Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Mol Ther ; 29(2): 597-610, 2021 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-33309883

RESUMEN

Evaluation of immune responses to adeno-associated virus (AAV)-mediated gene therapies prior to and following dose administration plays a key role in determining therapeutic safety and efficacy. This report describes up to 3 years of immunogenicity data following administration of valoctocogene roxaparvovec (BMN 270), an AAV5-mediated gene therapy encoding human B domain-deleted FVIII (hFVIII-SQ) in a phase 1/2 clinical study of adult males with severe hemophilia A. Patients with pre-existing humoral immunity to AAV5 or with a history of FVIII inhibitors were excluded from the trial. Blood plasma and peripheral blood mononuclear cell (PBMC) samples were collected at regular intervals following dose administration for assessment of humoral and cellular immune responses to both the AAV5 vector and transgene-expressed hFVIII-SQ. The predominant immune response elicited by BMN 270 administration was largely limited to the development of antibodies against the AAV5 capsid that were cross-reactive with other common AAV serotypes. No FVIII inhibitor responses were observed within 3 years following dose administration. In a context of prophylactic or on-demand corticosteroid immunosuppression given after vector infusion, AAV5 and hFVIII-SQ peptide-specific cellular immune responses were intermittently detected by an interferon (IFN)-γ and tumor necrosis factor (TNF)-α FluoroSpot assay, but they were not clearly associated with detrimental safety events or changes in efficacy measures.


Asunto(s)
Dependovirus/genética , Terapia Genética , Vectores Genéticos/genética , Hemofilia A/genética , Hemofilia A/terapia , Adulto , Reacciones Cruzadas/inmunología , Dependovirus/inmunología , Factor VIII/genética , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Interacciones Microbiota-Huesped/inmunología , Humanos , Inmunidad Humoral , Masculino , Transgenes , Resultado del Tratamiento
2.
Mol Ther ; 28(3): 723-746, 2020 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-31972133

RESUMEN

Gene therapy with adeno-associated virus (AAV) vectors has demonstrated safety and long-term efficacy in a number of trials across target organs, including eye, liver, skeletal muscle, and the central nervous system. Since the initial evidence that AAV vectors can elicit capsid T cell responses in humans, which can affect the duration of transgene expression, much progress has been made in understanding and modulating AAV vector immunogenicity. It is now well established that exposure to wild-type AAV results in priming of the immune system against the virus, with development of both humoral and T cell immunity. Aside from the neutralizing effect of antibodies, the impact of pre-existing immunity to AAV on gene transfer is still poorly understood. Herein, we review data emerging from clinical trials across a broad range of gene therapy applications. Common features of immune responses to AAV can be found, suggesting, for example, that vector immunogenicity is dose-dependent, and that innate immunity plays an important role in the outcome of gene transfer. A range of host-specific factors are also likely to be important, and a comprehensive understanding of the mechanisms driving AAV vector immunogenicity in humans will be key to unlocking the full potential of in vivo gene therapy.


Asunto(s)
Dependovirus/inmunología , Vectores Genéticos/efectos adversos , Inmunidad , Animales , Ensayos Clínicos como Asunto , Dependovirus/genética , Evaluación Preclínica de Medicamentos , Técnicas de Transferencia de Gen , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Celular , Inmunidad Humoral , Inmunidad Innata , Especificidad de Órganos , Linfocitos T/inmunología , Linfocitos T/metabolismo
3.
Hum Gene Ther ; 31(1-2): 70-79, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31650869

RESUMEN

Cocaine addiction continues to impose major burdens on affected individuals and broader society but is highly resistant to medical treatment or psychotherapy. This study was undertaken with the goal of Food and Drug Administration (FDA) permission for a first-in-human clinical trial of a gene therapy for treatment-seeking cocaine users to become and remain abstinent. The approach was based on intravenous administration of AAV8-hCocH, an adeno-associated viral vector encoding a modified plasma enzyme that metabolizes cocaine into harmless by-products. To assess systemic safety, we conducted "Good Laboratory Practice" (GLP) studies in cocaine-experienced and cocaine-naive mice at doses of 5E12 and 5E13 vector genomes/kg. Results showed total lack of viral vector-related adverse effects in all tests performed. Instead, mice given one injection of AAV8-hCocH and regular daily injections of cocaine had far less tissue pathology than cocaine-injected mice with no vector treatment. Biodistribution analysis showed the vector located almost exclusively in the liver. These results indicate that a liver-directed AAV8-hCocH gene transfer at reasonable dosage is safe, well tolerated, and effective. Thus, gene transfer therapy emerges as a radically new approach to treat compulsive cocaine abuse. In fact, based on these positive findings, the FDA recently accepted our latest request for investigational new drug application (IND 18579).


Asunto(s)
Hidrolasas de Éster Carboxílico/genética , Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/genética , Proteínas Recombinantes/genética , Animales , Biomarcadores , Trastornos Relacionados con Cocaína/genética , Trastornos Relacionados con Cocaína/terapia , Dependovirus/clasificación , Susceptibilidad a Enfermedades , Evaluación Preclínica de Medicamentos , Femenino , Orden Génico , Terapia Genética/métodos , Terapia Genética/normas , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Humanos , Masculino , Ratones , Mutación , Distribución Tisular , Resultado del Tratamiento
4.
Hum Gene Ther ; 29(10): 1183-1201, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30160169

RESUMEN

Genetically modified, autologous hematopoietic stem and progenitor cells (HSPCs) represent a new class of genetic medicine. Following this therapeutic paradigm, we are developing a product candidate, designated CD68-ET3-LV CD34+, for the treatment of the severe bleeding disorder, hemophilia A. The product consists of autologous CD34+ cells transduced with a human immunodeficiency virus 1-based, monocyte lineage-restricted, self-inactivating lentiviral vector (LV), termed CD68-ET3-LV, encoding a bioengineered coagulation factor VIII (fVIII) transgene, termed ET3, designed for enhanced expression. This vector was shown capable of high-titer manufacture under clinical scale and Good Manufacturing Practice. Biochemical and immunogenicity testing of recombinant ET3, as well as safety and efficacy testing of CD68-ET3-LV HSPCs, were utilized to demonstrate overall safety and efficacy in murine models. In the first model, administration of CD68-ET3-LV-transduced stem-cell antigen-1+ cells to hemophilia A mice resulted in sustained plasma fVIII production and hemostatic correction without signs of toxicity. Patient-derived, autologous mobilized peripheral blood (mPB) CD34+ cells are the clinical target cells for ex vivo transduction using CD68-ET3-LV, and the resulting genetically modified cells represent the investigational drug candidate. In the second model, CD68-ET3-LV gene transfer into mPB CD34+ cells isolated from normal human donors was utilized to obtain in vitro and in vivo pharmacology, pharmacokinetic, and toxicology assessment. CD68-ET3-LV demonstrated reproducible and efficient gene transfer into mPB CD34+ cells, with vector copy numbers in the range of 1 copy per diploid genome equivalent without affecting clonogenic potential. Differentiation of human CD34+ cells into monocytes was associated with increased fVIII production, supporting the designed function of the CD68 promoter. To assess in vivo pharmacodynamics, CD68-ET3-LV CD34+ cell product was administered to immunodeficient mice. Treated mice displayed sustained plasma fVIII levels and no signs of product related toxicity. Collectively, the findings of the current study support the preclinical safety and efficacy of CD68-ET3-LV CD34+.


Asunto(s)
Factor VIII/genética , Ingeniería Genética , Vectores Genéticos/genética , Células Madre Hematopoyéticas/metabolismo , Hemofilia A/genética , Hemofilia A/terapia , Lentivirus/genética , Animales , Coagulación Sanguínea , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Expresión Génica , Orden Génico , Técnicas de Transferencia de Gen , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Humanos , Masculino , Ratones , Ratones Transgénicos , Mutagénesis Insercional , Porcinos , Transducción Genética , Transgenes , Resultado del Tratamiento , Integración Viral
5.
Mol Ther ; 26(10): 2418-2430, 2018 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-30057240

RESUMEN

The present study was designed to characterize transduction of non-human primate brain and spinal cord with a modified adeno-associated virus serotype 2, incapable of binding to the heparan sulfate proteoglycan receptor, referred to as AAV2-HBKO. AAV2-HBKO was infused into the thalamus, intracerebroventricularly or via a combination of both intracerebroventricular and thalamic delivery. Thalamic injection of this modified vector encoding GFP resulted in widespread CNS transduction that included neurons in deep cortical layers, deep cerebellar nuclei, several subcortical regions, and motor neuron transduction in the spinal cord indicative of robust bidirectional axonal transport. Intracerebroventricular delivery similarly resulted in widespread cortical transduction, with one striking distinction that oligodendrocytes within superficial layers of the cortex were the primary cell type transduced. Robust motor neuron transduction was also observed in all levels of the spinal cord. The combination of thalamic and intracerebroventricular delivery resulted in transduction of oligodendrocytes in superficial cortical layers and neurons in deeper cortical layers. Several subcortical regions were also transduced. Our data demonstrate that AAV2-HBKO is a powerful vector for the potential treatment of a wide number of neurological disorders, and highlight that delivery route can significantly impact cellular tropism and pattern of CNS transduction.


Asunto(s)
Terapia Genética , Vectores Genéticos/efectos adversos , Neuronas/efectos de los fármacos , Parvovirinae/genética , Médula Espinal/efectos de los fármacos , Animales , Transporte Axonal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Proteínas de la Cápside/administración & dosificación , Proteínas de la Cápside/genética , Sistema Nervioso Central/efectos de los fármacos , Sistema Nervioso Central/patología , Dependovirus , Modelos Animales de Enfermedad , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Proteoglicanos de Heparán Sulfato/administración & dosificación , Proteoglicanos de Heparán Sulfato/genética , Humanos , Infusiones Intraventriculares , Neuronas Motoras/efectos de los fármacos , Neuronas/patología , Primates , Médula Espinal/patología , Tálamo/efectos de los fármacos
6.
Hum Gene Ther ; 28(6): 510-522, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28132521

RESUMEN

GM2 gangliosidoses, including Tay-Sachs disease and Sandhoff disease, are lysosomal storage disorders caused by deficiencies in ß-N-acetylhexosaminidase (Hex). Patients are afflicted primarily with progressive central nervous system (CNS) dysfunction. Studies in mice, cats, and sheep have indicated safety and widespread distribution of Hex in the CNS after intracranial vector infusion of AAVrh8 vectors encoding species-specific Hex α- or ß-subunits at a 1:1 ratio. Here, a safety study was conducted in cynomolgus macaques (cm), modeling previous animal studies, with bilateral infusion in the thalamus as well as in left lateral ventricle of AAVrh8 vectors encoding cm Hex α- and ß-subunits. Three doses (3.2 × 1012 vg [n = 3]; 3.2 × 1011 vg [n = 2]; or 1.1 × 1011 vg [n = 2]) were tested, with controls infused with vehicle (n = 1) or transgene empty AAVrh8 vector at the highest dose (n = 2). Most monkeys receiving AAVrh8-cmHexα/ß developed dyskinesias, ataxia, and loss of dexterity, with higher dose animals eventually becoming apathetic. Time to onset of symptoms was dose dependent, with the highest-dose cohort producing symptoms within a month of infusion. One monkey in the lowest-dose cohort was behaviorally asymptomatic but had magnetic resonance imaging abnormalities in the thalami. Histopathology was similar in all monkeys injected with AAVrh8-cmHexα/ß, showing severe white and gray matter necrosis along the injection track, reactive vasculature, and the presence of neurons with granular eosinophilic material. Lesions were minimal to absent in both control cohorts. Despite cellular loss, a dramatic increase in Hex activity was measured in the thalamus, and none of the animals presented with antibody titers against Hex. The high overexpression of Hex protein is likely to blame for this negative outcome, and this study demonstrates the variations in safety profiles of AAVrh8-Hexα/ß intracranial injection among different species, despite encoding for self-proteins.


Asunto(s)
Dependovirus/genética , Discinesias/etiología , Gangliosidosis GM2/terapia , Vectores Genéticos/efectos adversos , Necrosis/etiología , Neuronas/metabolismo , beta-N-Acetilhexosaminidasas/genética , Animales , Apatía , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Discinesias/genética , Discinesias/metabolismo , Discinesias/patología , Femenino , Gangliosidosis GM2/genética , Gangliosidosis GM2/metabolismo , Gangliosidosis GM2/patología , Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Sustancia Gris/metabolismo , Sustancia Gris/patología , Inyecciones Intraventriculares , Macaca fascicularis , Masculino , Necrosis/genética , Necrosis/metabolismo , Necrosis/patología , Neuronas/patología , Subunidades de Proteína/efectos adversos , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Tálamo/metabolismo , Tálamo/patología , Transgenes , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , beta-N-Acetilhexosaminidasas/efectos adversos , beta-N-Acetilhexosaminidasas/metabolismo
7.
Cancer Gene Ther ; 24(1): 13-19, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27934883

RESUMEN

The incidence of melanoma in the United States continues to rise, with metastatic lesions notoriously recalcitrant to therapy. There are limited effective treatment options available and a great need for more effective therapies that can be rapidly integrated in the clinic. In this study, we demonstrate that the combination of RGD-targeted adeno-associated virus phage (RGD-AAVP-TNF) with hypofractionated radiation therapy results in synergistic inhibition of primary syngeneic B16 melanoma in a C57 mouse model. Furthermore, this combination appeared to modify the tumor microenvironment, resulting in decreased Tregs in the draining LN and increased tumor-associated macrophages within the primary tumor. Finally, there appeared to be a reduction in metastatic potential and a prolongation of overall survival in the combined treatment group. These results indicate the use of targeted TNF gene therapy vector with radiation treatment could be a valuable treatment option for patients with metastatic melanoma.


Asunto(s)
Dependovirus/genética , Dependovirus/metabolismo , Vectores Genéticos/genética , Melanoma/genética , Melanoma/patología , Oligopéptidos/metabolismo , Factor de Necrosis Tumoral alfa/genética , Animales , Línea Celular Tumoral , Terapia Combinada , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Melanoma/metabolismo , Melanoma/terapia , Melanoma Experimental , Ratones , Metástasis de la Neoplasia , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Neovascularización Patológica/metabolismo , Neovascularización Patológica/terapia , Radioterapia/métodos , Radioterapia Guiada por Imagen , Análisis de Supervivencia , Resultado del Tratamiento , Carga Tumoral/genética , Carga Tumoral/inmunología , Carga Tumoral/efectos de la radiación , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
8.
Expert Rev Hematol ; 9(7): 649-59, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27153210

RESUMEN

Here we review the recent literature on Hemophilia gene transfer/therapy. Gene therapy is one of several new technologies being developed as a treatment for bleeding disorders. We will discuss current and pending clinical efforts and attempt to relate how the field is trending. In doing so, we will focus on the use of recombinant Adeno-associated viral (rAAV) vector-mediated gene transfer since all currently active trials are using this vector. Recent exciting results embody nearly 20 years of preclinical and translational research. After several early clinical attempts, therapeutic factor levels that can now be achieved reflect several modifications of the original vectors. Patterns of results are slowly starting to emerge as different AAV vectors are being tested. As with any new technology, there are drawbacks, and the potential for immune/inflammatory and oncogenic risks have emerged and will be discussed.


Asunto(s)
Terapia Genética , Hemofilia A/genética , Hemofilia A/terapia , Hemofilia B/genética , Hemofilia B/terapia , Animales , Ensayos Clínicos como Asunto , Dependovirus/clasificación , Dependovirus/genética , Evaluación Preclínica de Medicamentos , Factor IX/genética , Factor VIII/genética , Edición Génica , Marcación de Gen , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/efectos adversos , Vectores Genéticos/genética , Humanos , Reparación del Gen Blanco
9.
Toxicol Appl Pharmacol ; 268(3): 318-30, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23403069

RESUMEN

Adenoviral vectors (Ads) are promising gene delivery vehicles due to their high transduction efficiency; however, their clinical usefulness has been hampered by their immunogenicity and the presence of anti-Ad immunity in humans. We reported the efficacy of a gene therapy approach for glioma consisting of intratumoral injection of Ads encoding conditionally cytotoxic herpes simplex type 1 thymidine kinase (Ad-TK) and the immunostimulatory cytokine fms-like tyrosine kinase ligand 3 (Ad-Flt3L). Herein, we report the biodistribution, efficacy, and neurological and systemic effects of a bicistronic high-capacity Ad, i.e., HC-Ad-TK/TetOn-Flt3L. HC-Ads elicit sustained transgene expression, even in the presence of anti-Ad immunity, and can encode large therapeutic cassettes, including regulatory elements to enable turning gene expression "on" or "off" according to clinical need. The inclusion of two therapeutic transgenes within a single vector enables a reduction of the total vector load without adversely impacting efficacy. Because clinically the vectors will be delivered into the surgical cavity, normal regions of the brain parenchyma are likely to be transduced. Thus, we assessed any potential toxicities elicited by escalating doses of HC-Ad-TK/TetOn-Flt3L (1×10(8), 1×10(9), or 1×10(10) viral particles [vp]) delivered into the rat brain parenchyma. We assessed neuropathology, biodistribution, transgene expression, systemic toxicity, and behavioral impact at acute and chronic time points. The results indicate that doses up to 1×10(9) vp of HC-Ad-TK/TetOn-Flt3L can be safely delivered into the normal rat brain and underpin further developments for its implementation in a phase I clinical trial for glioma.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Ensayos Clínicos Fase I como Asunto/métodos , Citotoxinas/administración & dosificación , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Glioblastoma/tratamiento farmacológico , Inmunización/métodos , Adenoviridae/metabolismo , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Citotoxinas/efectos adversos , Citotoxinas/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Quimioterapia Combinada , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Vectores Genéticos/metabolismo , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Masculino , Ratas , Ratas Endogámicas Lew , Distribución Tisular/efectos de los fármacos , Distribución Tisular/fisiología , Resultado del Tratamiento
10.
J Virol ; 86(9): 5304-13, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22379082

RESUMEN

Herpes simplex virus 1 (HSV-1) mutants that lack the γ(1)34.5 gene are unable to replicate in the central nervous system but maintain replication competence in dividing cell populations, such as those found in brain tumors. We have previously demonstrated that a γ(1)34.5-deleted HSV-1 expressing murine interleukin-12 (IL-12; M002) prolonged survival of immunocompetent mice in intracranial models of brain tumors. We hypothesized that M002 would be suitable for use in clinical trials for patients with malignant glioma. To test this hypothesis, we (i) compared the efficacy of M002 to three other HSV-1 mutants, R3659, R8306, and G207, in murine models of brain tumors, (ii) examined the safety and biodistribution of M002 in the HSV-1-sensitive primate Aotus nancymae following intracerebral inoculation, and (iii) determined whether murine IL-12 produced by M002 was capable of activating primate lymphocytes. Results are summarized as follows: (i) M002 demonstrated superior antitumor activity in two different murine brain tumor models compared to three other genetically engineered HSV-1 mutants; (ii) no significant clinical or magnetic resonance imaging evidence of toxicity was observed following direct inoculation of M002 into the right frontal lobes of A. nancymae; (iii) there was no histopathologic evidence of disease in A. nancymae 1 month or 5.5 years following direct inoculation; and (iv) murine IL-12 produced by M002 activates A. nancymae lymphocytes in vitro. We conclude that the safety and preclinical efficacy of M002 warrants the advancement of a Δγ(1)34.5 virus expressing IL-12 to phase I clinical trials for patients with recurrent malignant glioma.


Asunto(s)
Vectores Genéticos/genética , Interleucina-12/genética , Simplexvirus/genética , Aciclovir/farmacología , Animales , Antivirales/farmacología , Aotidae , Encéfalo/patología , Línea Celular , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Femenino , Expresión Génica , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Glioma/genética , Glioma/mortalidad , Glioma/terapia , Humanos , Interleucina-12/metabolismo , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones SCID , Simplexvirus/efectos de los fármacos , Análisis de Supervivencia , Replicación Viral/efectos de los fármacos , Replicación Viral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Hear Res ; 277(1-2): 28-36, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21530627

RESUMEN

Gene-based therapeutics are being developed as novel treatments for genetic hearing loss. One roadblock to effective gene therapy is the identification of vectors which will safely deliver therapeutics to targeted cells. The cellular heterogeneity that exists within the cochlea makes viral tropism a vital consideration for effective inner ear gene therapy. There are compelling reasons to identify a viral vector with tropism for organ of Corti supporting cells. Supporting cells are the primary expression site of connexin 26 gap junction proteins that are mutated in the most common form of congenital genetic deafness (DFNB1). Supporting cells are also primary targets for inducing hair cell regeneration. Since many genetic forms of deafness are congenital it is necessary to administer gene transfer-based therapeutics prior to the onset of significant hearing loss. We have used transuterine microinjection of the fetal murine otocyst to investigate viral tropism in the developing inner ear. For the first time we have characterized viral tropism for supporting cells following in utero delivery to their progenitors. We report the inner ear tropism and potential ototoxicity of three previously untested vectors: early-generation adenovirus (Ad5.CMV.GFP), advanced-generation adenovirus (Adf.11D) and bovine adeno-associated virus (BAAV.CMV.GFP). Adenovirus showed robust tropism for organ of Corti supporting cells throughout the cochlea but induced increased ABR thresholds indicating ototoxicity. BAAV also showed tropism for organ of Corti supporting cells, with preferential transduction toward the cochlear apex. Additionally, BAAV readily transduced spiral ganglion neurons. Importantly, the BAAV-injected ears exhibited normal hearing at 5 weeks of age when compared to non-injected ears. Our results support the use of BAAV for safe and efficient targeting of supporting cell progenitors in the developing murine inner ear.


Asunto(s)
Adenoviridae/genética , Sordera/terapia , Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos , Células Laberínticas de Soporte/virología , Órgano Espiral/virología , Tropismo Viral , Estimulación Acústica , Animales , Audiometría de Tonos Puros , Umbral Auditivo , Sordera/genética , Sordera/fisiopatología , Potenciales Evocados Auditivos del Tronco Encefálico , Técnicas de Transferencia de Gen/efectos adversos , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Edad Gestacional , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Células Laberínticas de Soporte/metabolismo , Ratones , Ratones Endogámicos BALB C , Microinyecciones , Microscopía Fluorescente , Órgano Espiral/embriología , Órgano Espiral/metabolismo , Órgano Espiral/fisiopatología , Células Madre/virología , Transducción Genética
12.
Microbiol Immunol ; 55(5): 304-17, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21338384

RESUMEN

Listeria monocytogenes vectors have shown promise for delivery of viral and tumor antigens in animals. We used two mutant vector strains deleted for actA/plcB (BMB72) and actA/inlB (BMB54), and engineered both strains to secrete a heterologous nucleoprotein antigen from the Influenza A virus. Strains were evaluated in vitro and in mice. Twenty-two healthy volunteers received single oral doses of either strain in a physiological study of safety, shedding, and immunogenicity. Volunteers were observed in the hospital for seven days and had daily blood cultures, routine safety blood tests (complete blood count with differential; hepatic and renal function), and fecal cultures; none had fever, positive blood cultures, prolonged shedding, or serious or unexpected events. Four of 12 volunteers who received the actA/plcB-deleted strain had minor, transient, asymptomatic serum transaminase elevations (maximum increase 1.4× upper normal). Six of six volunteers who received ≥4 × 10(9) colony forming units had detectable mucosal immune responses to listerial antigens, but not to the vectored influenza antigen. Approximately half the volunteers had modest interferon-γ ELISpot responses to a complex listerial antigen, but none had increases over their baseline responses to the influenza antigen. Comparison with prior work suggests that foreign antigen expression, and perhaps also freezing, may adversely affect the organisms' immunogenicity.


Asunto(s)
Vectores Genéticos/efectos adversos , Inmunidad Mucosa , Vacunas contra la Influenza/efectos adversos , Interferón gamma/biosíntesis , Listeria monocytogenes/genética , Proteínas de Unión al ARN/efectos adversos , Vacunas Atenuadas/efectos adversos , Proteínas del Núcleo Viral/efectos adversos , Animales , Formación de Anticuerpos/inmunología , Toxinas Bacterianas/genética , Toxinas Bacterianas/inmunología , Toxinas Bacterianas/metabolismo , Evaluación Preclínica de Medicamentos , Ensayo de Immunospot Ligado a Enzimas , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/inmunología , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/inmunología , Proteínas Hemolisinas/metabolismo , Humanos , Inmunoglobulina G/biosíntesis , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/inmunología , Listeria monocytogenes/inmunología , Listeria monocytogenes/metabolismo , Ratones , Proteínas de la Nucleocápside , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Proteínas de Unión al ARN/metabolismo , Resultado del Tratamiento , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/metabolismo , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/inmunología , Proteínas del Núcleo Viral/metabolismo
13.
Hum Gene Ther ; 20(9): 943-54, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19416079

RESUMEN

A phase 1 clinical trial evaluating the safety of gene therapy for patients with wet age-related macular degeneration (AMD) or retinoblastoma has been completed without problems. The efficacy of gene therapy for Leber's congenital amaurosis (LCA) was reported by three groups. Gene therapy may thus hold promise as a therapeutic method for the treatment of intractable ocular diseases. However, it will first be important to precisely evaluate the efficiency and safety of alternative gene transfer vectors in a preclinical study using large animals. In the present study, we evaluated the acute local (ophthalmic) and systemic toxicity of our simian immunodeficiency virus from African green monkeys (SIVagm)-based lentiviral vectors carrying human pigment epithelium-derived factor (SIV-hPEDF) for transferring genes into nonhuman primate retinas. Transient inflammation and elevation of intraocular pressure were observed in some animals, but these effects were not dose dependent. Electroretinograms (ERGs), including multifocal ERGs, revealed no remarkable change in retinal function. Histopathologically, SIV-hPEDF administration resulted in a certain degree of inflammatory reaction and no apparent structural destruction in retinal tissue. Regarding systemic toxicity, none of the animals died, and none showed any serious side effects during the experimental course. No vector leakage was detected in serum or urine samples. We thus propose that SIVagm-mediated stable gene transfer might be useful and safe for ocular gene transfer in a clinical setting.


Asunto(s)
Proteínas del Ojo/genética , Vectores Genéticos/efectos adversos , Factores de Crecimiento Nervioso/genética , Retina/virología , Serpinas/genética , Virus de la Inmunodeficiencia de los Simios/genética , Transducción Genética , Animales , Línea Celular , Evaluación Preclínica de Medicamentos , Electrorretinografía , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Proteínas Fluorescentes Verdes , Humanos , Macaca fascicularis , Modelos Animales , Retina/patología , Transgenes , Resultado del Tratamiento
15.
Med Sci (Paris) ; 23(3): 303-9, 2007 Mar.
Artículo en Francés | MEDLINE | ID: mdl-17349293

RESUMEN

Osteoarticular disorders are the major cause of disability in Europe and North America. It is estimated that rheumatoid arthritis affects 1 % of the population and that more than two third of people over age 55 develop osteoarthritis. Because there are no satisfactory treatments, gene therapy offers a new therapeutic approach. The delivery of cDNA encoding anti-arthritic proteins to articular cells has shown therapeutic efficacy in numerous animal models in vivo. Through the development and the experimental progresses that have been made for both rheumatoid arthritis and osteoarthritis, this review discusses the different gene therapy strategies available today and the safety issues with which they may be associated. Among the different vectors available today, adeno-associated virus seems the best candidate for a direct in vivo gene delivery approach for the treatment of joint disorders.


Asunto(s)
Artritis Reumatoide/terapia , Terapia Genética , Osteoartritis/terapia , Anciano , Animales , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/fisiopatología , Cartílago Articular/metabolismo , Cartílago Articular/patología , Citocinas/antagonistas & inhibidores , Citocinas/genética , ADN Complementario/administración & dosificación , ADN Complementario/uso terapéutico , Dependovirus/genética , Perros , Doxiciclina/farmacología , Etanercept , Expresión Génica/efectos de los fármacos , Genes Sintéticos , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/efectos adversos , Vectores Genéticos/uso terapéutico , Haplorrinos , Caballos , Humanos , Inmunoglobulina G/uso terapéutico , Inyecciones Intraarticulares , Ratones , Persona de Mediana Edad , Osteoartritis/fisiopatología , Receptores del Factor de Necrosis Tumoral/uso terapéutico , Receptores Tipo II del Factor de Necrosis Tumoral/genética , Sirolimus/farmacología
16.
Blood ; 103(9): 3303-4, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-14739218

RESUMEN

We delivered the homologous erythropoietin (Epo) cDNA driven from a doxycycline-regulated promoter via recombinant adeno-associated virus in skeletal muscle of 9 cynomolgus macaques. Upon induction, rapid supraphysiologic levels of Epo were obtained. Unexpectedly, some individuals developed a profound anemia that correlated with the appearance of neutralizing antibodies against the endogenous Epo. Both the endogenous erythropoietin and vector sequences were identical. This is the first example of the inadvertent development of an autoimmune disease in primates as a result of gene transfer of a gene expressing a self-antigen. It raises some concerns when a therapeutic protein is produced at high levels from an ectopic site.


Asunto(s)
Anemia Hemolítica Autoinmune/inducido químicamente , Eritropoyetina/administración & dosificación , Eritropoyetina/efectos adversos , Terapia Genética/efectos adversos , Animales , Autoanticuerpos/sangre , Autoinmunidad , Dependovirus/genética , Doxiciclina/farmacología , Evaluación Preclínica de Medicamentos , Eritropoyetina/inmunología , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Macaca , Regiones Promotoras Genéticas/efectos de los fármacos
17.
Blood ; 103(9): 3300-2, 2004 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-14695227

RESUMEN

Gene therapy is being considered for the delivery of therapeutic proteins. We evaluated the delivery of the hormone erythropoietin (EPO) into cynomolgus macaques through intramuscularly administered adeno-associated virus (AAV) vectors. As expected, the animals developed supraphysiologic levels of EPO and polycythemia. However, severe anemia ensued in some animals because of an autoimmune response to endogenous and transgene derived EPO. This is the first example of gene therapy leading to inadvertent auto-immunity in primates.


Asunto(s)
Anemia Hemolítica Autoinmune/inducido químicamente , Eritropoyetina/efectos adversos , Eritropoyetina/uso terapéutico , Terapia Genética/efectos adversos , Animales , Autoinmunidad , Dependovirus/genética , Evaluación Preclínica de Medicamentos , Eritropoyetina/genética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/efectos adversos , Macaca , Policitemia/inducido químicamente , Transgenes
18.
Vaccine ; 19(13-14): 1567-72, 2001 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-11166876

RESUMEN

In regulating vaccines, the US Food and Drug Administration (FDA) is governed by the Code of Federal Regulations. These regulations serve as the framework for product characterization, as well as preclinical and clinical testing strategies. Novel vaccine approaches such as combination vaccines, vectored vaccines, new adjuvants, and novel delivery systems pose unique regulatory challenges for the FDA. If US licensure is sought, communication with the FDA throughout the clinical development of a product is essential to identify and implement the appropriate strategies for demonstrating the safety and effectiveness of a new product.


Asunto(s)
Industria Farmacéutica/normas , United States Food and Drug Administration/legislación & jurisprudencia , United States Food and Drug Administration/tendencias , Vacunas/normas , Adyuvantes Inmunológicos/efectos adversos , Adyuvantes Inmunológicos/normas , Adyuvantes Inmunológicos/uso terapéutico , Ensayos Clínicos como Asunto/efectos adversos , Ensayos Clínicos como Asunto/normas , Sistemas de Liberación de Medicamentos/efectos adversos , Sistemas de Liberación de Medicamentos/normas , Evaluación Preclínica de Medicamentos/efectos adversos , Evaluación Preclínica de Medicamentos/normas , Vectores Genéticos/efectos adversos , Vectores Genéticos/genética , Vectores Genéticos/normas , Concesión de Licencias , Control de Calidad , Estados Unidos , Vacunas/efectos adversos , Vacunas/uso terapéutico , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/normas , Vacunas Atenuadas/uso terapéutico , Vacunas Combinadas/efectos adversos , Vacunas Combinadas/normas , Vacunas Combinadas/uso terapéutico
19.
J Natl Cancer Inst ; 91(9): 763-71, 1999 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-10328106

RESUMEN

BACKGROUND: Preclinical studies in animal models have demonstrated tumor regression following intratumoral administration of an adenovirus vector containing wild-type p53 complementary DNA (Ad-p53). Therefore, in a phase I clinical trial, we administered Ad-p53 to 28 patients with non-small-cell lung cancer (NSCLC) whose cancers had progressed on conventional treatments. METHODS: Patients received up to six, monthly intratumoral injections of Ad-p53 by use of computed tomography-guided percutaneous fine-needle injection (23 patients) or bronchoscopy (five patients). The doses ranged from 10(6) plaque-forming units (PFU) to 10(11) PFU. RESULTS: Polymerase chain reaction (PCR) analysis showed the presence of adenovirus vector DNA in 18 (86%) of 21 patients with evaluable posttreatment biopsy specimens; vector-specific p53 messenger RNA was detected by means of reverse transcription-PCR analysis in 12 (46%) of 26 patients. Apoptosis (programmed cell death) was demonstrated by increased terminal deoxynucleotide transferase-mediated biotin uridine triphosphate nick-end labeling (TUNEL) staining in posttreatment biopsy specimens from 11 patients. Vector-related toxicity was minimal (National Cancer Institute's Common Toxicity Criteria: grade 3 = one patient; grade 4 = no patients) in 84 courses of treatment, despite repeated injections (up to six) in 23 patients. Therapeutic activity in 25 evaluable patients included partial responses in two patients (8%) and disease stabilization (range, 2-14 months) in 16 patients (64%); the remaining seven patients (28%) exhibited disease progression. CONCLUSIONS: Repeated intratumoral injections of Ad-p53 appear to be well tolerated, result in transgene expression of wild-type p53, and seem to mediate antitumor activity in a subset of patients with advanced NSCLC.


Asunto(s)
Adenoviridae , Carcinoma de Pulmón de Células no Pequeñas/terapia , Técnicas de Transferencia de Gen , Genes p53 , Terapia Genética/métodos , Neoplasias Pulmonares/terapia , Adenoviridae/genética , Adulto , Anciano , Broncoscopía , Carcinoma de Pulmón de Células no Pequeñas/diagnóstico por imagen , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , ADN Viral/aislamiento & purificación , Progresión de la Enfermedad , Femenino , Genes p53/genética , Vectores Genéticos/efectos adversos , Humanos , Etiquetado Corte-Fin in Situ , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Selección de Paciente , Análisis de Supervivencia , Tomografía Computarizada por Rayos X , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA