Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 122
Filtrar
1.
Rev Prat ; 63(4): 457-61, 2013 Apr.
Artículo en Francés | MEDLINE | ID: mdl-23682464

RESUMEN

Gene therapy is a form of molecular medicine based on the addition of a corrected copy of a gene to the somatic cells of an individual in order to cure or to alleviate the underlying disease. This approach provides new targeted therapies for an increasing number of acquired or inherited diseases. Since its very first beginning in the 90's essentially for cancer treatment, this therapeutical approach has met unpredicted obstacles as well as true success. Recently, significant clinical benefits have been reported for several inherited diseases of the hematopoietic system but also of the retina and some cancers, widening its application provided that severe adverse events can be efficiently and stably prevented.


Asunto(s)
Terapia Genética , Terapia Genética/métodos , Terapia Genética/tendencias , Vectores Genéticos , Humanos
2.
Gene ; 525(2): 170-3, 2013 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-23583799

RESUMEN

Gene therapy of severe combined immunodeficiencies has been proven to be effective to provide sustained correction of the T cell immunodeficiencies. This has been achieved for 2 forms of SCID, i.e SCID-X1 (γc deficiency) and adenosine deaminase deficiency. Occurrence of gene toxicity generated by integration of first generation retroviral vectors, as observed in the SCID-X1 trials has led to replace these vectors by self inactivated (SIN) retro(or lenti) viruses that may provide equivalent efficacy with a better safety profile. Results of ongoing clinical studies in SCID as well as in other primary immunodeficiencies, such as the Wiskott Aldrich syndrome, will be thus very informative.


Asunto(s)
Terapia Genética/métodos , Inmunodeficiencia Combinada Grave/terapia , Linfocitos T/inmunología , Síndrome de Wiskott-Aldrich/terapia , Adenosina Desaminasa/deficiencia , Agammaglobulinemia/terapia , Ensayos Clínicos como Asunto , Vectores Genéticos/efectos adversos , Humanos , Lentivirus/genética , Retroviridae/genética , Inmunodeficiencia Combinada Grave/genética , Resultado del Tratamiento
3.
Blood ; 121(19): 3981-7, 2013 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-23493783

RESUMEN

We report transplantation outcomes of 258 children with Hurler syndrome (HS) after a myeloablative conditioning regimen from 1995 to 2007. Median age at transplant was 16.7 months and median follow-up was 57 months. The cumulative incidence of neutrophil recovery at day 60 was 91%, acute graft-versus-host disease (GVHD) (grade II-IV) at day 100 was 25%, and chronic GVHD and 5 years was 16%. Overall survival and event-free survival (EFS) at 5 years were 74% and 63%, respectively. EFS after HLA-matched sibling donor (MSD) and 6/6 matched unrelated cord blood (CB) donor were similar at 81%, 66% after 10/10 HLA-matched unrelated donor (UD), and 68% after 5/6 matched CB donor. EFS was lower after transplantation in 4/6 matched unrelated CB (UCB) (57%; P = .031) and HLA-mismatched UD (41%; P = .007). Full-donor chimerism (P = .039) and normal enzyme levels (P = .007) were higher after CB transplantation (92% and 98%, respectively) compared with the other grafts sources (69% and 59%, respectively). In conclusion, results of allogeneic transplantation for HS are encouraging, with similar EFS rates after MSD, 6/6 matched UCB, 5/6 UCB, and 10/10 matched UD. The use of mismatched UD and 4/6 matched UCB was associated with lower EFS.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/citología , Mucopolisacaridosis I/terapia , Agonistas Mieloablativos/uso terapéutico , Acondicionamiento Pretrasplante/métodos , Adolescente , Niño , Preescolar , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Prueba de Histocompatibilidad , Humanos , Lactante , Masculino , Mucopolisacaridosis I/epidemiología , Mucopolisacaridosis I/mortalidad , Agonistas Mieloablativos/efectos adversos , Estudios Retrospectivos , Donantes de Tejidos , Obtención de Tejidos y Órganos/métodos , Acondicionamiento Pretrasplante/efectos adversos , Resultado del Tratamiento , Adulto Joven
4.
Transfusion ; 53(3): 564-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22725259

RESUMEN

BACKGROUND: This report describes the specific kinetics of the peripheral blood (PB) CD34+ cell concentration in a selected group of very poor stem cell mobilizer patients treated with granulocyte-colony-stimulating factor (G-CSF) and plerixafor and determines the kinetics' impact on apheresis. STUDY DESIGN AND METHODS: All patients had previously experienced at least two failures of mobilization (without use of plerixafor). The present salvage therapy consisted in the administration of 10 µg/kg/day G-CSF for 5 days added to a dose of plerixafor administered at between 5 a.m. and 6 a.m. on Day 5. The PB CD34+ cell counts were tested every 3 hours thereafter. Apheresis was initiated as soon as the PB CD34+ cell count reached 10 × 10(6) /L. RESULTS: A PB CD34+ cell count higher than 10 × 10(6) /L was observed as soon as 3 hours after plerixafor administration in 10 of the 11 patients who reached this threshold at some point in the monitoring process. Interestingly, all patients presented an early decrease in the PB CD34+ cell count 8 to 12 hours after plerixafor administration (below 10 × 10(6) /L for seven patients). CONCLUSION: Had such patients been tested for PB CD34+ cell mobilization according to conventional criteria (i.e., 11 hr after plerixafor administration), apheresis would not have been performed at the optimal timing. For very poor stem cell mobilizer patients, early monitoring of PB CD34+ cell count may be required for the optimal initiation of apheresis.


Asunto(s)
Antígenos CD34/metabolismo , Eliminación de Componentes Sanguíneos/métodos , Movilización de Célula Madre Hematopoyética/métodos , Compuestos Heterocíclicos/administración & dosificación , Adulto , Anciano , Antígenos CD34/sangre , Bencilaminas , Recuento de Células Sanguíneas , Células Sanguíneas/metabolismo , Ciclamas , Esquema de Medicación , Sinergismo Farmacológico , Femenino , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos/farmacología , Compuestos Heterocíclicos/farmacología , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/terapia , Trasplante de Células Madre de Sangre Periférica , Factores de Tiempo , Insuficiencia del Tratamiento
5.
Br J Haematol ; 160(2): 146-52, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23167301

RESUMEN

Severe combined immunodeficiencies (SCIDs) correspond to the most severe form of primary immunodeficiency. The extreme severity of the clinical presentation in SCID has legitimately led physicians to consider these conditions as medical emergencies. Hundreds of patients worldwide have undergone allogeneic haematopoietic stem cell transplantation (HCST) in the last 40 years. The complete absence of the T cell compartment in SCID prompted the development (starting in the early 1980s) of haploidentical, parental HSCT for the many patients who do not have a human leucocyte antigen (HLA)-identical sibling. Despite the undeniable progress made in this field over recent years, the long-lasting immunodeficiency that follows partially HLA-incompatible transplantation is still responsible for a mortality rate of 30% at one year post-transplantation. New approaches for reconstituting T cell compartments more rapidly are under intense preclinical development and are discussed herein.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave/cirugía , Trasplante de Células Madre de Sangre del Cordón Umbilical , Citocinas/inmunología , Europa (Continente)/epidemiología , Terapia Genética , Rechazo de Injerto/inmunología , Enfermedad Injerto contra Huésped/epidemiología , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/mortalidad , Histocompatibilidad , Humanos , Lactante , Recién Nacido , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/patología , Subgrupos Linfocitarios/trasplante , Transfusión de Linfocitos , Linfopoyesis , Tamizaje Neonatal , Inmunodeficiencia Combinada Grave/clasificación , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/epidemiología , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Timo/embriología , Timo/inmunología , Timo/patología , Donantes de Tejidos , Trasplante Homólogo , Resultado del Tratamiento , Estados Unidos/epidemiología
6.
Blood ; 121(9): 1510-6, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23264593

RESUMEN

On the basis of a nationwide database of 160 patients with Wiskott-Aldrich syndrome (WAS), we identified a subset of infants who were significantly more likely to be attributed with an Ochs score of 5 before the age of 2 (n = 26 of 47 [55%], P = 2.8 × 10(−7)). A retrospective analysis revealed that these patients often had severe refractory thrombocytopenia (n = 13), autoimmune hemolytic anemia (n = 15), and vasculitis (n = 6). One patient had developed 2 distinct cancers. Hemizygous mutations predictive of the absence of WAS protein were identified in 19 of the 24 tested patients, and the absence of WAS protein was confirmed in all 10 investigated cases. Allogeneic hematopoietic stem cell transplantation (HSCT) was found to be a curative treatment with a relatively good prognosis because it was successful in 17 of 22 patients. Nevertheless, 3 patients experienced significant disease sequelae and 4 patients died before HSCT. Therefore, the present study identifies a distinct subgroup of WAS patients with early-onset, life-threatening manifestations. We suggest that HSCT is a curative strategy in this subgroup of patients and should be performed as early in life as possible, even when a fully matched donor is lacking.


Asunto(s)
Síndrome de Wiskott-Aldrich/diagnóstico , Adolescente , Adulto , Edad de Inicio , Niño , Preescolar , Estudios de Cohortes , Humanos , Lactante , Recién Nacido , Pronóstico , Proyectos de Investigación , Índice de Severidad de la Enfermedad , Síndrome de Wiskott-Aldrich/epidemiología , Síndrome de Wiskott-Aldrich/patología , Adulto Joven
7.
Hum Mutat ; 34(2): 374-84, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23111928

RESUMEN

Ataxia telangiectasia-mutated (ATM) and ataxia telangiectasia and Rad3-related (ATR) kinases are two key regulators of DNA-damage responses (DDR) that are mainly activated in response to DNA double-strand breaks and single-stranded DNA damages, respectively. Seckel syndrome, a rare genetic disorder characterized by a microcephaly and a markedly reduced body size, has been associated with defective ATR-dependent DNA damage signaling. However, the only human genetic ATR defect reported so far is a hypomorphic splicing mutation identified in five related individuals with Seckel syndrome. Here, we report the first case of primary microcephaly with compound heterozygous mutations in ATR: a 540 kb genomic deletion on one allele and a missense mutation leading to splice dysregulation on the other, which ultimately lead to a sharp decrease in ATR expression. DNA combing technology revealed a profound spontaneous alteration of several DNA replication parameters in patient's cells and FISH analyses highlighted the genomic instability caused by ATR deficiency. Collectively, our results emphasize the crucial role for ATR in the control of DNA replication, and reinforce the complementary and nonredundant contributions of ATM and ATR in human cells to face DNA damages and warrant genome integrity.


Asunto(s)
Proteínas de Ciclo Celular/genética , Replicación del ADN , ADN de Cadena Simple , Proteínas de Unión al ADN/genética , Inestabilidad Genómica , Microcefalia/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Supresoras de Tumor/genética , Alelos , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Secuencia de Bases , Western Blotting , Línea Celular , Niño , Hibridación Genómica Comparativa , Roturas del ADN de Doble Cadena , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Eliminación de Gen , Estudio de Asociación del Genoma Completo , Heterocigoto , Humanos , Hibridación Fluorescente in Situ , Masculino , Ratones , Datos de Secuencia Molecular , Mutación Missense , Empalme del ARN , Transducción de Señal
8.
Curr Opin Immunol ; 24(5): 580-4, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22981681

RESUMEN

Over 60 patients affected by SCID due to IL2RG deficiency (SCID-X1) or adenosine deaminase (ADA)-SCID have received hematopoietic stem cell gene therapy in the past 15 years using gammaretroviral vectors, resulting in immune reconstitution and clinical benefit in the majority of them. However, the occurrence of insertional oncogenesis in the SCID-X1 trials has led to the development of new clinical trials based on integrating vectors with improved safety design as well as investigation on new technologies for highly efficient gene targeting and site-specific gene editing. Here we will present the experience and perspectives of gene therapy for SCID-X1 and ADA-SCID and discuss the pros and cons of gene therapy in comparison to allogeneic transplantation.


Asunto(s)
Terapia Genética/métodos , Terapia Genética/tendencias , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/terapia , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Animales , Ensayos Clínicos como Asunto/métodos , Ensayos Clínicos como Asunto/tendencias , Modelos Animales de Enfermedad , Trasplante de Células Madre Hematopoyéticas/métodos , Trasplante de Células Madre Hematopoyéticas/tendencias , Humanos , Síndromes de Inmunodeficiencia/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia
10.
Curr Opin Immunol ; 24(5): 585-91, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22909900

RESUMEN

Gene therapy has become an attractive alternative therapeutic strategy to allogeneic transplant for primary immunodeficiencies (PIDs) owing to known genetic defects. Clinical trials using gammaretroviral vectors have demonstrated the proof of principle of gene therapy for Wiskott-Aldrich syndrome (WAS) and chronic granulomatous disease (CGD), but have also highlighted limitations of the technology. New strategies based on vectors that can achieve more robust correction with less risk of insertional mutagenesis are being developed. In this review we present the status of gene therapy for WAS and CGD, and discuss the emerging application of similar strategies to a broader range of PIDs, such as IPEX syndrome.


Asunto(s)
Terapia Genética/métodos , Terapia Genética/tendencias , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/terapia , Animales , Ensayos Clínicos como Asunto/tendencias , Síndrome de Exfoliación/genética , Síndrome de Exfoliación/inmunología , Síndrome de Exfoliación/terapia , Predicción/métodos , Técnicas de Transferencia de Gen/tendencias , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Vectores Genéticos/uso terapéutico , Enfermedad Granulomatosa Crónica/genética , Enfermedad Granulomatosa Crónica/inmunología , Enfermedad Granulomatosa Crónica/terapia , Humanos , Síndromes de Inmunodeficiencia/inmunología , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/terapia , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/inmunología , Síndrome de Wiskott-Aldrich/terapia
11.
Blood ; 120(17): 3615-24; quiz 3626, 2012 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-22791287

RESUMEN

Deficiency of the purine salvage enzyme adenosine deaminase leads to SCID (ADA-SCID). Hematopoietic cell transplantation (HCT) can lead to a permanent cure of SCID; however, little data are available on outcome of HCT for ADA-SCID in particular. In this multicenter retrospective study, we analyzed outcome of HCT in 106 patients with ADA-SCID who received a total of 119 transplants. HCT from matched sibling and family donors (MSDs, MFDs) had significantly better overall survival (86% and 81%) in comparison with HCT from matched unrelated (66%; P < .05) and haploidentical donors (43%; P < .001). Superior overall survival was also seen in patients who received unconditioned transplants in comparison with myeloablative procedures (81% vs 54%; P < .003), although in unconditioned haploidentical donor HCT, nonengraftment was a major problem. Long-term immune recovery showed that regardless of transplant type, overall T-cell numbers were similar, although a faster rate of T-cell recovery was observed after MSD/MFD HCT. Humoral immunity and donor B-cell engraftment was achieved in nearly all evaluable surviving patients and was seen even after unconditioned HCT. These data detail for the first time the outcomes of HCT for ADA-SCID and show that, if patients survive HCT, long-term cellular and humoral immune recovery is achieved.


Asunto(s)
Agammaglobulinemia/tratamiento farmacológico , Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave/tratamiento farmacológico , Acondicionamiento Pretrasplante , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/inmunología , Agammaglobulinemia/inmunología , Agammaglobulinemia/mortalidad , Agammaglobulinemia/patología , Niño , Preescolar , Femenino , Supervivencia de Injerto , Prueba de Histocompatibilidad , Humanos , Inmunidad Celular , Inmunidad Humoral , Lactante , Recién Nacido , Estimación de Kaplan-Meier , Recuento de Linfocitos , Masculino , Agonistas Mieloablativos/uso terapéutico , Estudios Retrospectivos , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/mortalidad , Inmunodeficiencia Combinada Grave/patología , Hermanos , Linfocitos T/inmunología , Resultado del Tratamiento , Donante no Emparentado
12.
Stem Cells ; 30(8): 1771-80, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22689616

RESUMEN

Slow T-cell reconstitution is a major clinical concern after transplantation of cord blood (CB)-derived hematopoietic stem cells. Adoptive transfer of in vitro-generated T-cell progenitors has emerged as a promising strategy for promoting de novo thymopoiesis and thus accelerating T-cell reconstitution. Here, we describe the development of a new culture system based on the immobilized Notch ligand Delta-like-4 (DL-4). Culture of human CD34(+) CB cells in this new DL-4 system enabled the in vitro generation of large amounts of T-cell progenitor cells that (a) displayed the phenotypic and molecular signatures of early thymic progenitors and (b) had high T lymphopoietic potential. When transferred into NOD/SCID/γc(-/-) (NSG) mice, DL-4 primed T-cell progenitors migrated to the thymus and developed into functional, mature, polyclonal αß T cells that subsequently left the thymus and accelerated T-cell reconstitution. T-cell reconstitution was even faster and more robust when ex vivo-manipulated and nonmanipulated CB samples were simultaneously injected into NSG mice (i.e., a situation reminiscent of the double CB transplant setting). This work provides further evidence of the ability of in vitro-generated human T-cell progenitors to accelerate T-cell reconstitution and also introduces a feeder-cell-free culture technique with the potential for rapid, safe transfer to a clinical setting.


Asunto(s)
Células Madre Hematopoyéticas/citología , Péptidos y Proteínas de Señalización Intercelular/inmunología , Linfocitos T/citología , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de Unión al Calcio , Diferenciación Celular/fisiología , Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/inmunología , Humanos , Inmunoterapia , Péptidos y Proteínas de Señalización Intercelular/genética , Linfopoyesis/fisiología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T/inmunología
13.
Blood ; 120(10): 2144-54, 2012 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-22730537

RESUMEN

Invariant natural killer T (iNKT) cells can experimentally dissociate GVL from graft-versus-host-disease (GVHD). Their role in human conventional allogeneic hematopoietic stem cell transplantation (HSCT) is unknown. Here, we analyzed the post-HSCT recovery of iNKT cells in 71 adult allografted patients. Results were compared with conventional T- and NK-cell recovery and correlated to the occurrence of GVHD, relapse, and survival. We observed that posttransplantation iNKT cells, likely of donor origin, recovered independently of T and NK cells in the first 90 days after HSCT and reached greater levels in recipient younger than 45 years (P = .003) and after a reduced-intensity conditioning regimen (P = .03). Low posttransplantation iNKT/T ratios (ie, < 10(-3)) were an independent factor associated with the occurrence of acute GVHD (aGVHD; P = .001). Inversely, reaching iNKT/T ratios > 10(-3) before day 90 was associated with reduced nonrelapse mortality (P = .009) without increased risk of relapse and appeared as an independent predictive factor of an improved overall survival (P = .028). Furthermore, an iNKT/T ratio on day 15 > 0.58 × 10(-3) was associated with a 94% risk reduction of aGVHD. These findings provide a proof of concept that early postallogeneic HSCT iNKT cell recovery can predict the occurrence of aGVHD and an improved overall survival.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre Hematopoyéticas , Células T Asesinas Naturales/inmunología , Acondicionamiento Pretrasplante/métodos , Enfermedad Aguda , Adolescente , Adulto , Factores de Edad , Femenino , Enfermedad Injerto contra Huésped/patología , Enfermedad Injerto contra Huésped/terapia , Efecto Injerto vs Leucemia/inmunología , Neoplasias Hematológicas/inmunología , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/terapia , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Células T Asesinas Naturales/patología , Recurrencia , Factores de Riesgo , Análisis de Supervivencia , Linfocitos T/inmunología , Linfocitos T/patología , Factores de Tiempo , Donantes de Tejidos , Trasplante Homólogo
14.
Cell Stem Cell ; 11(1): 36-49, 2012 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-22683204

RESUMEN

Fanconi anemia (FA) is an inherited DNA repair deficiency syndrome. FA patients undergo progressive bone marrow failure (BMF) during childhood, which frequently requires allogeneic hematopoietic stem cell transplantation. The pathogenesis of this BMF has been elusive to date. Here we found that FA patients exhibit a profound defect in hematopoietic stem and progenitor cells (HSPCs) that is present before the onset of clinical BMF. In response to replicative stress and unresolved DNA damage, p53 is hyperactivated in FA cells and triggers a late p21(Cdkn1a)-dependent G0/G1 cell-cycle arrest. Knockdown of p53 rescued the HSPC defects observed in several in vitro and in vivo models, including human FA or FA-like cells. Taken together, our results identify an exacerbated p53/p21 "physiological" response to cellular stress and DNA damage accumulation as a central mechanism for progressive HSPC elimination in FA patients, and have implications for clinical care.


Asunto(s)
Médula Ósea/patología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patología , Células Madre Hematopoyéticas/patología , Proteína p53 Supresora de Tumor/metabolismo , Adolescente , Adulto , Células Madre Adultas/metabolismo , Células Madre Adultas/patología , Envejecimiento/patología , Animales , Médula Ósea/metabolismo , Niño , Preescolar , Modelos Animales de Enfermedad , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/patología , Proteína del Grupo de Complementación C de la Anemia de Fanconi/deficiencia , Proteína del Grupo de Complementación C de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular , Puntos de Control de la Fase G2 del Ciclo Celular , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Células Madre Hematopoyéticas/metabolismo , Humanos , Lactante , Ratones , Persona de Mediana Edad , Fase S
15.
Stem Cells ; 30(8): 1675-84, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22644669

RESUMEN

Mutations in the survival of motor neuron gene (SMN1) are responsible for spinal muscular atrophy, a fatal neuromuscular disorder. Mice carrying a homozygous deletion of Smn exon 7 directed to skeletal muscle (HSA-Cre, Smn(F7/F7) mice) present clinical features of human muscular dystrophies for which new therapeutic approaches are highly warranted. Herein we demonstrate that tail vein transplantation of mouse amniotic fluid stem (AFS) cells enhances the muscle strength and improves the survival rate of the affected animals. Second, after cardiotoxin injury of the Tibialis Anterior, only AFS-transplanted mice efficiently regenerate. Most importantly, secondary transplants of satellite cells (SCs) derived from treated mice show that AFS cells integrate into the muscle stem cell compartment and have long-term muscle regeneration capacity indistinguishable from that of wild-type-derived SC. This is the first study demonstrating the functional and stable integration of AFS cells into the skeletal muscle, highlighting their value as cell source for the treatment of muscular dystrophies.


Asunto(s)
Líquido Amniótico/citología , Músculo Esquelético/citología , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/cirugía , Nicho de Células Madre/fisiología , Trasplante de Células Madre/métodos , Células Madre/citología , Líquido Amniótico/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/fisiopatología , Atrofia Muscular Espinal/patología , Atrofia Muscular Espinal/fisiopatología , Distribución Aleatoria , Células Madre/metabolismo , Proteína 1 para la Supervivencia de la Neurona Motora/genética
16.
Blood ; 119(12): 2949-55, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22308292

RESUMEN

Pediatric patients with SCID constitute medical emergencies. In the absence of an HLA-identical hematopoietic stem cell (HSC) donor, mismatched related-donor transplantation (MMRDT) or unrelated-donor umbilical cord blood transplantation (UCBT) are valuable treatment options. To help transplantation centers choose the best treatment option, we retrospectively compared outcomes after 175 MMRDTs and 74 UCBTs in patients with SCID or Omenn syndrome. Median follow-up time was 83 months and 58 months for UCBT and MMRDT, respectively. Most UCB recipients received a myeloablative conditioning regimen; most MMRDT recipients did not. UCB recipients presented a higher frequency of complete donor chimerism (P = .04) and faster total lymphocyte count recovery (P = .04) without any statistically significance with the preparative regimen they received. The MMRDT and UCBT groups did not differ in terms of T-cell engraftment, CD4(+) and CD3(+) cell recoveries, while Ig replacement therapy was discontinued sooner after UCBT (adjusted P = .02). There was a trend toward a greater incidence of grades II-IV acute GVHD (P = .06) and more chronic GVHD (P = .03) after UCBT. The estimated 5-year overall survival rates were 62% ± 4% after MMRDT and 57% ± 6% after UCBT. For children with SCID and no HLA-identical sibling donor, both UCBT and MMRDT represent available HSC sources for transplantation with quite similar outcomes.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical , Enfermedad Injerto contra Huésped/epidemiología , Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/cirugía , Preescolar , Trasplante de Células Madre de Sangre del Cordón Umbilical/efectos adversos , Trasplante de Células Madre de Sangre del Cordón Umbilical/mortalidad , Femenino , Enfermedad Injerto contra Huésped/mortalidad , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/mortalidad , Histocompatibilidad , Humanos , Incidencia , Lactante , Recién Nacido , Estimación de Kaplan-Meier , Masculino , Modelos de Riesgos Proporcionales , Estudios Retrospectivos , Inmunodeficiencia Combinada Grave/mortalidad , Acondicionamiento Pretrasplante/métodos , Resultado del Tratamiento
17.
Methods Enzymol ; 507: 15-27, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22365767

RESUMEN

Severe combined immunodeficiencies (SCIDs) appear as optimal disease targets to challenge potential efficacy of gene therapy. Ex vivo, retrovirally mediated gene transfer into hematopoietic progenitor cells has been shown to provide sustained correction of two forms of SCID, that is, SCID-X1 and adenosine deaminase deficiencies. In the former case, however, genotoxicity was observed in a minority of patients as a consequence of retroviral integration into proto-oncogenes loci and transactivation. Design of vectors in which the enhancer element of retroviral LTR has been deleted and an internal promoter added (self-inactivated vectors) could provide both safe and efficient gene transfer as being presently tested.


Asunto(s)
Terapia Genética , Retroviridae/genética , Inmunodeficiencia Combinada Grave/terapia , Adenosina Desaminasa/deficiencia , Agammaglobulinemia/terapia , Ensayos Clínicos como Asunto , Técnicas de Transferencia de Gen , Ingeniería Genética , Vectores Genéticos , Humanos , Resultado del Tratamiento , Integración Viral
18.
Methods Enzymol ; 507: 187-98, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22365775

RESUMEN

X-linked adrenoleukodystrophy (X-ALD) is a severe genetic demyelinating disease caused by a deficiency in ALD protein, an adenosine triphosphate-binding cassette transporter encoded by the ABCD1 gene. When performed at an early stage of the disease, allogeneic hematopoietic stem cell transplantation (HCT) can arrest the progression of cerebral demyelinating lesions. To overcome the limitations of allogeneic HCT, hematopoietic stem cell (HSC) gene therapy strategy aiming to perform autologous transplantation of lentivirally corrected cells was developed. We demonstrated the preclinical feasibility of HSC gene therapy for ALD based on the correction of CD34+ cells from X-ALD patients using an HIV1-derived lentiviral vector. These results prompted us to initiate an HSC gene therapy trial in two X-ALD patients who had developed progressive cerebral demyelination, were candidates for allogeneic HCT, but had no HLA-matched donors or cord blood. Autologous CD34+ cells were purified from the peripheral blood after G-CSF stimulation, genetically corrected ex vivo with a lentiviral vector encoding wild-type ABCD1 cDNA, and then reinfused into the patients after they had received full myeloablative conditioning. Over 3 years of follow-up, the hematopoiesis remained polyclonal in the two patients treated with 7-14% of granulocytes, monocytes, and T and B lymphocytes expressing the lentivirally encoded ALD protein. There was no evidence of clonal dominance or skewing based on the retrieval of lentiviral insertion repertoire in different hematopoietic lineages by deep sequencing. Cerebral demyelination was arrested 14 and 16months, respectively, in the two treated patients, without further progression up to the last follow-up, a clinical outcome that is comparable to that observed after allogeneic HCT. Longer follow-up of these two treated patients and HSC gene therapy performed in additional ALD patients are however needed to evaluate the safety and efficacy of lentiviral HSC gene therapy in cerebral forms of X-ALD.


Asunto(s)
Adrenoleucodistrofia/terapia , Terapia Genética , Lentivirus/genética , Miembro 1 de la Subfamilia D de Transportador de Casetes de Unión al ATP , Transportadoras de Casetes de Unión a ATP/genética , Niño , Técnicas de Transferencia de Gen , Vectores Genéticos , Células HEK293 , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/virología , Humanos , Masculino , Resultado del Tratamiento
19.
Mol Ther ; 19(11): 2031-9, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21862999

RESUMEN

Vector-associated side effects in clinical gene therapy have provided insights into the molecular mechanisms of hematopoietic regulation in vivo. Surprisingly, many retrovirus insertion sites (RIS) present in engrafted cells have been found to cluster nonrandomly in close association with specific genes. Our data demonstrate that these genes directly influence the in vivo fate of hematopoietic cell clones. Analysis of insertions thus far has been limited to individual clinical studies. Here, we studied >7,000 insertions retrieved from various studies. More than 40% of all insertions found in engrafted gene-modified cells were clustered in the same genomic areas covering only 0.36% of the genome. Gene classification analyses displayed significant overrepresentation of genes associated with hematopoietic functions and relevance for cell growth and survival in vivo. The similarity of insertion distributions indicates that vector insertions in repopulating cells cluster in predictable patterns. Thus, insertion analyses of preclinical in vitro and murine in vivo studies as well as vector insertion repertoires in clinical trials yielded concerted results and mark a small number of interesting genomic loci and genes that warrants further investigation of the biological consequences of vector insertions.


Asunto(s)
Gammaretrovirus/genética , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Genoma , Integración Viral , Animales , Mapeo Cromosómico , Redes Reguladoras de Genes , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Primates , Trasplantes , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia
20.
J Allergy Clin Immunol ; 127(6): 1356-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21624615

RESUMEN

Gene therapy has become an option for the treatment of 2 forms of severe combined immunodeficiency (SCID): X-linked SCID and adenosine deaminase deficiency. The results of clinical trials initiated more than 10 years ago testify to sustained and reproducible correction of the underlying T-cell immunodeficiency. Successful treatment is based on the selective advantage conferred on T-cell precursors through their expression of the therapeutic transgene. However, "first-generation" retroviral vectors also caused leukemia in some patients with X-linked SCID because of the constructs' tendency to insert into active genes (eg, proto-oncogenes) in progenitor cells and transactivate an oncogene through a viral element in the long terminal repeat. These elements have been deleted from the vectors now in use. Together with the use of lentiviral vectors (which are more potent for transducing stem cells), these advances should provide a basis for the safe and effective extension of gene therapy's indications in the field of primary immunodeficiencies. Nevertheless, this extension will have to be proved by examining the results of the ongoing clinical trials.


Asunto(s)
Terapia Genética/métodos , Síndromes de Inmunodeficiencia/terapia , Inmunidad Adaptativa , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Adenosina Desaminasa/inmunología , Agammaglobulinemia/genética , Agammaglobulinemia/inmunología , Agammaglobulinemia/terapia , Ensayos Clínicos como Asunto , Terapia Genética/efectos adversos , Terapia Genética/tendencias , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Humanos , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/inmunología , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/terapia , Resultado del Tratamiento , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/inmunología , Síndrome de Wiskott-Aldrich/terapia , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA