Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Clin Exp Allergy ; 50(12): 1342-1351, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32909660

RESUMEN

BACKGROUND: The anti-interleukin 13 (IL-13) monoclonal antibody lebrikizumab improves lung function in patients with moderate-to-severe uncontrolled asthma, but its effects on airway inflammation and remodelling are unknown. CLAVIER was designed to assess lebrikizumab's effect on eosinophilic inflammation and remodelling. OBJECTIVE: To report safety and efficacy results from enrolled participants with available data from CLAVIER. METHODS: We performed bronchoscopy on patients with uncontrolled asthma before and after 12 weeks of randomized double-blinded treatment with lebrikizumab (n = 31) or placebo (n = 33). The pre-specified primary end-point was relative change in airway subepithelial eosinophils per mm2 of basement membrane (cells/mm2 ). Pre-specified secondary and exploratory outcomes included change in IL-13-associated biomarkers and measures of airway remodelling. RESULTS: There was a baseline imbalance in tissue eosinophils and high variability between treatment groups. There was no discernible change in adjusted mean subepithelial eosinophils/mm2 in response to lebrikizumab (95% CI, -82.5%, 97.5%). As previously observed, FEV1 increased after lebrikizumab treatment. Moreover, subepithelial collagen thickness decreased 21.5% after lebrikizumab treatment (95% CI, -32.9%, -10.2%), and fractional exhaled nitric oxide, CCL26 and SERPINB2 mRNA expression in bronchial tissues also reduced. Lebrikizumab was well tolerated, with a safety profile consistent with other lebrikizumab asthma studies. CONCLUSIONS & CLINICAL RELEVANCE: We did not observe reduced tissue eosinophil numbers in association with lebrikizumab treatment. However, in pre-specified exploratory analyses, lebrikizumab treatment was associated with reduced degree of subepithelial fibrosis, a feature of airway remodelling, as well as improved lung function and reduced key pharmacodynamic biomarkers in bronchial tissues. These results reinforce the importance of IL-13 in airway pathobiology and suggest that neutralization of IL-13 may reduce asthmatic airway remodelling. CLINICAL TRIAL REGISTRATION: NCT02099656.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Antiasmáticos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Asma/tratamiento farmacológico , Eosinófilos/efectos de los fármacos , Interleucina-13/antagonistas & inhibidores , Pulmón/efectos de los fármacos , Adolescente , Adulto , Anciano , Antiasmáticos/efectos adversos , Antiasmáticos/farmacocinética , Anticuerpos Monoclonales/efectos adversos , Anticuerpos Monoclonales/farmacocinética , Asma/inmunología , Asma/fisiopatología , Método Doble Ciego , Eosinófilos/inmunología , Eosinófilos/metabolismo , Femenino , Humanos , Pulmón/inmunología , Pulmón/fisiopatología , Masculino , Persona de Mediana Edad , Transducción de Señal , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
2.
Cancer Immunol Immunother ; 59(9): 1313-23, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20499060

RESUMEN

A critical factor in clinical development of cancer immunotherapies is the identification of tumor-associated antigens that may be related to immunotherapy potency. In this study, protein microarrays containing >8,000 human proteins were screened with serum from prostate cancer patients (N = 13) before and after treatment with a granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting whole cell immunotherapy. Thirty-three proteins were identified that displayed significantly elevated (P

Asunto(s)
Anticuerpos/sangre , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Proteínas HSP90 de Choque Térmico/biosíntesis , Inmunoterapia Adoptiva , Proteínas Nucleares/biosíntesis , Neoplasias de la Próstata/inmunología , Anticuerpos/inmunología , Biomarcadores Farmacológicos , Biomarcadores de Tumor/inmunología , Ensayos Clínicos como Asunto , Progresión de la Enfermedad , Galectinas/inmunología , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/inmunología , Ensayos Analíticos de Alto Rendimiento , Humanos , Masculino , Metástasis de la Neoplasia , Proteínas Nucleares/genética , Proteínas Nucleares/inmunología , Pronóstico , Neoplasias de la Próstata/diagnóstico , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , Análisis por Matrices de Proteínas , Estudios Retrospectivos , Análisis de Supervivencia , Resultado del Tratamiento
3.
Clin Immunol ; 133(2): 184-97, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19664962

RESUMEN

Clinical studies of cell-based immunotherapies have included both patient-specific (autologous) and non-patient-specific (allogeneic) approaches. Major concerns in using allogeneic immunotherapies are that the induced immune responses may be predominantly directed against the allogeneic HLA molecules of the cellular immunotherapy and not against its potential tumor antigens and that only the allogeneic responses will be enhanced when the immunotherapies are combined with immune checkpoint regulators in an effort to enhance overall immunotherapy potency. To evaluate these possibilities, studies were performed using the GM-CSF-secreting B16F1 cell line as autologous immunotherapy (Auto) and the same cell line modified to over-express the MHC molecule K(d) to generate an immunotherapy that expresses an allogeneic component (Allo) when injected into C57/Bl6 mice. The goal was to compare the specific anti-tumor immune responses induced by these two immunotherapies, which share an identical antigen repertoire, with the exception of the allogeneic MHC class I molecule expressed by the Allo cells, and have identical GM-CSF-secretion levels. Both immunotherapies provided similar therapeutic benefit to tumor-bearing animals with a trend towards a more pronounced tumor growth delay in animals injected with the Allo immunotherapy. This correlated with a significant increase in the number of activated DCs and T-cells in the DLN of Allo-treated animals. In addition, persistent infiltration of effector CD8(+) T-cells was detected in the tumors of animals treated with the Allo immunotherapy, which correlated with a trend towards a greater antigen-specific T-cell response in these animals. When combined with the immune checkpoint regulator anti-PD-1, tumor-specific and allogeneic immune responses were equally enhanced. Thus, the ability of an allogeneic tumor cell immunotherapy to induce a therapeutic anti-tumor immune response is comparable, if not superior, to an autologous tumor cell immunotherapy and its anti-tumor potency can be enhanced when combined with immunomodulatory compounds.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Trasplante de Células/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Trasplante Autólogo/inmunología , Trasplante Homólogo/inmunología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Movimiento Celular/inmunología , Citocinas/metabolismo , Células Dendríticas/citología , Células Dendríticas/inmunología , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/sangre , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoterapia/métodos , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Subgrupos Linfocitarios/citología , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/metabolismo , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/inmunología , Fragmentos de Péptidos/inmunología , Bazo/citología , Bazo/inmunología , Análisis de Supervivencia , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología , Transfección
4.
Clin Cancer Res ; 14(12): 3933-41, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18559615

RESUMEN

PURPOSE: Evaluate the codelivery of hyaluronidase enzyme with oncolytic adenoviruses to determine whether it improves the spread of the virus throughout tumors, thereby leading to a greater overall antitumor efficacy in tumor models. EXPERIMENTAL DESIGN: The optimal dose of hyaluronidase that provided best transduction efficiency and spread of a green fluorescent protein (GFP)-expressing adenovirus within tumors was combined with oncolytic viruses in tumor models to determine whether the combination treatment results in an improvement of antitumor efficacy. RESULTS: In mice injected with the adenovirus Ad5/35GFP and an optimal dose of hyaluronidase (50 U), a significant increase in the number of GFP-expressing cells was observed when compared with animals injected with virus only (P < 0.0001). When the oncolytic adenoviruses Ad5OV or Ad5/35 OV (OV-5 or OV5T35H) were codelivered with 50 U of hyaluronidase, a significant delay in tumor progression was observed, which translated into a significant increase in the mean survival time of tumor-bearing mice compared with either of the monotherapy-treated groups (P < 0.0001). Furthermore, the mice that received the combination of Ad5/35 OV and hyaluronidase showed the best antitumor efficacy. Importantly, the combination treatment did not increase the metastatic potential of the tumors. Lastly, the increase in virus potency observed in animals injected with both enzyme and virus correlated with enhanced virus spread throughout tumors. CONCLUSION: Antitumor activity and overall survival of mice bearing highly aggressive tumors are significantly improved by codelivery of oncolytic adenoviruses and hyaluronidase when compared with either of the monotherapy-treated groups, and it may prove to be a potent and novel approach to treating patients with cancer.


Asunto(s)
Adenoviridae/efectos de los fármacos , Hialuronoglucosaminidasa/administración & dosificación , Hialuronoglucosaminidasa/farmacología , Virus Oncolíticos/efectos de los fármacos , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , Adenoviridae/fisiología , Animales , Línea Celular Tumoral , Terapia Combinada , Femenino , Humanos , Inyecciones Intralesiones , Masculino , Ratones , Ratones Desnudos , Metástasis de la Neoplasia , Viroterapia Oncolítica , Virus Oncolíticos/fisiología , Resultado del Tratamiento , Replicación Viral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Cancer Res ; 67(9): 4399-407, 2007 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-17483354

RESUMEN

Selective replication of oncolytic viruses in tumor cells provides a promising approach for the treatment of human cancers. One of the limitations observed with oncolytic viruses currently used in the treatment of solid tumors is the inefficient spread of virus throughout the tumor mass following intratumoral injection. Data are presented showing that oncolytic adenoviruses expressing the relaxin gene and containing an Ad5/Ad35 chimeric fiber showed significantly enhanced transduction and increased virus spread throughout the tumor when compared with non-relaxin-expressing, Ad5-based viruses. The increased spread of such viruses throughout tumors correlated well with improved antitumor efficacy and overall survival in two highly metastatic tumor models. Furthermore, nonreplicating viruses expressing relaxin did not increase metastases, suggesting that high level expression of relaxin will not enhance metastatic spread of tumors. In summary, the data show that relaxin may play a role in rearranging matrix components within tumors, which helps recombinant oncolytic adenoviruses to spread effectively throughout the tumor mass and thereby increase the extent of viral replication within the tumor. Expressing relaxin from Ad5/Ad35 fiber chimeric adenoviruses may prove a potent and novel approach to treating patients with cancer.


Asunto(s)
Adenoviridae/fisiología , Viroterapia Oncolítica/métodos , Neoplasias de la Próstata/terapia , Relaxina/biosíntesis , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Línea Celular Tumoral , Colágeno/metabolismo , Femenino , Vectores Genéticos/genética , Humanos , Masculino , Ratones , Metástasis de la Neoplasia , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/virología , Relaxina/genética , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Hum Gene Ther ; 17(8): 807-20, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16942441

RESUMEN

In this study, recombinant AAV vectors pseudotyped with viral capsids derived from AAV serotypes 7 and 8 were evaluated for gene transfer in the murine striatum relative to vectors pseudotyped with AAV serotypes 2, 5, and 6. In comparison with rAAV serotype 2, pseudotyped vectors derived from AAV-7 and AAV-8 have increased transduction efficiency in the murine CNS, with the rank order rAAV-7 > rAAV-8 > rAAV-5 > rAAV-2 = rAAV-6, with all vectors demonstrating a marked tropism for neuronal transduction. Pseudotyped rAAV vector gene transfer in the brain after preimplantation of a murine 4C8 glioblastoma tumor was also evaluated. Efficiency of gene transfer to the orthotopic tumor was increased when using AAV-6, -7, and -8 capsid proteins in comparison with serotype 2, with the order rAAV-8 = rAAV-7 > rAAV-6 > rAAV-2 > rAAV-5. The increased gene transfer efficiency of rAAV vectors pseudotyped with the rAAV-8 capsid also provided enhanced therapeutic efficacy in a mouse model of glioblastoma multiforme, using vectors encoding an inhibitor of the vascular endothelial growth factor pathway. These studies demonstrate that rAAV vectors pseudotyped with capsids derived from AAV serotypes 7 and 8 provide enhanced gene transfer in the murine CNS and may offer increased therapeutic efficacy in the treatment of neurological disease.


Asunto(s)
Neoplasias Encefálicas/terapia , Cuerpo Estriado , Dependovirus , Terapia Genética , Glioblastoma/terapia , Neoplasias Experimentales/terapia , Animales , Cápside , Línea Celular Tumoral , Terapia Genética/métodos , Vectores Genéticos , Glioblastoma/genética , Humanos , Ratones , Ratones Transgénicos , Trasplante de Neoplasias/métodos , Neoplasias Experimentales/genética , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Especificidad de la Especie , Transducción Genética/métodos , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/genética
7.
Hum Gene Ther ; 20(10): 1201-13, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19572803

RESUMEN

The limited efficacy of adenovirus type 5 (Ad5)-based oncolytic viruses seen in the clinic thus far may be attributable in part to variable expression of its receptor on tumor cells. Replacement of the Ad5 fiber knob with the Ad35 fiber knob generated the Ad5/35 chimeric virus, which has previously been demonstrated to have significant antitumor activity in murine tumor models, presumably by virtue of its recognition of the CD46 receptor, which is abundant on many types of tumor cells. In the current study, a CD46 receptor transgenic mouse strain (hCD46Ge) that expresses the CD46 receptor in a pattern closely mirroring that in humans was used to study the in vivo properties of Ad5/Ad35 chimeric viruses. Vector distribution was evaluated after intravenous administration to hCD46Ge mice of an Ad5-based oncolytic adenovirus or an Ad5/35 chimeric oncolytic adenovirus (designated OV-5 and OV-5T35H, respectively), a wild-type Ad5 virus (Ad5wt), or an Ad5-based, E1-deleted adenovirus (Addl312) at 1.25 x 10(12) viral particles/kg. The amount of OV-5T35H vector genomes in the liver was at least two orders of magnitude lower than that of Ad5-based viruses. Moreover, animals injected with OV-5T35H virus had significantly lower elevations of serum proinflammatory cytokines and liver enzyme levels. Mice injected with Ad5wt lost more than 20% of their body weight and died or required euthanasia because of poor clinical condition within 4 days of virus administration. Mice treated with OV-5 lost as much as 15% of their body weight over 8-9 days, but recovered within 14 days. Mice that were treated with Addl312 or OV-5T35H exhibited no body weight loss during the study period. These studies suggest that the Ad5/35-based chimeric viruses may have a better safety profile after intravenous injection compared with Ad5-based viruses.


Asunto(s)
Adenoviridae/genética , Adenoviridae/fisiología , Proteína Cofactora de Membrana/genética , Virus Oncolíticos/genética , Virus Oncolíticos/fisiología , Replicación Viral/fisiología , Animales , Biomarcadores/metabolismo , Recuento de Células Sanguíneas , Células Sanguíneas/metabolismo , Células Endoteliales/virología , Proteínas Fluorescentes Verdes/metabolismo , Heterocigoto , Homocigoto , Humanos , Inmunidad Innata/inmunología , Hígado/enzimología , Hígado/inmunología , Hígado/virología , Ratones , Viroterapia Oncolítica/métodos , Distribución Tisular , Transducción Genética
8.
Mol Ther ; 15(5): 912-20, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17245354

RESUMEN

Regulated gene expression may be required for the clinical development of certain gene therapies. Several approaches have been developed that allow pharmacologic control of transgene expression, including the dimerizer-regulated transcriptional system in which rapamycin or its analogs function as transcriptional inducers. These compounds can also act as direct antitumor agents via inhibition of mammalian target of rapamycin (mTOR). We describe the development of an optimized recombinant adeno-associated virus (AAV) expression cassette that allows dimerizer-regulated gene expression from a single vector in vitro and in vivo. After demonstrating multiple cycles of rapamycin-dependent transgene induction following a single administration of an AAV vector in vivo, application of this regulated AAV gene expression system to the pharmacologic control of antiangiogenic therapy was evaluated in preclinical tumor models. Dimerizer-regulated vectors were constructed encoding a soluble inhibitor of the vascular endothelial growth factor (VEGF) pathway. In two subcutaneous models of glioblastoma, regulated expression of the VEGF inhibitor via recombinant AAV-mediated gene transfer, in combination with rapamycin, was shown to decrease tumor growth rate significantly. The dual properties of rapamycin--as a transcriptional inducer and mTOR inhibitor--are exploited in combination with an AAV-encoded antiangiogenic agent to provide a novel approach for the treatment of malignant diseases.


Asunto(s)
Dependovirus/genética , Terapia Genética/métodos , Neoplasias/terapia , Sirolimus/farmacología , Angiostatinas/genética , Angiostatinas/metabolismo , Animales , Western Blotting , Línea Celular , Línea Celular Tumoral , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Glioblastoma/genética , Glioblastoma/metabolismo , Glioblastoma/terapia , Humanos , Inmunosupresores/farmacología , Ratones , Ratones Desnudos , Neoplasias/irrigación sanguínea , Neoplasias/genética , Prealbúmina/genética , Regiones Promotoras Genéticas/genética , Receptores de Factores de Crecimiento Endotelial Vascular/genética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Cancer Immunol Immunother ; 56(10): 1653-65, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17410360

RESUMEN

Granulocyte-macrophage colony-stimulating factor (GM-CSF)-secreting tumor cell immunotherapies have demonstrated long-lasting, and specific anti-tumor immune responses in animal models. The studies reported here specifically evaluate two aspects of the immune response generated by such immunotherapies: the persistence of irradiated tumor cells at the immunization site, and the breadth of the immune response elicited to tumor associated antigens (TAA) derived from the immunotherapy. To further define the mechanism of GM-CSF-secreting cancer immunotherapies, immunohistochemistry studies were performed using the B16F10 melanoma tumor model. In contrast to previous reports, our data revealed that the irradiated tumor cells persisted and secreted high levels of GM-CSF at the injection site for more than 21 days. Furthermore, dense infiltrates of dendritic cells were observed only in mice treated with GM-CSF-secreting B16F10 cells, and not in mice treated with unmodified B16F10 cells with or without concurrent injection of rGM-CSF. In addition, histological studies also revealed enhanced neutrophil and CD4+ T cell infiltration, as well as the presence of apoptotic cells, at the injection site of mice treated with GM-CSF-secreting tumor cells. To evaluate the scope of the immune response generated by GM-CSF-secreting cancer immunotherapies, several related B16 melanoma tumor cell subclones that exist as a result of genetic drift in the original cell line were used to challenge mice previously immunized with GM-CSF-secreting B16F10 cells. These studies revealed that GM-CSF-secreting cancer immunotherapies elicit T cell responses that effectively control growth of related but antigenically distinct tumors. Taken together, these studies provide important new insights into the mechanism of action of this promising novel cancer immunotherapy.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Inmunoterapia Adoptiva/métodos , Neoplasias/terapia , Animales , Apoptosis , Línea Celular Tumoral , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/administración & dosificación , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Neoplasias/tratamiento farmacológico , Proteínas Recombinantes , Linfocitos T/inmunología
10.
Mol Ther ; 13(5): 956-66, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16580881

RESUMEN

The presence of the blood-brain barrier complicates drug delivery in the development of therapeutic agents for the treatment of glioblastoma multiforme (GBM). The use of local gene transfer in the brain has the potential to overcome this delivery barrier by allowing the expression of therapeutic agents directly at the tumor site. In this study, we describe the development of a recombinant adeno-associated (rAAV) serotype 8 vector that encodes an optimized soluble inhibitor, termed sVEGFR1/R2, of vascular endothelial growth factor (VEGF). VEGF is an angiogenic factor highly up-regulated in GBM tumor tissue and correlates with disease progression. In subcutaneous models of GBM, VEGF inhibition following rAAV-mediated gene transfer significantly reduces overall tumor volume and increases median survival time following a single administration of vector. Using orthotopic brain tumor models of GBM, we find that direct intracranial administration of the rAAV-sVEGFR1/R2 vector to the tumor site demonstrates anti-tumor efficacy at doses that are not efficacious following systemic delivery of the vector. We propose that rAAV-mediated gene transfer of a potent soluble VEGF inhibitor in the CNS represents an effective antiangiogenic treatment strategy for GBM.


Asunto(s)
Sistema Nervioso Central/metabolismo , Dependovirus/genética , Técnicas de Transferencia de Gen , Glioblastoma/terapia , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Línea Celular , Línea Celular Tumoral , Dependovirus/clasificación , Femenino , Terapia Genética/métodos , Vectores Genéticos , Humanos , Masculino , Ratones , Ratones Endogámicos , Ratones Desnudos , Trasplante de Neoplasias , Ratas , Ratas Desnudas , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Serotipificación , Solubilidad , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA