Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
2.
J Clin Invest ; 131(7)2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33571163

RESUMEN

BackgroundWe conducted a phase I clinical trial that infused CCR5 gene-edited CD4+ T cells to determine how these T cells can better enable HIV cure strategies.MethodsThe aim of trial was to develop RNA-based approaches to deliver zinc finger nuclease (ZFN), evaluate the effect of CCR5 gene-edited CD4+ T cells on the HIV-specific T cell response, test the ability of infused CCR5 gene-edited T cells to delay viral rebound during analytical treatment interruption, and determine whether individuals heterozygous for CCR5 Δ32 preferentially benefit. We enrolled 14 individuals living with HIV whose viral load was well controlled by antiretroviral therapy (ART). We measured the time to viral rebound after ART withdrawal, the persistence of CCR5-edited CD4+ T cells, and whether infusion of 10 billion CCR5-edited CD4+ T cells augmented the HIV-specific immune response.ResultsInfusion of the CD4+ T cells was well tolerated, with no serious adverse events. We observed a modest delay in the time to viral rebound relative to historical controls; however, 3 of the 14 individuals, 2 of whom were heterozygous for CCR5 Δ32, showed post-viral rebound control of viremia, before ultimately losing control of viral replication. Interestingly, only these individuals had substantial restoration of HIV-specific CD8+ T cell responses. We observed immune escape for 1 of these reinvigorated responses at viral recrudescence, illustrating a direct link between viral control and enhanced CD8+ T cell responses.ConclusionThese findings demonstrate how CCR5 gene-edited CD4+ T cell infusion could aid HIV cure strategies by augmenting preexisting HIV-specific immune responses.REGISTRATIONClinicalTrials.gov NCT02388594.FundingNIH funding (R01AI104400, UM1AI126620, U19AI149680, T32AI007632) was provided by the National Institute of Allergy and Infectious Diseases (NIAID), the National Institute on Drug Abuse (NIDA), the National Institute of Mental Health (NIMH), and the National Institute of Neurological Disorders and Stroke (NINDS). Sangamo Therapeutics also provided funding for these studies.


Asunto(s)
Antirretrovirales/administración & dosificación , Linfocitos T CD4-Positivos , Edición Génica , Infecciones por VIH , VIH-1/fisiología , Transfusión de Linfocitos , Receptores CCR5 , Replicación Viral/inmunología , Adulto , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Linfocitos T CD8-positivos/inmunología , Femenino , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Infecciones por VIH/terapia , Humanos , Masculino , Persona de Mediana Edad , Receptores CCR5/genética , Receptores CCR5/inmunología , Carga Viral/genética , Carga Viral/inmunología , Replicación Viral/efectos de los fármacos , Replicación Viral/genética
3.
Mol Ther Methods Clin Dev ; 18: 839-855, 2020 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-32953934

RESUMEN

Salivary gland hypofunction causes significant morbidity and loss of quality of life for head and neck cancer patients treated with radiotherapy. Preventing hypofunction is an unmet therapeutic need. We used an adeno-associated virus serotype 2 (AAV2) vector expressing the human neurotrophic factor neurturin (CERE-120) to treat murine submandibular glands either pre- or post-irradiation (IR). Treatment with CERE-120 pre-IR, not post-IR, prevented hypofunction. RNA sequencing (RNA-seq) analysis showed reduced gene expression associated with fibrosis and the innate and humoral immune responses. We then used a minipig model with CERE-120 treatment pre-IR and also compared outcomes of the contralateral non-IR gland. Analysis of gene expression, morphology, and immunostaining showed reduced IR-related immune responses and improved secretory mechanisms. CERE-120 prevented IR-induced hypofunction and restored immune homeostasis, and there was a coordinated contralateral gland response to either damage or treatment. CERE-120 gene therapy is a potential treatment for head and neck cancer patients to influence communication among neuronal, immune, and epithelial cells to prevent IR-induced salivary hypofunction and restore immune homeostasis.

4.
Hematol Oncol Clin North Am ; 31(5): 797-808, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28895848

RESUMEN

The clinical application and regulatory strategy of genome editing for ex vivo cell therapy is derived from the intersection of two fields of study: viral vector gene therapy trials; and clinical trials with ex vivo purification and engraftment of CD34+ hematopoietic stem cells, T cells, and tumor cell vaccines. This article covers the regulatory and translational preclinical activities needed for a genome editing clinical trial modifying hematopoietic stem cells and the genesis of this current strategy based on previous clinical trials using genome-edited T cells. The SB-728 zinc finger nuclease platform is discussed because this is the most clinically advanced genome editing technology.


Asunto(s)
Edición Génica , Investigación Biomédica Traslacional , Animales , Transformación Celular Neoplásica , Endonucleasas/química , Endonucleasas/metabolismo , Edición Génica/legislación & jurisprudencia , Edición Génica/métodos , Técnicas de Inactivación de Genes , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Humanos , Pruebas de Mutagenicidad , ARN Mensajero/química , ARN Mensajero/genética , Receptores CCR5/química , Receptores CCR5/metabolismo , Medición de Riesgo , Investigación Biomédica Traslacional/legislación & jurisprudencia , Investigación Biomédica Traslacional/métodos , Dedos de Zinc
5.
Proc Natl Acad Sci U S A ; 114(5): 1165-1170, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28028237

RESUMEN

Mutations in leucine-rich repeat kinase 2 (LRRK2) and α-synuclein lead to Parkinson's disease (PD). Disruption of protein homeostasis is an emerging theme in PD pathogenesis, making mechanisms to reduce the accumulation of misfolded proteins an attractive therapeutic strategy. We determined if activating nuclear factor erythroid 2-related factor (Nrf2), a potential therapeutic target for neurodegeneration, could reduce PD-associated neuron toxicity by modulating the protein homeostasis network. Using a longitudinal imaging platform, we visualized the metabolism and location of mutant LRRK2 and α-synuclein in living neurons at the single-cell level. Nrf2 reduced PD-associated protein toxicity by a cell-autonomous mechanism that was time-dependent. Furthermore, Nrf2 activated distinct mechanisms to handle different misfolded proteins. Nrf2 decreased steady-state levels of α-synuclein in part by increasing α-synuclein degradation. In contrast, Nrf2 sequestered misfolded diffuse LRRK2 into more insoluble and homogeneous inclusion bodies. By identifying the stress response strategies activated by Nrf2, we also highlight endogenous coping responses that might be therapeutically bolstered to treat PD.


Asunto(s)
Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/antagonistas & inhibidores , Factor 2 Relacionado con NF-E2/fisiología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Enfermedad de Parkinson/metabolismo , alfa-Sinucleína/antagonistas & inhibidores , Animales , Corteza Cerebral/citología , Genes Reporteros , Células HEK293 , Humanos , Hidroquinonas/farmacología , Cuerpos de Inclusión , Células Madre Pluripotentes Inducidas/citología , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/metabolismo , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina/toxicidad , Factor 2 Relacionado con NF-E2/biosíntesis , Factor 2 Relacionado con NF-E2/genética , Neuronas/metabolismo , Cultivo Primario de Células , Agregación Patológica de Proteínas , Proteostasis , Ratas , Proteínas Recombinantes de Fusión/metabolismo , Análisis de la Célula Individual , Factores de Tiempo , alfa-Sinucleína/metabolismo , alfa-Sinucleína/toxicidad
7.
Mol Ther ; 23(8): 1380-1390, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25939491

RESUMEN

Programmed cell death-1 (PD-1) is expressed on activated T cells and represents an attractive target for gene-editing of tumor targeted T cells prior to adoptive cell transfer (ACT). We used zinc finger nucleases (ZFNs) directed against the gene encoding human PD-1 (PDCD-1) to gene-edit melanoma tumor infiltrating lymphocytes (TIL). We show that our clinical scale TIL production process yielded efficient modification of the PD-1 gene locus, with an average modification frequency of 74.8% (n = 3, range 69.9-84.1%) of the alleles in a bulk TIL population, which resulted in a 76% reduction in PD-1 surface-expression. Forty to 48% of PD-1 gene-edited cells had biallelic PD-1 modification. Importantly, the PD-1 gene-edited TIL product showed improved in vitro effector function and a significantly increased polyfunctional cytokine profile (TNFα, GM-CSF, and IFNγ) compared to unmodified TIL in two of the three donors tested. In addition, all donor cells displayed an effector memory phenotype and expanded approximately 500-2,000-fold in vitro. Thus, further study to determine the efficiency and safety of adoptive cell transfer using PD-1 gene-edited TIL for the treatment of metastatic melanoma is warranted.


Asunto(s)
Endorribonucleasas/genética , Regulación Neoplásica de la Expresión Génica , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma/terapia , Receptor de Muerte Celular Programada 1/genética , Dedos de Zinc , Alelos , Animales , Separación Celular , Citocinas/metabolismo , Femenino , Citometría de Flujo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Memoria Inmunológica , Inmunoterapia Adoptiva , Interferón gamma/metabolismo , Activación de Linfocitos/inmunología , Ratones , Metástasis de la Neoplasia , Trasplante de Neoplasias , Fenotipo , Receptor de Muerte Celular Programada 1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
8.
PLoS One ; 9(5): e96137, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24846143

RESUMEN

Following spinal cord injury (SCI) there are drastic changes that occur in the spinal microvasculature, including ischemia, hemorrhage, endothelial cell death and blood-spinal cord barrier disruption. Vascular endothelial growth factor-A (VEGF-A) is a pleiotropic factor recognized for its pro-angiogenic properties; however, VEGF has recently been shown to provide neuroprotection. We hypothesized that delivery of AdV-ZFP-VEGF--an adenovirally delivered bio-engineered zinc-finger transcription factor that promotes endogenous VEGF-A expression--would result in angiogenesis, neuroprotection and functional recovery following SCI. This novel VEGF gene therapy induces the endogenous production of multiple VEGF-A isoforms; a critical factor for proper vascular development and repair. Briefly, female Wistar rats--under cyclosporin immunosuppression--received a 35 g clip-compression injury and were administered AdV-ZFP-VEGF or AdV-eGFP at 24 hours post-SCI. qRT-PCR and Western Blot analysis of VEGF-A mRNA and protein, showed significant increases in VEGF-A expression in AdV-ZFP-VEGF treated animals (p<0.001 and p<0.05, respectively). Analysis of NF200, TUNEL, and RECA-1 indicated that AdV-ZFP-VEGF increased axonal preservation (p<0.05), reduced cell death (p<0.01), and increased blood vessels (p<0.01), respectively. Moreover, AdV-ZFP-VEGF resulted in a 10% increase in blood vessel proliferation (p<0.001). Catwalk™ analysis showed AdV-ZFP-VEGF treatment dramatically improves hindlimb weight support (p<0.05) and increases hindlimb swing speed (p<0.02) when compared to control animals. Finally, AdV-ZFP-VEGF administration provided a significant reduction in allodynia (p<0.01). Overall, the results of this study indicate that AdV-ZFP-VEGF administration can be delivered in a clinically relevant time-window following SCI (24 hours) and provide significant molecular and functional benefits.


Asunto(s)
Adenoviridae , Terapia Genética/métodos , Hiperalgesia/terapia , Traumatismos de la Médula Espinal/terapia , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Dedos de Zinc , Animales , Femenino , Células HEK293 , Humanos , Hiperalgesia/etiología , Hiperalgesia/metabolismo , Hiperalgesia/patología , Neovascularización Fisiológica/genética , Ratas , Ratas Wistar , Traumatismos de la Médula Espinal/complicaciones , Traumatismos de la Médula Espinal/metabolismo , Traumatismos de la Médula Espinal/patología , Factor A de Crecimiento Endotelial Vascular/genética
9.
N Engl J Med ; 370(10): 901-10, 2014 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-24597865

RESUMEN

BACKGROUND: CCR5 is the major coreceptor for human immunodeficiency virus (HIV). We investigated whether site-specific modification of the gene ("gene editing")--in this case, the infusion of autologous CD4 T cells in which the CCR5 gene was rendered permanently dysfunctional by a zinc-finger nuclease (ZFN)--is safe. METHODS: We enrolled 12 patients in an open-label, nonrandomized, uncontrolled study of a single dose of ZFN-modified autologous CD4 T cells. The patients had chronic aviremic HIV infection while they were receiving highly active antiretroviral therapy. Six of them underwent an interruption in antiretroviral treatment 4 weeks after the infusion of 10 billion autologous CD4 T cells, 11 to 28% of which were genetically modified with the ZFN. The primary outcome was safety as assessed by treatment-related adverse events. Secondary outcomes included measures of immune reconstitution and HIV resistance. RESULTS: One serious adverse event was associated with infusion of the ZFN-modified autologous CD4 T cells and was attributed to a transfusion reaction. The median CD4 T-cell count was 1517 per cubic millimeter at week 1, a significant increase from the preinfusion count of 448 per cubic millimeter (P<0.001). The median concentration of CCR5-modified CD4 T cells at 1 week was 250 cells per cubic millimeter. This constituted 8.8% of circulating peripheral-blood mononuclear cells and 13.9% of circulating CD4 T cells. Modified cells had an estimated mean half-life of 48 weeks. During treatment interruption and the resultant viremia, the decline in circulating CCR5-modified cells (-1.81 cells per day) was significantly less than the decline in unmodified cells (-7.25 cells per day) (P=0.02). HIV RNA became undetectable in one of four patients who could be evaluated. The blood level of HIV DNA decreased in most patients. CONCLUSIONS: CCR5-modified autologous CD4 T-cell infusions are safe within the limits of this study. (Funded by the National Institute of Allergy and Infectious Diseases and others; ClinicalTrials.gov number, NCT00842634.).


Asunto(s)
Linfocitos T CD4-Positivos/trasplante , Terapia Genética , Infecciones por VIH/terapia , Transfusión de Linfocitos , Receptores CCR5/genética , Adulto , Terapia Antirretroviral Altamente Activa , Transfusión de Sangre Autóloga , Linfocitos T CD4-Positivos/química , Terapia Combinada , ADN Viral/sangre , Femenino , Terapia Genética/efectos adversos , Terapia Genética/métodos , VIH/genética , VIH/aislamiento & purificación , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , ARN Viral/sangre , Recto/inmunología , Carga Viral
10.
Proc Natl Acad Sci U S A ; 110(12): 4697-702, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-23401527

RESUMEN

Glial proliferation and activation are associated with disease progression in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar dementia. In this study, we describe a unique platform to address the question of cell autonomy in transactive response DNA-binding protein (TDP-43) proteinopathies. We generated functional astroglia from human induced pluripotent stem cells carrying an ALS-causing TDP-43 mutation and show that mutant astrocytes exhibit increased levels of TDP-43, subcellular mislocalization of TDP-43, and decreased cell survival. We then performed coculture experiments to evaluate the effects of M337V astrocytes on the survival of wild-type and M337V TDP-43 motor neurons, showing that mutant TDP-43 astrocytes do not adversely affect survival of cocultured neurons. These observations reveal a significant and previously unrecognized glial cell-autonomous pathological phenotype associated with a pathogenic mutation in TDP-43 and show that TDP-43 proteinopathies do not display an astrocyte non-cell-autonomous component in cell culture, as previously described for SOD1 ALS. This study highlights the utility of induced pluripotent stem cell-based in vitro disease models to investigate mechanisms of disease in ALS and other TDP-43 proteinopathies.


Asunto(s)
Esclerosis Amiotrófica Lateral , Astrocitos , Células Madre Pluripotentes Inducidas , Neuronas Motoras , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Astrocitos/metabolismo , Astrocitos/patología , Línea Celular , Proliferación Celular , Supervivencia Celular , Técnicas de Cocultivo , Proteínas de Unión al ADN/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Masculino , Persona de Mediana Edad , Neuronas Motoras/metabolismo , Neuronas Motoras/patología , Mutación
11.
Hum Gene Ther ; 24(3): 245-58, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23360514

RESUMEN

Since HIV requires CD4 and a co-receptor, most commonly C-C chemokine receptor 5 (CCR5), for cellular entry, targeting CCR5 expression is an attractive approach for therapy of HIV infection. Treatment of CD4(+) T cells with zinc-finger protein nucleases (ZFNs) specifically disrupting chemokine receptor CCR5 coding sequences induces resistance to HIV infection in vitro and in vivo. A chimeric Ad5/F35 adenoviral vector encoding CCR5-ZFNs permitted efficient delivery and transient expression following anti-CD3/anti-CD28 costimulation of T lymphocytes. We present data showing CD3/CD28 costimulation substantially improved transduction efficiency over reported methods for Ad5/F35 transduction of T lymphocytes. Modifications to the laboratory scale process, incorporating clinically compatible reagents and methods, resulted in a robust ex vivo manufacturing process capable of generating >10(10) CCR5 gene-edited CD4+ T cells from healthy and HIV+ donors. CD4+ T-cell phenotype, cytokine production, and repertoire were comparable between ZFN-modified and control cells. Following consultation with regulatory authorities, we conducted in vivo toxicity studies that showed no detectable ZFN-specific toxicity or T-cell transformation. Based on these findings, we initiated a clinical trial testing the safety and feasibility of CCR5 gene-edited CD4+ T-cell transfer in study subjects with HIV-1 infection.


Asunto(s)
Enzimas de Restricción del ADN/genética , Vectores Genéticos/normas , Infecciones por VIH/genética , Infecciones por VIH/inmunología , Receptores CCR5/genética , Dedos de Zinc/genética , Adenovirus Humanos/genética , Traslado Adoptivo , Animales , Antígenos CD28/inmunología , Complejo CD3/inmunología , Enzimas de Restricción del ADN/metabolismo , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Vectores Genéticos/genética , Infecciones por VIH/terapia , Humanos , Activación de Linfocitos/inmunología , Masculino , Ratones , Fenotipo , Receptores CCR5/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transducción Genética/métodos , Transducción Genética/normas , Trasplante Heterólogo
12.
J Neurotrauma ; 29(17): 2647-59, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23016562

RESUMEN

Vascular endothelial growth factor (VEGF) plays a role in angiogenesis and has been shown to be neuroprotective following central nervous system trauma. In the present study we evaluated the pro-angiogenic and neuroprotective effects of an engineered zinc-finger protein transcription factor transactivator targeting the vascular endothelial growth factor A (VEGF-ZFP). We used two virus delivery systems, adeno-virus and adeno-associated virus, to examine the effects of early and delayed VEGF-A upregulation after brain trauma, respectively. Male Sprague-Dawley rats were subject to a unilateral fluid percussion injury (FPI) of moderate severity (2.2-2.5 atm) followed by intracerebral microinjection of either adenovirus vector (Adv) or an adeno-associated vector (AAV) carrying the VEGF-ZFP construct. Adv-VEGF-ZFP-treated animals had significantly fewer TUNEL positive cells in the injured penumbra of the cortex (p<0.001) and hippocampus (p=0.001) relative to untreated rats at 72 h post-injury. Adv-VEGF-ZFP treatment significantly improved fEPSP values (p=0.007) in the CA1 region relative to injury alone. Treatment with AAV2-VEGF-ZFP resulted in improved post-injury microvascular diameter and improved functional recovery on the balance beam and rotarod task at 30 days post-injury. Collectively, the results provide supportive evidence for the concept of acute and delayed treatment following TBI using VEGF-ZFP to induce angiogenesis, reduce cell death, and enhance functional recovery.


Asunto(s)
Lesiones Encefálicas/terapia , Terapia Genética , Factor A de Crecimiento Endotelial Vascular/genética , Dedos de Zinc/genética , Animales , Western Blotting , Lesiones Encefálicas/patología , Lesiones Encefálicas/psicología , Región CA1 Hipocampal/patología , Capilares/patología , Dependovirus/genética , Potenciales Postsinápticos Excitadores/fisiología , Vectores Genéticos , Etiquetado Corte-Fin in Situ , Potenciación a Largo Plazo , Masculino , Microinyecciones , Neovascularización Fisiológica/fisiología , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Factor A de Crecimiento Endotelial Vascular/fisiología
13.
J Neurotrauma ; 28(9): 1863-79, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21902538

RESUMEN

Recent studies have identified anti-apoptotic functions for vascular endothelial growth factor (VEGF) in the central nervous system (CNS). However, VEGF therapy has been hampered by a tendency to promote vascular permeability, edema, and inflammation. Recently, engineered zinc finger proteins (ZFPs) that upregulate multiple forms of VEGF in their natural biological ratios, have been developed to overcome these negative side effects. We used retinal trauma and ischemia models, and a cortical pial strip ischemia model to determine if VEGF upregulating ZFPs are neuroprotective in the adult CNS. Optic nerve transection and ophthalmic artery ligation lead to the apoptotic degeneration of retinal ganglion cells (RGCs) and are, respectively, two highly reproducible models for CNS trauma or ischemia. Adeno-associated vectors (AAV) vectors encoding VEGF-ZFPs (AAV-VEGF-ZFP) significantly increased RGC survival by ∼twofold at 14 days after optic nerve transection or ophthalmic artery ligation. Furthermore, AAV-VEGF-ZFP enhanced recovery of the pupillary light reflex. RECA-1 immunostaining demonstrated no appreciable differences between retinas treated with AAV-VEGF-ZFP and controls, suggesting that AAV-VEGF-ZFP treatment did not affect retinal vasculature. Following pial strip of the forelimb motor cortex, brains treated with an adenovirus encoding VEGF ZFPs (AdV-ZFP) showed higher neuronal survival, accelerated wound contraction, and reduced lesion volume between 1 and 6 weeks after injury. Behavioral testing using the cylinder test for vertical exploration showed that AdV-VEGF-ZFP treatment enhanced contralateral forelimb function within the first 2 weeks after injury. Our results indicate that VEGF ZFP therapy is neuroprotective following traumatic injury or stroke in the adult mammalian CNS.


Asunto(s)
Lesiones Encefálicas/terapia , Terapia Genética/métodos , Accidente Cerebrovascular/terapia , Regulación hacia Arriba/genética , Factor A de Crecimiento Endotelial Vascular/genética , Dedos de Zinc/genética , Animales , Conducta Animal/fisiología , Lesiones Encefálicas/genética , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/terapia , Ingeniería de Proteínas , Ratas , Recuperación de la Función/genética , Accidente Cerebrovascular/genética , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
Neurobiol Dis ; 37(2): 384-93, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19879362

RESUMEN

Spinal cord injury (SCI) leads to local vascular disruption and progressive ischemia, which contribute to secondary degeneration. Enhancing angiogenesis through the induction of vascular endothelial growth factor (VEGF)-A expression therefore constitutes an attractive therapeutic approach. Moreover, emerging evidence suggests that VEGF-A may also exhibit neurotrophic, neuroprotective, and neuroproliferative effects. Building on this previous work, we seek to examine the potential therapeutic benefits of an engineered zinc finger protein (ZFP) transcription factor designed to activate expression of all isoforms of endogenous VEGF-A (ZFP-VEGF). Administration of ZFP-VEGF resulted in increased VEGF-A mRNA and protein levels, an attenuation of axonal degradation, a significant increase in vascularity and decreased levels of apoptosis. Furthermore, ZFP-VEGF treated animals showed significant improvements in tissue preservation and neurobehavioural outcomes. These data suggest that activation of VEGF-A via the administration of an engineered ZFP transcription factor holds promise as a therapy for SCI and potentially other forms of neurotrauma.


Asunto(s)
Técnicas de Transferencia de Gen , Terapia Genética/métodos , Traumatismos de la Médula Espinal/genética , Traumatismos de la Médula Espinal/terapia , Factores de Transcripción/genética , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Apoptosis/genética , Vasos Sanguíneos/citología , Vasos Sanguíneos/metabolismo , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/farmacología , Vectores Genéticos/uso terapéutico , Neovascularización Fisiológica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Recuperación de la Función/genética , Traumatismos de la Médula Espinal/metabolismo , Factores de Transcripción/farmacología , Activación Transcripcional/genética , Resultado del Tratamiento , Regulación hacia Arriba/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Degeneración Walleriana/genética , Degeneración Walleriana/metabolismo , Degeneración Walleriana/terapia , Dedos de Zinc/genética
15.
Nat Biotechnol ; 26(7): 808-16, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18587387

RESUMEN

Homozygosity for the naturally occurring Delta32 deletion in the HIV co-receptor CCR5 confers resistance to HIV-1 infection. We generated an HIV-resistant genotype de novo using engineered zinc-finger nucleases (ZFNs) to disrupt endogenous CCR5. Transient expression of CCR5 ZFNs permanently and specifically disrupted approximately 50% of CCR5 alleles in a pool of primary human CD4(+) T cells. Genetic disruption of CCR5 provided robust, stable and heritable protection against HIV-1 infection in vitro and in vivo in a NOG model of HIV infection. HIV-1-infected mice engrafted with ZFN-modified CD4(+) T cells had lower viral loads and higher CD4(+) T-cell counts than mice engrafted with wild-type CD4(+) T cells, consistent with the potential to reconstitute immune function in individuals with HIV/AIDS by maintenance of an HIV-resistant CD4(+) T-cell population. Thus adoptive transfer of ex vivo expanded CCR5 ZFN-modified autologous CD4(+) T cells in HIV patients is an attractive approach for the treatment of HIV-1 infection.


Asunto(s)
Traslado Adoptivo/métodos , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD4-Positivos/trasplante , Desoxirribonucleasas/genética , Infecciones por VIH/prevención & control , Infecciones por VIH/cirugía , Dedos de Zinc/genética , Animales , Células Cultivadas , Mapeo Cromosómico/métodos , Ingeniería Genética/métodos , Humanos , Inmunidad Innata , Ratones , Resultado del Tratamiento
16.
Nat Biotechnol ; 25(11): 1298-306, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17965707

RESUMEN

Achieving the full potential of zinc-finger nucleases (ZFNs) for genome engineering in human cells requires their efficient delivery to the relevant cell types. Here we exploited the infectivity of integrase-defective lentiviral vectors (IDLV) to express ZFNs and provide the template DNA for gene correction in different cell types. IDLV-mediated delivery supported high rates (13-39%) of editing at the IL-2 receptor common gamma-chain gene (IL2RG) across different cell types. IDLVs also mediated site-specific gene addition by a process that required ZFN cleavage and homologous template DNA, thus establishing a platform that can target the insertion of transgenes into a predetermined genomic site. Using IDLV delivery and ZFNs targeting distinct loci, we observed high levels of gene addition (up to 50%) in a panel of human cell lines, as well as human embryonic stem cells (5%), allowing rapid, selection-free isolation of clonogenic cells with the desired genetic modification.


Asunto(s)
Reparación del ADN , Desoxirribonucleasas de Localización Especificada Tipo II/metabolismo , Células Madre Embrionarias/enzimología , Ingeniería Genética/métodos , Lentivirus/genética , Dedos de Zinc , Desoxirribonucleasas de Localización Especificada Tipo II/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Integrasas/genética , Subunidad gamma Común de Receptores de Interleucina/genética , Lentivirus/enzimología , Mutación Puntual , Moldes Genéticos , Transgenes , Integración Viral/genética
17.
Clin Cancer Res ; 13(13): 3883-91, 2007 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-17606721

RESUMEN

PURPOSE: This trial evaluated the safety, clinical activity, and immunogenicity of an allogeneic cellular immunotherapy in 55 chemotherapy-naïve patients with hormone-refractory prostate cancer (HRPC). The immunotherapy, based on the GVAX platform, is a combination of two prostate carcinoma cell lines modified with the granulocyte macrophage colony-stimulating factor (GM-CSF) gene. EXPERIMENTAL DESIGN: HRPC patients with radiologic metastases (n = 34) or rising prostate-specific antigen (PSA) only (n = 21) received a prime dose of 500 million cells and 12 boost doses of either 100 million cells (low dose) or 300 million cells (high dose) biweekly for 6 months. End points were changes in PSA, time to progression, and survival. RESULTS: Median survival was 26.2 months (95% confidence interval, 17, 36) in the radiologic group: 34.9 months (8, 57) after treatment with the high dose (n = 10) of immunotherapy and 24.0 months (11, 35) with the low dose (n = 24). The median time to bone scan progression in the radiologic group was 5.0 months (2.6, 11.6) with the high dose and 2.8 months (2.8, 5.7) with the low dose. In the rising-PSA group (n = 21) receiving the low dose, the median time to bone scan progression was 5.9 months (5.6, not reached), and median survival was 37.5 months (29, 56). No dose-limiting or autoimmune toxicities were seen; the most common adverse events were injection site reaction and fatigue. CONCLUSIONS: These results suggest that this GM-CSF-secreting, allogeneic cellular immunotherapy is well tolerated and may have clinical activity in patients with metastatic HRPC. Phase 3 trials to confirm these results are under way.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunoterapia/métodos , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/terapia , Anciano , Anciano de 80 o más Años , Progresión de la Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia , Antígeno Prostático Específico/biosíntesis , Factores de Tiempo , Resultado del Tratamiento
18.
Clin Cancer Res ; 12(11 Pt 1): 3394-401, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16740763

RESUMEN

PURPOSE: To determine the toxicity, immunologic, and clinical activity of immunotherapy with irradiated, allogeneic, prostate cancer cells expressing granulocyte macrophage colony-stimulating factor (GM-CSF) in patients with recurrent prostate cancer. PATIENTS AND METHODS: A single-institution phase I/II trial was done in hormone therapy-naïve patients with prostate-specific antigen (PSA) relapse following radical prostatectomy and absence of radiologic metastases. Treatments were administered weekly via intradermal injections of 1.2 x 10(8) GM-CSF gene-transduced, irradiated, cancer cells (6 x 10(7) LNCaP cells and 6 x 10(7) PC-3 cells) for 8 weeks. RESULTS: Twenty-one patients were enrolled and treated. Toxicities included local injection-site reactions, pruritus, and flu-like symptoms. One patient had a partial PSA response of 7-month duration. At 20 weeks post first treatment, 16 of 21 (76%) patients showed a statistically significant decrease in PSA velocity (slope) compared with prevaccination (P < 0.001). Injection site biopsies showed intradermal infiltrates consisting of CD1a+ dendritic cells and CD68+ macrophages, similar to previous clinical trials using autologous GM-CSF-transduced cancer cells. Posttreatment, patients developed new oligoclonal antibodies reactive against at least five identified antigens present in LNCaP or PC-3 cells. A high-titer antibody response against a 250-kDa antigen expressed on normal prostate epithelial cells was induced in a patient with partial PSA remission; titers of this antibody decreased when treatment ended, and subsequent PSA relapse occurred. CONCLUSIONS: This non-patient-specific prostate cancer immunotherapy has a favorable safety profile and is immunologically active. Continued clinical investigation at higher doses and with longer boosting schedules is warranted.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Inmunoterapia Activa/métodos , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/terapia , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/terapia , Anciano , Reacciones Antígeno-Anticuerpo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Inyecciones Intradérmicas , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/cirugía , Estadificación de Neoplasias , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/cirugía , Recurrencia , Medición de Riesgo , Resultado del Tratamiento
19.
Mol Ther ; 14(1): 107-17, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16690359

RESUMEN

CG7870 is a replication-selective oncolytic adenovirus genetically engineered to replicate preferentially in prostate tissue. In a previous phase I/II clinical trial of intraprostatic delivery of CG7870 for locally recurrent prostate cancer this virus was well tolerated. In this phase I study CG7870 was administered as a single intravenous infusion in a group-sequential dose escalation design (1 x 10(10) to 6 x 10(12) viral particles (vp)) to 23 patients with hormone-refractory metastatic prostate cancer. Flulike symptoms (fever, fatigue, rigors, nausea, and/or vomiting) were the most common adverse events. Three therapy-related grade 3 adverse events were reported, one of which (fatigue) was serious. At doses greater than 10(12) vp all five patients experienced asymptomatic grade 1 to 2 transaminitis and/or isolated d-dimer elevations starting on day 2 through 8; dose escalation was therefore halted at 6 x 10(12) vp. All tested patients had CG7870 genomes present in the peripheral blood for at least 90 minutes after infusion; patients in the highest dose group had persistence of genomes through 29 days. A "secondary" or "delayed" peak in plasma CG7870 genome copies (defined as a >10-fold increase in CG7870 genomes from nadir concentration) suggestive of active viral replication and shedding into the bloodstream was detected in 16/23 (70%) patients. CG7870 was detected in the saliva of 3 patients, whereas all urine samples tested negative. All patients developed antibodies to CG7870. Dose-related increases in interleukins 6 and 10 (IL-6, IL-10) blood levels were detected. The peak IL-6 concentration after CG7870 treatment was associated with a transient, asymptomatic decrease in blood pressure. No partial or complete prostate-specific antigen (PSA) responses were observed; however, 5 patients had a decrease in serum PSA of 25% to 49% following a single treatment, including 3 of 8 patients at the highest dose levels.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Antígeno Prostático Específico/sangre , Neoplasias de la Próstata/terapia , Adenoviridae/inmunología , Anciano , Anciano de 80 o más Años , Anticuerpos Antivirales/sangre , Presión Sanguínea/efectos de los fármacos , Citocinas/sangre , Replicación del ADN/genética , Relación Dosis-Respuesta a Droga , Terapia Genética/efectos adversos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Vectores Genéticos/farmacocinética , Humanos , Infusiones Intravenosas , Interleucina-10/sangre , Interleucina-6/sangre , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/genética , Saliva/metabolismo , Distribución Tisular , Resultado del Tratamiento
20.
Mol Ther ; 10(4): 799-816, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15451464

RESUMEN

We produced lethally irradiated retrovirally GM-CSF-transduced autologous renal tumor cell vaccines (GVAX) from six Japanese patients with stage IV renal cell cancer (RCC). Four patients received GVAX ranging from 1.4 x 10(8) to 3.7 x 10(8) cells on 6-17 occasions. Throughout a total of 48 vaccinations, there were no severe adverse events. After vaccination, DTH skin tests became positive to autologous RCC (auto-RCC) in all patients. The vaccination sites showed significant infiltration by CD4(+) T cells, eosinophils, and HLA-DR-positive cells. The kinetic analyses of cellular immune responses using peripheral blood lymphocytes revealed an enhanced proliferative response against auto-RCC in four patients, and cytotoxicity against auto-RCC was augmented in three patients. T cell receptor beta-chain analysis revealed oligoclonal expansion of T cells in the peripheral blood, skin biopsy specimens from DTH sites, and tumors. Western blot analysis demonstrated the induction of a humoral immune response against auto-RCC. Two of the four patients are currently alive 58 and 40 months after the initial vaccination with low-dose interleukin-2. Our results suggest that GVAX substantially enhanced the antitumor cellular and humoral immune responses, which might have contributed to the relatively long survival times of our patients in the present study.


Asunto(s)
Vacunas contra el Cáncer/uso terapéutico , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/terapia , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Neoplasias Renales/inmunología , Neoplasias Renales/terapia , Anciano , Antígenos CD/análisis , Relación CD4-CD8 , Vacunas contra el Cáncer/efectos adversos , Vacunas contra el Cáncer/genética , Carcinoma de Células Renales/metabolismo , Citocinas/inmunología , Femenino , Antígenos HLA-DR/análisis , Humanos , Neoplasias Renales/patología , Neoplasias Pulmonares/secundario , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Receptores de Antígenos de Linfocitos T alfa-beta/análisis , Pruebas Cutáneas , Transducción Genética , Vacunación/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA