Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 17(3): 508-15, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19127252

RESUMEN

Intravascular delivery (1.5 x 10(9) particles and higher) of recombinant adenovirus (rAd) induces myeloid cell mediated, self-limiting hemodynamic responses in normal mice. However, we observed anaphylactoid-type reactions and exacerbated hemodynamic events following rAd injection in mice bearing malignant 4T1 mammary carcinoma. Because 4T1 tumors induce significant CD11b(+)Gr-1(+) myeloid cell expansion and activation, we set to determine whether this causes rAd-induced exaggerated responses. When treated with a single intravenous dose (1 x 10(10) particles) of rAd, mice implanted with 4T1 carcinoma succumbed due to the anaphylactoid-type reactions. In contrast, normal mice and mice implanted with a related mammary carcinoma (66cl4) that does not induce CD11b(+)Gr-1(+) cell expansion, showed minimal responses. Depletion of phagocytic CD11b(+)Gr-1(+) cells prior to rAd delivery protected 4T1 tumor-bearing animals, whereas passive transfer of CD11b(+)Gr-1(+) cells from 4T1 tumor-bearing animals was sufficient to convey susceptibility to anaphylactoid-type reactions in normal animals. We further show that there is upregulation of nitric oxide and leukotriene signaling pathways in the 4T1 tumor-induced CD11b(+)Gr-1(+) myeloid cells and that pretreating mice with inhibitors of nitric oxide synthetase and leukotrienes can attenuate the anaphylactoid-type reactions. These data show that malignant tumor growth can alter CD11b(+)Gr-1(+) myeloid cells, rendering hosts susceptible to anaphylactoid-type reactions upon intravascular treatment with rAd.


Asunto(s)
Adenoviridae/inmunología , Anafilaxia/metabolismo , Antígeno CD11b/inmunología , Diferenciación Celular , Terapia Genética/efectos adversos , Neoplasias/patología , Receptores de Quimiocina/metabolismo , Adenoviridae/genética , Anafilaxia/fisiopatología , Animales , Sistema Cardiovascular/metabolismo , Línea Celular Tumoral , Susceptibilidad a Enfermedades , Femenino , Frecuencia Cardíaca , Humanos , Antagonistas de Leucotrieno/farmacología , Liposomas , Ratones , Células Mieloides/metabolismo , Trasplante de Neoplasias , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/terapia , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Fagocitos , Receptores de Quimiocina/inmunología , Receptores de Leucotrienos/metabolismo
2.
Viral Immunol ; 21(4): 469-76, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19115936

RESUMEN

Realization of the potential clinical utility of recombinant adenovirus for gene therapy or vaccine development depends on a better understanding of the role of naturally occurring or therapy-induced anti-adenovirus antibodies. This study addresses the impact of anti-adenovirus neutralizing antibodies and the complement protein C1q on adenovirus infection of coxsackie and adenovirus receptor (CAR)-positive, and especially CAR-negative cells. Initially, transduction efficiency of adenovirus vectors was assessed in the presence or absence of human sera derived from healthy individuals that were seropositive for anti-adenovirus neutralizing antibodies. Infection was monitored by transgene expression in vitro using a replication-deficient adenovirus encoding green fluorescent protein (Ad-GFP). HeLa cells (CAR-positive) were readily infected by Ad-GFP and increasing concentrations of pooled sera increasingly inhibited infection. In contrast, rhabdomyosarcoma (RD) cells, a CAR-negative cell, were poorly infected by Ad-GFP. However, in the presence of human serum, robust GFP expression was observed. This expression was completely abrogated if the human serum was heat-inactivated. Addition of purified human C1q protein to the heat-inactivated serum restored GFP expression. Similar results were seen when human C1q protein was added to purified anti-hexon antibodies, but not to anti-fiber or anti-penton base antibodies, thus implicating anti-hexon antibodies as the infective antibody component of the human sera. These studies suggest that complement protein C1q and anti-hexon antibodies together can mediate efficient adenovirus infection in CAR-negative cell types.


Asunto(s)
Infecciones por Adenovirus Humanos/inmunología , Adenovirus Humanos/inmunología , Proteínas de la Cápside/inmunología , Complemento C1q/inmunología , Receptores Virales/inmunología , Rabdomiosarcoma/virología , Infecciones por Adenovirus Humanos/virología , Anticuerpos Antivirales/inmunología , Anticuerpos Antivirales/aislamiento & purificación , Proteínas de la Cápside/aislamiento & purificación , Proteína de la Membrana Similar al Receptor de Coxsackie y Adenovirus , Células HeLa , Humanos , Rabdomiosarcoma/inmunología
3.
Mol Ther ; 12(2): 254-63, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16043097

RESUMEN

Intravascular administration of recombinant adenovirus (rAd) in cancer patients has been well tolerated. However, dose-limiting hemodynamic responses associated with suppression of cardiac output have been observed at doses of 7.5 x 10(13) particles. While analysis of hemodynamic responses induced by small-molecule pharmaceuticals is well established, little is known about the cardiovascular effects of rAd. Telemetric cardiovascular (CV) monitoring in mice was utilized to measure hemodynamic events following intravascular rAd administration. Electrocardiogram analysis revealed a block in the SA node 3-4 min postinfusion, resulting in secondary pacemaking initiated at the AV node. This was associated with acute bradycardia, reduced blood pressure, and hypothermia followed by gradual recovery. Adenovirus-primed murine sera with high neutralizing antibody (nAb) titers could inhibit CV responses, whereas human sera with equivalent nAb titers induced by natural infection were, surprisingly, not inhibitory. Interestingly, repeat dosing within 2-4 h of the primary injection resulted in desensitization, resembling tachyphylaxis, for subsequent CV responses. Last, depletion of Kupffer cells prior to rAd infusion precluded induction of CV responses. These inhibitory effects suggest that rAd interactions with certain cells of the reticular endothelial system are associated with induction of CV responses. Significantly, these studies may provide insight into management of acute adverse effects following rAd systemic delivery, enabling a broadening of therapeutic index.


Asunto(s)
Adenoviridae/genética , Terapia Genética , Vectores Genéticos/farmacología , Frecuencia Cardíaca/efectos de los fármacos , beta-Galactosidasa/uso terapéutico , Animales , Presión Sanguínea/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas , Gasto Cardíaco/efectos de los fármacos , Sistema Cardiovascular/efectos de los fármacos , Electrocardiografía , Vectores Genéticos/uso terapéutico , Humanos , Virus de la Influenza A , Macrófagos del Hígado/fisiología , Ratones , Ratones Endogámicos BALB C
4.
Shock ; 23(6): 507-15, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15897802

RESUMEN

Control of dendritic cell (DC) function is critical for strategies to modulate innate and acquired immune responses. We examined whether transduction of murine DCs with adenoviral vectors (Adv) expressing interleukin (IL)-10 could alter their phenotype and T cell stimulatory function. Murine bone marrow-derived DCs were transduced with AdV encoding human IL-10 or green fluorescent protein (GFP). Whereas transduction of immature DCs with AdV/GFP resulted in dose-dependent maturation, DCs transduced with Adv/IL-10 maintained an immature state with low major histocompatibility complex (MHC) class II, CD86, and IL-12 expression. The Adv/IL-10 transduced DCs were phenotypically unique, characterized by suppression of IL-12 expression, failure to stimulate Th1 or Th2 cytokine responses, and retained capacity to endocytose antigen. Importantly, Adv/IL-10-transduced DCs were biologically active in vivo, in that administration of these DCs into mice before a generalized peritonitis significantly improved survival. We conclude that Adv/IL-10 transduction of DCs provides an efficient means to modulate DC function. The capacity to modify DCs by adenoviral expression of IL-10 may provide a novel ex vivo or in vivo approach to mitigate acute and chronic inflammatory diseases like sepsis.


Asunto(s)
Adenoviridae/genética , Células Dendríticas/citología , Interleucina-10/genética , Interleucina-10/uso terapéutico , Sepsis/terapia , Adenoviridae/metabolismo , Animales , Antígenos CD/biosíntesis , Antígeno B7-2 , Complejo CD3/biosíntesis , Antígenos CD40/biosíntesis , Supervivencia Celular , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/metabolismo , Endocitosis , Femenino , Citometría de Flujo , Terapia Genética/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Interleucina-12/metabolismo , Lipopolisacáridos/metabolismo , Ganglios Linfáticos/patología , Linfocitos/citología , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Endogámicos C57BL , Fenotipo , Sepsis/metabolismo , Linfocitos T/citología , Células TH1 , Células Th2/metabolismo , Factores de Tiempo
5.
Clin Cancer Res ; 10(21): 7199-206, 2004 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-15534093

RESUMEN

PURPOSE: The purpose of this study was to assess the impact of anti-adenovirus neutralizing antibodies (AdNAbs) on the distribution, tolerability, and efficacy of intravenously administered oncolytic adenovirus. A translational model was developed to evaluate the impact of humoral immunity on intravenous administration of oncolytic adenovirus in humans. EXPERIMENTAL DESIGN: Initially, severe combined immunodeficient (SCID)/beige mice were passively immunized with various amounts of human sera to establish a condition of preexisting humoral immunity similar to humans. A replication-deficient adenovirus encoding beta-galactosidase (rAd-betagal) was injected intravenously into these mice. An AdNAb titer that mitigated galactosidase transgene expression was determined. A xenograft tumor-bearing nude mouse model was developed to assess how a similar in vivo titer would impact the activity of 01/PEME, an oncolytic adenovirus, after intravenous administration. RESULTS: In SCID/beige mice, there was a dose dependence between AdNAbs and galactosidase transgene expression; 90% of transgene expression was inhibited when the titer was 80. A similar titer reconstituted in the nude mice with human serum, as was done in the SCID/beige mice, did not abrogate the antitumor efficacy of the replicating adenovirus after intravenous administration. Viral DNA increased in tumors over time. CONCLUSIONS: In intravenous administration, preexisting AdNAb titer of 80 significantly attenuated the activity of a 2.5 x 10(12) particles per kilogram dose of nonreplicating adenovirus; the same titer had no affect on the activity of an equivalent dose of replicating adenovirus. Our results suggest that a majority of patients with preexisting adenovirus immunity would be candidates for intravenous administration of oncolytic adenovirus.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Animales , Peso Corporal , Línea Celular Tumoral , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Vectores Genéticos , Humanos , Ratones , Ratones Desnudos , Ratones SCID , Trasplante de Neoplasias , Reacción en Cadena de la Polimerasa , Factores de Tiempo , beta-Galactosidasa/genética
6.
Virology ; 328(1): 52-61, 2004 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-15380358

RESUMEN

Replication competent adenoviruses with various E1 modifications designed to restrict their replication to tumor cells are being evaluated as oncolytic agents in clinical trials. In mouse models, we observed that such oncolytic adenoviruses showed greater hepatotoxicity than E1-deleted adenovirus vectors following intravenous administration. Additional studies in congenic BALB/c, nude, and beige/Scid mice demonstrated dose-dependent hepatotoxicity and indicated that beige/Scid was the most sensitive strain. Comparison of E1-containing viruses showed that hepatotoxicity correlated with expression of wild-type E1a in the liver. Pharmacokinetic analysis showed rapid increases in viral DNA levels in the liver with a virus containing wild-type E1a. This was correlated with rapid induction of TNF-alpha to high levels and with rapid elevation of serum ALT. Hepatotoxicity was significantly reduced for an adenovirus with deletions in the region E1a (dl01/07) or a virus lacking E1a. The results suggest a mechanism for hepatotoxicity involving virus-induced production of local TNF-alpha release and E1a-mediated sensitization of hepatocyte killing.


Asunto(s)
Adenoviridae/genética , Proteínas E1A de Adenovirus/metabolismo , Vectores Genéticos/farmacocinética , Hepatitis/virología , Hígado/virología , Factor de Necrosis Tumoral alfa/biosíntesis , Proteínas E1A de Adenovirus/análisis , Proteínas E1A de Adenovirus/genética , Alanina Transaminasa/sangre , Animales , ADN Viral/análisis , ADN Viral/biosíntesis , Femenino , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Hepatitis/metabolismo , Hepatitis/patología , Huésped Inmunocomprometido , Inyecciones Intravenosas , Hígado/metabolismo , Hígado/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones SCID , Modelos Animales , Mutación , Especificidad de la Especie , Factores de Tiempo , Factor de Necrosis Tumoral alfa/análisis , Virus/genética
7.
Cancer Res ; 63(14): 4003-8, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12873998

RESUMEN

Central to the development of oncolytic virotherapies for cancer will be a better understanding of the parameters that influence the outcome of virotherapy to treat disseminated cancer by i.v. administration versus regional disease by local treatment. Intratumoral administration of 01/PEME, an oncolytic adenovirus, required approximately 1000-fold less dose than i.v. administration to induce similar tumor growth inhibition. Despite the short (<10 min) circulating half-life of the virus DNA, we could monitor virus distribution to the tumor site and observed virus replication by >1000-fold increase in virus DNA copies over time. There were doses of 01/PEME for which the virus DNA concentration in the tumor increased over time but did not result in antitumor efficacy. Oncolytic virus replication at a tumor site may not be a relevant indication of antitumor efficacy. Efficient distribution to the tumor site may be one of the most critical parameters for antitumor efficacy with oncolytic virotherapy.


Asunto(s)
Adenoviridae/genética , Neoplasias de la Próstata/terapia , Neoplasias de la Próstata/virología , Adenoviridae/metabolismo , Animales , Genes p53 , Humanos , Inyecciones Intralesiones , Inyecciones Intravenosas , Masculino , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
8.
J Immunol ; 168(7): 3412-8, 2002 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-11907099

RESUMEN

The dendritic cell (DC) is the most potent APC of the immune system, capable of stimulating naive T cells to proliferate and differentiate into effector T cells. Recombinant adenovirus (Adv) readily transduces DCs in vitro allowing directed delivery of transgenes that modify DC function and immune responses. In this study we demonstrate that footpad injection of a recombinant Adv readily targets transduction of myeloid and lymphoid DCs in the draining popliteal lymph node, but not in other lymphoid organs. Popliteal DCs transduced with an empty recombinant Adv undergo maturation, as determined by high MHC class II and CD86 expression. However, transduction with vectors expressing human IL-10 limit DC maturation and associated T cell activation in the draining lymph node. The extent of IL-10 expression is dose dependent; transduction with low particle numbers (10(5)) yields only local expression, while transduction with higher particle numbers (10(7) and 10(10)) leads additionally to IL-10 appearance in the circulation. Furthermore, local DC expression of human IL-10 following in vivo transduction with low particle numbers (10(5)) significantly improves survival following cecal ligation and puncture, suggesting that compartmental modulation of DC function profoundly alters the sepsis-induced immune response.


Asunto(s)
Adenovirus Humanos/inmunología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Interleucina-10/biosíntesis , Sepsis/inmunología , Sepsis/mortalidad , Adenovirus Humanos/genética , Animales , Caspasa 3 , Inhibidores de Caspasas , Caspasas/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Dendríticas/citología , Células Dendríticas/virología , Relación Dosis-Respuesta Inmunológica , Regulación hacia Abajo/genética , Regulación hacia Abajo/inmunología , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/inmunología , Humanos , Inyecciones Subcutáneas , Interleucina-10/sangre , Interleucina-10/genética , Interleucina-6/antagonistas & inhibidores , Interleucina-6/sangre , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Sepsis/microbiología , Análisis de Supervivencia , Timo/enzimología , Timo/patología , Regulación hacia Arriba/genética , Regulación hacia Arriba/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA