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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
N Engl J Med ; 384(21): 2002-2013, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-33974366

RESUMEN

BACKGROUND: Severe combined immunodeficiency due to adenosine deaminase (ADA) deficiency (ADA-SCID) is a rare and life-threatening primary immunodeficiency. METHODS: We treated 50 patients with ADA-SCID (30 in the United States and 20 in the United Kingdom) with an investigational gene therapy composed of autologous CD34+ hematopoietic stem and progenitor cells (HSPCs) transduced ex vivo with a self-inactivating lentiviral vector encoding human ADA. Data from the two U.S. studies (in which fresh and cryopreserved formulations were used) at 24 months of follow-up were analyzed alongside data from the U.K. study (in which a fresh formulation was used) at 36 months of follow-up. RESULTS: Overall survival was 100% in all studies up to 24 and 36 months. Event-free survival (in the absence of reinitiation of enzyme-replacement therapy or rescue allogeneic hematopoietic stem-cell transplantation) was 97% (U.S. studies) and 100% (U.K. study) at 12 months; 97% and 95%, respectively, at 24 months; and 95% (U.K. study) at 36 months. Engraftment of genetically modified HSPCs persisted in 29 of 30 patients in the U.S. studies and in 19 of 20 patients in the U.K. study. Patients had sustained metabolic detoxification and normalization of ADA activity levels. Immune reconstitution was robust, with 90% of the patients in the U.S. studies and 100% of those in the U.K. study discontinuing immunoglobulin-replacement therapy by 24 months and 36 months, respectively. No evidence of monoclonal expansion, leukoproliferative complications, or emergence of replication-competent lentivirus was noted, and no events of autoimmunity or graft-versus-host disease occurred. Most adverse events were of low grade. CONCLUSIONS: Treatment of ADA-SCID with ex vivo lentiviral HSPC gene therapy resulted in high overall and event-free survival with sustained ADA expression, metabolic correction, and functional immune reconstitution. (Funded by the National Institutes of Health and others; ClinicalTrials.gov numbers, NCT01852071, NCT02999984, and NCT01380990.).


Asunto(s)
Agammaglobulinemia/terapia , Terapia Genética/métodos , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Lentivirus/genética , Inmunodeficiencia Combinada Grave/terapia , Adenosina Desaminasa/deficiencia , Adolescente , Niño , Preescolar , Terapia Genética/efectos adversos , Humanos , Lactante , Recuento de Linfocitos , Supervivencia sin Progresión , Estudios Prospectivos , Trasplante Autólogo
2.
Blood ; 130(11): 1327-1335, 2017 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-28716862

RESUMEN

Until recently, hematopoietic stem cell transplantation was the only curative option for Wiskott-Aldrich syndrome (WAS). The first attempts at gene therapy for WAS using a ϒ-retroviral vector improved immunological parameters substantially but were complicated by acute leukemia as a result of insertional mutagenesis in a high proportion of patients. More recently, treatment of children with a state-of-the-art self-inactivating lentiviral vector (LV-w1.6 WASp) has resulted in significant clinical benefit without inducing selection of clones harboring integrations near oncogenes. Here, we describe a case of a presplenectomized 30-year-old patient with severe WAS manifesting as cutaneous vasculitis, inflammatory arthropathy, intermittent polyclonal lymphoproliferation, and significant chronic kidney disease and requiring long-term immunosuppressive treatment. Following reduced-intensity conditioning, there was rapid engraftment and expansion of a polyclonal pool of transgene-positive functional T cells and sustained gene marking in myeloid and B-cell lineages up to 20 months of observation. The patient was able to discontinue immunosuppression and exogenous immunoglobulin support, with improvement in vasculitic disease and proinflammatory markers. Autologous gene therapy using a lentiviral vector is a viable strategy for adult WAS patients with severe chronic disease complications and for whom an allogeneic procedure could present an unacceptable risk. This trial was registered at www.clinicaltrials.gov as #NCT01347242.


Asunto(s)
Terapia Genética , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/terapia , Adulto , Proliferación Celular , Preescolar , Ensayos Clínicos como Asunto , Células Clonales , Citocinas/sangre , Humanos , Subgrupos Linfocitarios/inmunología , Linfocitos T/inmunología , Vacunación , Síndrome de Wiskott-Aldrich/sangre
3.
N Engl J Med ; 371(15): 1407-17, 2014 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-25295500

RESUMEN

BACKGROUND: In previous clinical trials involving children with X-linked severe combined immunodeficiency (SCID-X1), a Moloney murine leukemia virus-based γ-retrovirus vector expressing interleukin-2 receptor γ-chain (γc) complementary DNA successfully restored immunity in most patients but resulted in vector-induced leukemia through enhancer-mediated mutagenesis in 25% of patients. We assessed the efficacy and safety of a self-inactivating retrovirus for the treatment of SCID-X1. METHODS: We enrolled nine boys with SCID-X1 in parallel trials in Europe and the United States to evaluate treatment with a self-inactivating (SIN) γ-retrovirus vector containing deletions in viral enhancer sequences expressing γc (SIN-γc). RESULTS: All patients received bone marrow-derived CD34+ cells transduced with the SIN-γc vector, without preparative conditioning. After 12.1 to 38.7 months of follow-up, eight of the nine children were still alive. One patient died from an overwhelming adenoviral infection before reconstitution with genetically modified T cells. Of the remaining eight patients, seven had recovery of peripheral-blood T cells that were functional and led to resolution of infections. The patients remained healthy thereafter. The kinetics of CD3+ T-cell recovery was not significantly different from that observed in previous trials. Assessment of insertion sites in peripheral blood from patients in the current trial as compared with those in previous trials revealed significantly less clustering of insertion sites within LMO2, MECOM, and other lymphoid proto-oncogenes in our patients. CONCLUSIONS: This modified γ-retrovirus vector was found to retain efficacy in the treatment of SCID-X1. The long-term effect of this therapy on leukemogenesis remains unknown. (Funded by the National Institutes of Health and others; ClinicalTrials.gov numbers, NCT01410019, NCT01175239, and NCT01129544.).


Asunto(s)
Gammaretrovirus/genética , Terapia Genética , Vectores Genéticos , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Animales , Antígenos CD34 , ADN Complementario/uso terapéutico , Expresión Génica , Silenciador del Gen , Terapia Genética/efectos adversos , Humanos , Lactante , Subunidad gamma Común de Receptores de Interleucina/genética , Masculino , Ratones , Mutación , Linfocitos T/inmunología , Transducción Genética , Transgenes/fisiología , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/inmunología
4.
Blood ; 121(7): 1073-6, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23223356

RESUMEN

X-linked lymphoproliferative disease (XLP1) arises from mutations in the gene encoding SLAM-associated protein (SAP) and leads to abnormalities of NKT-cell development, NK-cell cytotoxicity, and T-dependent humoral function. Curative treatment is limited to allogeneic hematopoietic stem cell (HSC) transplantation. We tested whether HSC gene therapy could correct the multilineage defects seen in SAP(-/-) mice. SAP(-/-) murine HSCs were transduced with lentiviral vectors containing either SAP or reporter gene before transplantation into irradiated recipients. NKT-cell development was significantly higher and NK-cell cytotoxicity restored to wild-type levels in mice receiving the SAP vector in comparison to control mice. Baseline immunoglobulin levels were significantly increased and T-dependent humoral responses to NP-CGG, including germinal center formation, were restored in SAP-transduced mice.We demonstrate for the first time that HSC gene transfer corrects the cellular and humoral defects in SAP(-/-) mice providing proof of concept for gene therapy in XLP1.


Asunto(s)
Terapia Genética/métodos , Péptidos y Proteínas de Señalización Intracelular/genética , Trastornos Linfoproliferativos/inmunología , Trastornos Linfoproliferativos/terapia , Animales , Linaje de la Célula , Modelos Animales de Enfermedad , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Trasplante de Células Madre Hematopoyéticas , Inmunidad Celular , Inmunidad Humoral , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Asesinas Naturales/inmunología , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/patología , Ratones , Ratones Noqueados , Células T Asesinas Naturales/inmunología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Proteína Asociada a la Molécula de Señalización de la Activación Linfocitaria
5.
JAMA ; 313(15): 1550-63, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25898053

RESUMEN

IMPORTANCE: Wiskott-Aldrich syndrome is a rare primary immunodeficiency associated with severe microthrombocytopenia. Partially HLA antigen-matched allogeneic hematopoietic stem cell (HSC) transplantation is often curative but is associated with significant comorbidity. OBJECTIVE: To assess the outcomes and safety of autologous HSC gene therapy in Wiskott-Aldrich syndrome. DESIGN, SETTING, AND PARTICIPANTS: Gene-corrected autologous HSCs were infused in 7 consecutive patients with severe Wiskott-Aldrich syndrome lacking HLA antigen-matched related or unrelated HSC donors (age range, 0.8-15.5 years; mean, 7 years) following myeloablative conditioning. Patients were enrolled in France and England and treated between December 2010 and January 2014. Follow-up of patients in this intermediate analysis ranged from 9 to 42 months. INTERVENTION: A single infusion of gene-modified CD34+ cells with an advanced lentiviral vector. MAIN OUTCOMES AND MEASURES: Primary outcomes were improvement at 24 months in eczema, frequency and severity of infections, bleeding tendency, and autoimmunity and reduction in disease-related days of hospitalization. Secondary outcomes were improvement in immunological and hematological characteristics and evidence of safety through vector integration analysis. RESULTS: Six of the 7 patients were alive at the time of last follow-up (mean and median follow-up, 28 months and 27 months, respectively) and showed sustained clinical benefit. One patient died 7 months after treatment of preexisting drug-resistant herpes virus infection. Eczema and susceptibility to infections resolved in all 6 patients. Autoimmunity improved in 5 of 5 patients. No severe bleeding episodes were recorded after treatment, and at last follow-up, all 6 surviving patients were free of blood product support and thrombopoietic agonists. Hospitalization days were reduced from a median of 25 days during the 2 years before treatment to a median of 0 days during the 2 years after treatment. All 6 surviving patients exhibited high-level, stable engraftment of functionally corrected lymphoid cells. The degree of myeloid cell engraftment and of platelet reconstitution correlated with the dose of gene-corrected cells administered. No evidence of vector-related toxicity was observed clinically or by molecular analysis. CONCLUSIONS AND RELEVANCE: This study demonstrated the feasibility of the use of gene therapy in patients with Wiskott-Aldrich syndrome. Controlled trials with larger numbers of patients are necessary to assess long-term outcomes and safety.


Asunto(s)
Terapia Genética , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Lentivirus , Familia de Proteínas del Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/terapia , Adolescente , Niño , Preescolar , Estudios de Factibilidad , Expresión Génica , Terapia Genética/efectos adversos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Lactante , Recién Nacido , Masculino , Índice de Severidad de la Enfermedad , Síndrome de Wiskott-Aldrich/genética , Síndrome de Wiskott-Aldrich/inmunología
6.
Nat Cancer ; 2(6): 629-642, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34345830

RESUMEN

Low-affinity CD19 chimeric antigen receptor (CAR) T cells display enhanced expansion and persistence, enabling fate tracking through integration site analysis. Here we show that integration sites from early (1 month) and late (>3yr) timepoints cluster separately, suggesting different clonal contribution to early responses and prolonged anti-leukemic surveillance. CAR T central and effector memory cells in patients with long-term persistence remained highly polyclonal, whereas diversity dropped rapidly in patients with limited CAR T persistence. Analysis of shared integrants between the CAR T cell product and post-infusion demonstrated that, despite their low frequency, T memory stem cell clones in the product contributed substantially to the circulating CAR T cell pools, during both early expansion and long-term persistence. Our data may help identify patients at risk of early loss of CAR T cells and highlight the critical role of T memory stem cells both in mediating early anti-leukemic responses and in long-term surveillance by CAR T cells.


Asunto(s)
Receptores Quiméricos de Antígenos , Antígenos CD19 , Humanos , Inmunoterapia Adoptiva/efectos adversos , Receptores de Antígenos de Linfocitos T/genética , Receptores Quiméricos de Antígenos/genética , Células Madre
7.
Nat Commun ; 12(1): 1622, 2021 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-33712608

RESUMEN

Our mathematical model of integration site data in clinical gene therapy supported the existence of long-term lymphoid progenitors capable of surviving independently from hematopoietic stem cells. To date, no experimental setting has been available to validate this prediction. We here report evidence of a population of lymphoid progenitors capable of independently maintaining T and NK cell production for 15 years in humans. The gene therapy patients of this study lack vector-positive myeloid/B cells indicating absence of engineered stem cells but retain gene marking in both T and NK. Decades after treatment, we can still detect and analyse transduced naïve T cells whose production is likely maintained by a population of long-term lymphoid progenitors. By tracking insertional clonal markers overtime, we suggest that these progenitors can support both T and NK cell production. Identification of these long-term lymphoid progenitors could be utilised for the development of next generation gene- and cancer-immunotherapies.


Asunto(s)
Células Asesinas Naturales/fisiología , Linfocitos/fisiología , Células Progenitoras Linfoides/fisiología , Linfocitos T/fisiología , Linfocitos B , Terapia Genética/métodos , Células Madre Hematopoyéticas , Humanos , Interferón gamma/metabolismo , Mutagénesis , Células Mieloides/fisiología , Proto-Oncogenes/genética , Proto-Oncogenes/fisiología
8.
Hum Gene Ther ; 32(17-18): 949-958, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33740872

RESUMEN

Chronic granulomatous disease (CGD) is an inherited blood disorder of phagocytic cells that renders patients susceptible to infections and inflammation. A recent clinical trial of lentiviral gene therapy for the most frequent form of CGD, X-linked, has demonstrated stable correction over time, with no adverse events related to the gene therapy procedure. We have recently developed a parallel lentiviral vector for p47phox-deficient CGD (p47phoxCGD), the second most common form of this disease. Using this vector, we have observed biochemical correction of CGD in a mouse model of the disease. In preparation for clinical trial approval, we have performed standardized preclinical studies following Good Laboratory Practice (GLP) principles, to assess the safety of the gene therapy procedure. We report no evidence of adverse events, including mutagenesis and tumorigenesis, in human hematopoietic stem cells transduced with the lentiviral vector. Biodistribution studies of transduced human CD34+ cells indicate that the homing properties or engraftment ability of the stem cells is not negatively affected. CD34+ cells derived from a p47phoxCGD patient were subjected to an optimized transduction protocol and transplanted into immunocompromised mice. After the procedure, patient-derived neutrophils resumed their function, suggesting that gene correction was successful. These studies pave the way to a first-in-man clinical trial of lentiviral gene therapy for the treatment of p47phoxCGD.


Asunto(s)
Enfermedad Granulomatosa Crónica , Animales , Humanos , Ratones , Terapia Genética , Enfermedad Granulomatosa Crónica/genética , Enfermedad Granulomatosa Crónica/terapia , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Distribución Tisular
9.
Nat Med ; 26(2): 200-206, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31988463

RESUMEN

Chronic granulomatous disease (CGD) is a rare inherited disorder of phagocytic cells1,2. We report the initial results of nine severely affected X-linked CGD (X-CGD) patients who received ex vivo autologous CD34+ hematopoietic stem and progenitor cell-based lentiviral gene therapy following myeloablative conditioning in first-in-human studies (trial registry nos. NCT02234934 and NCT01855685). The primary objectives were to assess the safety and evaluate the efficacy and stability of biochemical and functional reconstitution in the progeny of engrafted cells at 12 months. The secondary objectives included the evaluation of augmented immunity against bacterial and fungal infection, as well as assessment of hematopoietic stem cell transduction and engraftment. Two enrolled patients died within 3 months of treatment from pre-existing comorbidities. At 12 months, six of the seven surviving patients demonstrated stable vector copy numbers (0.4-1.8 copies per neutrophil) and the persistence of 16-46% oxidase-positive neutrophils. There was no molecular evidence of either clonal dysregulation or transgene silencing. Surviving patients have had no new CGD-related infections, and six have been able to discontinue CGD-related antibiotic prophylaxis. The primary objective was met in six of the nine patients at 12 months follow-up, suggesting that autologous gene therapy is a promising approach for CGD patients.


Asunto(s)
Cromosomas Humanos X , Terapia Genética/métodos , Enfermedad Granulomatosa Crónica/genética , Lentivirus/genética , Adolescente , Antígenos CD34/genética , Niño , Preescolar , Comorbilidad , Silenciador del Gen , Genes Reguladores , Vectores Genéticos , Enfermedad Granulomatosa Crónica/terapia , Células Madre Hematopoyéticas/citología , Humanos , Masculino , NADPH Oxidasas/genética , Neutrófilos/metabolismo , Seguridad del Paciente , Regiones Promotoras Genéticas , Acondicionamiento Pretrasplante , Resultado del Tratamiento , Reino Unido , Estados Unidos , Adulto Joven
10.
Cancer Res ; 65(1): 195-202, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15665295

RESUMEN

Signal transducer and activator of transcription (STAT) 3 is overexpressed or activated in most types of human tumors and has been classified as an oncogene. In the present study, we investigated the contribution of the STAT3s to the proinvasive activity of trefoil factors (TFF) and vascular endothelial growth factor (VEGF) in human colorectal cancer cells HCT8/S11 expressing VEGF receptors. Both intestinal trefoil peptide (TFF3) and VEGF, but not pS2 (TFF1), activate STAT3 signaling through Tyr(705) phosphorylation of both STAT3alpha and STAT3beta isoforms. Blockade of STAT3 signaling by STAT3beta, depletion of the STAT3alpha/beta isoforms by RNA interference, and pharmacologic inhibition of STAT3alpha/beta phosphorylation by cucurbitacin or STAT3 inhibitory peptide abrogates TFF- and VEGF-induced cellular invasion and reduces the growth of HCT8/S11 tumor xenografts in athymic mice. Differential gene expression analysis using DNA microarrays revealed that overexpression of STAT3beta down-regulates the VEGF receptors Flt-1, neuropilins 1 and 2, and the inhibitor of DNA binding/differentiation (Id-2) gene product involved in the neoplastic transformation. Taken together, our data suggest that TFF3 and the essential tumor angiogenesis regulator VEGF(165) exert potent proinvasive activity through STAT3 signaling in human colorectal cancer cells. We also validate new therapeutic strategies targeting STAT3 signaling by pharmacologic inhibitors and RNA interference for the treatment of colorectal cancer patients.


Asunto(s)
Neoplasias del Colon/patología , Proteínas de Unión al ADN/fisiología , Mucinas/fisiología , Proteínas Musculares/fisiología , Transactivadores/fisiología , Factor A de Crecimiento Endotelial Vascular/fisiología , Apoptosis , Secuencia de Bases , División Celular , Línea Celular Tumoral , Cartilla de ADN , Humanos , Cinética , Invasividad Neoplásica , Péptidos , Isoformas de Proteínas/fisiología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3 , Transducción de Señal/fisiología , Factor Trefoil-3
11.
Mol Cancer Ther ; 5(8): 2070-7, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16928828

RESUMEN

We recently showed by DNA microarray analysis that vascular endothelial growth factor (VEGF) receptor (VEGFR) is expressed in HCT8/S11 human colon cancer cells, suggesting that several angiogenic factors may target colon cancer cells themselves. In this study, transcripts encoding the VEGF-165 and semaphorin 3A (Sema3A) receptors and coreceptors Flt-1, KDR/Flk-1, plexin A1, and neuropilins NP-1 and NP-2 were identified by reverse transcription-PCR in the human colon cancer cell lines HCT8/S11, HT29, HCT116, and PCmsrc. Collagen invasion induced by VEGF-165 and Sema3A in HCT8/S11 cells (EC(50), 0.4-1 nmol/L) required p42/44 mitogen-activated protein kinase and signaling through RhoA/Rho-kinase-dependent and -independent pathways, respectively. As expected, the VEGFR signaling inhibitor ZD4190 selectively abrogated the proinvasive activity of VEGF in collagen gels (IC(50), 10 nmol/L) and chick heart fragments. We identify a novel function for VEGF-165 and Sema3A as proinvasive factors for human colorectal cancer cells. Interestingly, oral administration of the single drug ZD4190 to athymic mice (50 mg/kg/d, once daily) inhibited by 70% the growth of HCT8/S11 tumor cell xenografts. Combinations between the src tyrosine kinase inhibitor M475271 and ZD4190 or cisplatin resulted in additive therapeutic activity against LNM35 human lung tumor xenografts. Our data have significant implications for new therapeutic approaches and individualized treatment targeting VEGFR and src signaling pathways in combination with established clinical drugs at primary tumors and distant metastases in colon and lung cancer patients.


Asunto(s)
Neoplasias del Colon/tratamiento farmacológico , Quinazolinas/farmacología , Semaforina-3A/metabolismo , Triazoles/farmacología , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Cisplatino/farmacología , Neoplasias del Colon/patología , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Ratones , Ratones Desnudos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Invasividad Neoplásica , Piperidinas/farmacología , Receptores de Factores de Crecimiento Endotelial Vascular/efectos de los fármacos , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Semaforina-3A/efectos de los fármacos , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Sci Transl Med ; 9(374)2017 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-28123068

RESUMEN

Autologous T cells engineered to express chimeric antigen receptor against the B cell antigen CD19 (CAR19) are achieving marked leukemic remissions in early-phase trials but can be difficult to manufacture, especially in infants or heavily treated patients. We generated universal CAR19 (UCART19) T cells by lentiviral transduction of non-human leukocyte antigen-matched donor cells and simultaneous transcription activator-like effector nuclease (TALEN)-mediated gene editing of T cell receptor α chain and CD52 gene loci. Two infants with relapsed refractory CD19+ B cell acute lymphoblastic leukemia received lymphodepleting chemotherapy and anti-CD52 serotherapy, followed by a single-dose infusion of UCART19 cells. Molecular remissions were achieved within 28 days in both infants, and UCART19 cells persisted until conditioning ahead of successful allogeneic stem cell transplantation. This bridge-to-transplantation strategy demonstrates the therapeutic potential of gene-editing technology.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Linfocitos T/citología , Nucleasas de los Efectores Tipo Activadores de la Transcripción/genética , Alemtuzumab/uso terapéutico , Antígenos CD19/metabolismo , Antígeno CD52/metabolismo , Ensayos de Uso Compasivo , Femenino , Edición Génica , Trasplante de Células Madre Hematopoyéticas , Humanos , Lactante , Lentivirus/genética , Recurrencia Local de Neoplasia , Receptores de Antígenos de Linfocitos T/genética , Inducción de Remisión , Trasplante de Células Madre , Efectores Tipo Activadores de la Transcripción , Trasplante Homólogo
13.
FASEB J ; 19(1): 144-6, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15507471

RESUMEN

Inappropriate activation of the Wnt/APC/beta-catenin signaling pathways plays a critical role at early stages in a variety of human cancers. However, their respective implication in tumor cell invasion is still hypothetical. Here, we show that two activators of the canonical Wnt/beta-catenin transcription pathway, namely Dvl-2, the Axin 501-560 fragment binding glycogen synthase kinase -3beta (GSK-3beta), and the negative Wnt regulator wt-Axin did not alter cell invasion into type I collagen. In addition, both Dvl-2 and Axin 501-560 exerted a permissive action on the proinvasive activity of HGF and intestinal trefoil factor. Upstream activation of Wnt signaling by the Wnt-2 and Wnt-3a ligands, stable overexpression of Wnt-2, as well as GSK-3beta inhibition by lithium, SB216763, and GSK-3beta dominant negative forms (K85R and R96E) conferred the invasive phenotype through several proinvasive pathways. Induction of the matrix metalloprotease MMP-7 (matrilysin) gene and protein by Wnt-2 was abolished by inactivation of the AP-1 binding site in the promoter. Accordingly, invasion induced by Wnt-2 was prevented by soluble FRP-3 and FRP-1, sequestration of Gbetagamma subunits, depletion of the GSK-3beta protein by RNA interference, the c-Jun dominant negative mutant TAM67 and was not reversed by wt-Axin. Thus, the proinvasive activity of Wnt-2 is mediated by a noncanonical Wnt pathway using GSK-3beta and the AP-1 oncogene. Our data provide a potential clue for our understanding of the action and crosstalk between Wnt activators and other proinvasive pathways, in relation with matrix substrates and proteases in human cancers.


Asunto(s)
Glucógeno Sintasa Quinasa 3/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Invasividad Neoplásica/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Transducción de Señal/genética , Factor de Transcripción AP-1/metabolismo , Animales , Anticuerpos Monoclonales/metabolismo , Proteína Axina , Línea Celular , Línea Celular Transformada , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Perros , Células Epiteliales/química , Células Epiteliales/metabolismo , Células Epiteliales/virología , Vectores Genéticos/genética , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta , Glicoproteínas/farmacología , Células HT29 , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Humanos , Indoles/farmacología , Péptidos y Proteínas de Señalización Intercelular/agonistas , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intracelular , Riñón/química , Riñón/citología , Riñón/embriología , Riñón/metabolismo , Riñón/virología , Neoplasias Renales/genética , Neoplasias Renales/patología , Ligandos , Cloruro de Litio/farmacología , Maleimidas/farmacología , Metaloproteinasa 7 de la Matriz/biosíntesis , Fragmentos de Péptidos/farmacología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/química , Proteínas Represoras/farmacología , Retroviridae , Transcripción Genética/fisiología , Proteínas Wnt , Proteína wnt2
14.
Cancer Res ; 62(17): 4879-83, 2002 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12208734

RESUMEN

The c-kit tyrosine kinase inhibitor STI571 exhibits a substantial therapeutic activity in patients with chronic myeloid leukemia and gastrointestinal stromal tumors respectively associated with constitutive activation of the BCR-ABL and c-kit tyrosine kinases. Human colorectal tumors also express the c-kit proto-oncogene. The present study focuses on the anticancer activity of STI571 in human colorectal tumor cells in vitro and in vivo. The c-kit receptor was identified as a M(r) 145,000 immunoreactive band in human colon cancer cells HT29, HCT8/S11, and HCT116. Cellular invasion induced by 10 ng/ml stem cell factor (EC(50) = 3 ng/ml) in HT29 cells was blocked by 1 micro M STI571 (IC(50) = 56 nM) and pharmacological inhibitors of several oncogenic signaling pathways, namely, phosphatidylinositol 3-kinase (LY294002), Rho GTPases (Clostridium botulinum exoenzyme C3 transferase), and Rho-kinase (Y27632). STI571 inhibited HT29 cell proliferation (IC(50) = 6 micro M) and induced apoptosis in vitro. These cellular effects were associated with a decrease in tumor growth. We also demonstrated that stem cell factor is a proangiogenic factor in vivo and in vitro. These encouraging results warrant further preclinical investigations and clinical trials on the use of the c-kit inhibitor STI571 as a chemotherapeutic agent in colon cancer prevention and in treatment of advanced colorectal cancers associated with liver metastases.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Inhibidores Enzimáticos/farmacología , Piperazinas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/efectos de los fármacos , Pirimidinas/farmacología , Animales , Apoptosis/efectos de los fármacos , Benzamidas , División Celular/efectos de los fármacos , Embrión de Pollo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/crecimiento & desarrollo , Humanos , Mesilato de Imatinib , Ratones , Ratones Desnudos , Invasividad Neoplásica , Neovascularización Fisiológica/efectos de los fármacos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-kit/biosíntesis , Proteínas Proto-Oncogénicas c-kit/metabolismo , Factor de Células Madre/antagonistas & inhibidores , Factor de Células Madre/farmacología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Oncogene ; 23(19): 3317-27, 2004 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-15077188

RESUMEN

STAT3 is frequently overexpressed and constitutively activated by tyrosine phosphorylation during malignant transformation. Despite the clear importance of STAT3 in cell proliferation and survival in diverse human cancers, its possible contribution to tumor cell adhesion, motility and invasion remains hypothetical. We therefore compared the transforming properties of STAT3wt, its constitutively activated dimeric form STAT3C, and the dominant negative mutant STAT3-Y705F in human colorectal HCT8/S11 cancer cells. Both STAT3wt and STAT3C exert a permissive action to the proinvasive activity of the scatter factor HGF in HCT8/S11 cells. In contrast, the monomeric and cytoplasmic mutant Y705F induces a constitutive invasive phenotype through Wnt/Rho-independent and EGFR/PI3-kinase-dependent pathways. Accordingly, Y705F decreases cell-cell homotypic adhesions, and increases cell motility and scattering, as well as lamellipodia-type cellular extensions. STAT3-Y705F-transfected HCT8/S11 cells display an increased tyrosine phosphorylation of the cell-cell adhesion regulator beta-catenin and its dissociation from the invasion suppressor E-cadherin at cell-cell contacts. Our data imply that both invasion promoter and repressor genes are controlled by the canonical STAT3 transcription pathways. Disruption of this cascade by Y705F reveals the proinvasive potential of altered forms of STAT3 as a persistent signaling adaptor in cytokine/transforming growth factor receptor scaffolds and oncogenic pathways.


Asunto(s)
Adhesión Celular , Proteínas de Unión al ADN/fisiología , Invasividad Neoplásica , Transducción de Señal , Transactivadores/fisiología , Cadherinas/análisis , Cadherinas/fisiología , Agregación Celular , Movimiento Celular , Proteínas del Citoesqueleto/análisis , Proteínas del Citoesqueleto/fisiología , Receptores ErbB/fisiología , Humanos , Factor de Unión 1 al Potenciador Linfoide , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Factor de Transcripción STAT3 , Transactivadores/análisis , Factores de Transcripción/fisiología , Proteínas Wnt , beta Catenina
16.
FASEB J ; 17(12): 1721-3, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12958188

RESUMEN

The matrix metalloprotease matrilysin is expressed in premalignant polyps and plays a key role in local invasion during the progression of digestive tumors. In the present work, we investigated the possible relationships between the activity of the mouse and human matrilysin promoters (Mp), endogenous matrilysin protein expression, and two early oncogenetic defects frequently observed in human colonic cancers, namely activation of the src oncogene and impairment of the Wnt/APC/beta-catenin pathway. Using transient transfection assays, we report here that src signaling and the HMG-box transcription factor LEF-1 act synergistically with the proximal (-61 to -67) AP-1 binding site to transactivate the Mp in premalignant and tumorigenic kidney and colonic epithelial cells, through beta-catenin- and axin-independent signaling pathways. This synergism involves the -109 and -194 Tcf/LEF-1 binding sites in the Mp and a physical interaction between LEF-1 and c-Jun. Furthermore, src coordinates accumulation of the c-Jun factor and matrilysin transcripts. Conversely, the c-Jun dominant negative mutant TAM67 and the src tyrosine kinase inhibitor M475271 impaired src-induced Mp activation, matrilysin protein accumulation, and invasion of type I collagen gels. This mechanism may thereby contribute to cellular invasion during the early-stage adenoma/adenocarcinoma conversion and the metastatic process of digestive tumors.


Asunto(s)
Neoplasias del Colon/enzimología , Neoplasias del Colon/genética , Proteínas de Unión al ADN/metabolismo , Metaloproteinasa 7 de la Matriz/genética , Proteína Oncogénica pp60(v-src)/metabolismo , Regiones Promotoras Genéticas , Factor de Transcripción AP-1/metabolismo , Factores de Transcripción/metabolismo , Neoplasias del Colon/patología , Inducción Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Factor de Unión 1 al Potenciador Linfoide , Modelos Biológicos , Invasividad Neoplásica , Elementos de Respuesta , Transducción de Señal , Activación Transcripcional , Células Tumorales Cultivadas
17.
FASEB J ; 16(6): 565-76, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11919159

RESUMEN

Thrombin and proteinase-activated receptors (PAR) specifically regulate several functions that markedly enhance the transformation phenotype such as inflammation, cell proliferation, tumor growth, and metastasis. We recently reported that thrombin inhibits cellular invasion induced by src, hepatocyte growth factor (HGF), and leptin in kidney and colonic epithelial cells via predominant activation of the pertussis toxin (PTx) -sensitive G-proteins Galphao/Galphai. We provide pharmacological and biochemical evidence that in the presence of PTx, PAR-1 induced cellular invasion through Galpha12/Galpha13- and RhoA/Rho kinase (ROCK) -dependent signaling. However, inhibition of the endogenous small GTPase RhoA by the C3 exoenzyme, dominant-negative N19-RhoA, activated G26V-RhoD, and activators of the nitric oxide/cGMP pathways conferred invasive activity to PAR-1 via a signaling cascade using Galphaq, phospholipase C (PLC), Ca(2+)/calmodulin myosin light chain kinase (CaM-MLCK), and phosphorylation of MLC. We found that cellular invasion induced by the src oncogene is abrogated by inhibitors of the RhoA/ROCK pathway and is independent of PLC/CaM-MLCK signaling. Our data demonstrate that the RhoA and RhoD small GTPases are acting as a molecular switch of cellular invasion and reveal a novel critical mechanism by which PAR-1 bypass Galphao/i and RhoA inhibition via differential coupling to heterotrimeric G-proteins linked to divergent or convergent biological responses. Our data also indicate that Rho GTPases and ROCK mediate a src-dependent invasion signal in kidney and colonic cancer cells. We conclude that dynamic regulation of Rho GTPases activation and inactivation by oncogenes, growth factors, cGMP-inducing agents, and adhesion molecules can initiate convergent invasion signals controlled by the thrombin PAR-1 in cancer cells.-Nguyen, Q.-D., Faivre, S., Bruyneel, E., Rivat, C., Seto, M., Endo, T., Mareel, M., Emami, S., Gespach, C. RhoA- and RhoD-dependent regulatory switch of Galpha subunit signaling by PAR-1 receptors in cellular invasion.


Asunto(s)
Toxinas Botulínicas , Neoplasias del Colon/metabolismo , Proteínas de Drosophila , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Proteínas/metabolismo , Receptores de Trombina/metabolismo , Proteínas de Unión al GTP rho , Proteína de Unión al GTP rhoA/metabolismo , ADP Ribosa Transferasas/farmacología , Línea Celular , Neoplasias del Colon/patología , Subunidades alfa de la Proteína de Unión al GTP G12-G13 , Subunidades alfa de la Proteína de Unión al GTP Gq-G11 , Guanilato Ciclasa/metabolismo , Riñón/metabolismo , Modelos Biológicos , Mutación , Invasividad Neoplásica , Proteína Oncogénica pp60(v-src)/farmacología , Toxina del Pertussis , Proteínas/antagonistas & inhibidores , Receptor PAR-1 , Receptores de Trombina/agonistas , Transducción de Señal , Células Tumorales Cultivadas , Factores de Virulencia de Bordetella/farmacología , Proteína de Unión al GTP rhoA/antagonistas & inhibidores , Proteína de Unión al GTP rhoA/genética
18.
Peptides ; 25(5): 885-98, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15177885

RESUMEN

TFF peptides are involved in mucosal maintenance and repair through motogenic and antiapoptotic activities. These peptides are overexpressed during inflammatory processes and cancer progression. They also function as scatter factors, proinvasive and angiogenic agents. Such a divergence is related to the pathophysiological state of tissues submitted to persistent aggressive situations during digestive processes in the normal gastrointestinal tract, inflammatory and neoplastic diseases. In agreement with this model, TFF peptides are connected with multiple oncogenic pathways. As a consequence, the TFF signaling pathways may serve as potential targets in the control of chronic inflammation and progression of human solid tumors.


Asunto(s)
Invasividad Neoplásica/patología , Neoplasias/metabolismo , Péptidos/metabolismo , Transducción de Señal/fisiología , Humanos
19.
Hum Gene Ther ; 23(7): 668-75, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22691036

RESUMEN

For over 40 years, primary immunodeficiencies (PIDs) have featured prominently in the development and refinement of human allogeneic hematopoietic stem cell transplantation. More recently, ex vivo somatic gene therapy using autologous cells has provided remarkable evidence of clinical efficacy in patients without HLA-matched stem cell donors and in whom toxicity of allogeneic procedures is likely to be high. Together with improved preclinical models, a wealth of information has accumulated that has allowed development of safer, more sophisticated technologies and protocols that are applicable to a much broader range of diseases. In this review we summarize the status of these gene therapy trials and discuss the emerging application of similar strategies to other PIDs.


Asunto(s)
Terapia Genética , Síndromes de Inmunodeficiencia/terapia , Animales , Ensayos Clínicos como Asunto , Gammaretrovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos , Recombinación Homóloga , Humanos , Síndromes de Inmunodeficiencia/genética
20.
Blood ; 108(5): 1551-4, 2006 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16684963

RESUMEN

The biologic hallmark of polycythemia vera (PV) is the formation of endogenous erythroid colonies (EECs) with an erythropoietin-independent differentiation. Recently, it has been shown that an activating mutation of JAK2 (V617F) was at the origin of PV. In this work, we studied whether the STAT5/Bcl-xL pathway could be responsible for EEC formation. A constitutively active form of STAT5 was transduced into human erythroid progenitors and induced an erythropoietin-independent terminal differentiation and EEC formation. Furthermore, Bcl-xL overexpression in erythroid progenitors was also able to induce erythroid colonies despite the absence of erythropoietin. Conversely, siRNA-mediated STAT5 and Bcl-xL knock-down in human erythroid progenitors inhibited colony-forming unit-erythroid (CFU-E) formation in the presence of Epo. Altogether, these results demonstrate that a sustained level of the sole Bcl-xL is capable of giving rise to Epo-independent erythroid colony formation and suggest that, in PV patients, JAK2(V617F) may induce EEC via the STAT5/Bcl-xL pathway.


Asunto(s)
Eritropoyesis/fisiología , Factor de Transcripción STAT5/metabolismo , Células Madre/citología , Células Madre/fisiología , Proteína bcl-X/metabolismo , Diferenciación Celular , Ensayo de Unidades Formadoras de Colonias , Eritropoyetina/fisiología , Eliminación de Gen , Humanos , Policitemia Vera/sangre , ARN Interferente Pequeño/genética , Factor de Transcripción STAT5/genética , Proteína bcl-X/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA