animals transplanted with HSCs transduced with nonexpression or neo-expression vectors, and progeny lymphocytes marked with either vector persisted for more than 2 years. Furthermore, in recipients of transduced HSCs, infusion of mature lymphocytes transduced with a second neo-expressing vector did not result in elimination of the transduced lymphocytes. Our data show that introduction of a xenogeneic gene via HSCs induces tolerance to the foreign gene products. HSC gene therapy is therefore suitable for clinical applications where long-term expression of a therapeutic or foreign gene is required.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Tolerancia Inmunológica , Animales , Secuencia de Bases , Transfusión de Sangre Autóloga , Cartilla de ADN/genética , Expresión Génica , Terapia Genética/métodos , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Kanamicina Quinasa/genética , Kanamicina Quinasa/inmunología , Transfusión de Linfocitos , Linfocitos/inmunología , Linfocitos/metabolismo , Macaca mulatta , Modelos Biológicos , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Transducción Genética , Trasplante Autólogo
17.
Blood ; 96(1): 1-8, 2000 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-10891424

RESUMEN

Retroviral insertion site analysis was used to track the contribution of retrovirally transduced primitive progenitors to hematopoiesis after autologous transplantation in the rhesus macaque model. CD34-enriched mobilized peripheral blood cells were transduced with retroviral marking vectors containing the neo gene and were reinfused after total body irradiation. High-level gene transfer efficiency allowed insertion site analysis of individual myeloid and erythroid colony-forming units (CFU) and of highly purified B- and T-lymphoid populations in 2 animals. At multiple time points up to 1 year after transplantation, retroviral insertion sites were identified by performing inverse polymerase chain reaction and sequencing vector-containing CFU or more than 99% pure T- and B-cell populations. Forty-eight unique insertion sequences were detected in the first animal and also in the second animal, and multiple clones contributed to hematopoiesis at 2 or more time points. Multipotential clones contributing to myeloid and lymphoid lineages were identified. These results support the concept that hematopoiesis in large animals is polyclonal and that individual multipotential stem or progenitor cells can contribute to hematopoiesis for prolonged periods. Gene transfer to long-lived, multipotent clones is shown and is encouraging for human gene therapy applications.


Asunto(s)
Linfocitos B/citología , Hematopoyesis , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Linfocitos T/citología , Animales , Antígenos CD34/sangre , Diferenciación Celular , Ensayo de Unidades Formadoras de Colonias , Técnicas de Transferencia de Gen , Genes Reporteros , Terapia Genética/métodos , Vectores Genéticos , Movilización de Célula Madre Hematopoyética , Humanos , Kanamicina Quinasa/genética , Macaca mulatta , Retroviridae , Transfección , Irradiación Corporal Total
18.
Semin Hematol ; 37(1): 102-9, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10676915

RESUMEN

Severe aplastic anemia (SAA) has a poor prognosis in the absence of treatment. Current accepted therapeutic strategies include allogeneic stem-cell transplantation and immunosuppression, both resulting in long-term survival in the majority of patients. Although human leukocyte antigen (HLA)-matched sibling stem-cell transplantation is highly effective, the 25% probability of finding a suitable sibling donor within a family renders this approach available to only a minority of patients. Transplantation using HLA-matched, unrelated donors carries a high risk of treatment failure along with considerable toxicity. While combined immunosuppression with both antithymocyte globulin (ATG) and cyclosporine A (CSA) produces hematologic improvement in most patients, relapse is common. Late evolution of aplastic anemia to other serious hematologic disorders, including paroxysmal nocturnal hemoglobinuria (PNH), myelodysplasia, and acute leukemia, is also a significant problem following treatment with ATG/CSA. Recently, results of immunosuppression in SAA with another potent immunosuppressive agent, cyclophosphamide, were reported in a small number of patients. The overall response rate was similar to that seen with ATG/CSA, but relapse and late clonal disease were not observed during a long period of follow-up. A larger randomized trial comparing sustained hematologic response rates to either conventional immunosuppression with ATG/CSA or high-dose cyclophosphamide and CSA is now underway; secondary end points include response duration, event-free survival, and overall survival. Additionally, a number of protocols designed to test the efficacy of alternative immunosuppressive or immunomodulatory agents are being developed.


Asunto(s)
Anemia Aplásica/tratamiento farmacológico , Ciclofosfamida/uso terapéutico , Inmunosupresores/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Humanos , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/uso terapéutico , Receptores de Interleucina-2/antagonistas & inhibidores , Receptores de Interleucina-2/inmunología , Sirolimus/uso terapéutico , Tacrolimus/uso terapéutico
19.
Br J Haematol ; 108(1): 167-75, 2000 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-10651740

RESUMEN

Diamond-Blackfan anaemia (DBA) is a constitutional pure red cell aplasia presenting in early childhood. In some patients, neutropenia and/or thrombocytopenia have also been observed during the course of the disease. We have followed 28 patients with steroid-refractory DBA for up to 13 years with serial peripheral blood counts and bone marrow (BM) aspirates and biopsies. In 21/28 (75%) patients, moderate to severe generalized BM hypoplasia developed, with overall cellularities ranging from 0% to 30%. Marrow hypoplasia correlated with the development of neutropenia (9/21; 43%) and/or thrombocytopenia (6/21; 29%) in many patients. No patient had either cytogenetic abnormalities or progressed to acute leukaemia, although one 13-year-old developed marked marrow fibrosis and trilineage dysplasia. We used the in vitro long-term culture-initiating cell (LTC-IC) assay to quantify multilineage, primitive haematopoietic progenitors in a representative subset of these patients. LTC-IC assays showed equivalent frequencies of cobblestone area-forming cells (CAFCs) with a mean of 5.42/10(5) cells +/- 1.9 SD and 6.13/10(5) cells +/- 2.6 SD in nine patients and six normal controls respectively. The average clonogenic cell output per LTC-IC, however, was significantly lower in DBA patients (mean 2.16 +/- 1.2 SD vs. 7. 36 +/- 2.7 SD in normal controls, P = 0.0008). Our results suggest that the underlying defect in patients with severe refractory DBA may not be limited to the erythroid lineage, as was evidenced by the development of pancytopenia, bone marrow hypoplasia and reduced clonogenic cell output in LTC-IC assays.


Asunto(s)
Anemia de Fanconi/patología , Adolescente , Adulto , Células de la Médula Ósea/patología , Células Cultivadas , Niño , Preescolar , Enfermedad Crónica , Anemia de Fanconi/terapia , Femenino , Hematopoyesis/fisiología , Humanos , Lactante , Recién Nacido , Masculino , Neutropenia/etiología , Neutropenia/patología , Trombocitopenia/etiología , Trombocitopenia/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...