Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Pathog ; 9(6): e1003391, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23785279

RESUMEN

A potent therapeutic T-cell vaccine may be an alternative treatment of chronic hepatitis B virus (HBV) infection. Previously, we developed a DNA prime-adenovirus (AdV) boost vaccination protocol that could elicit strong and specific CD8+ T-cell responses to woodchuck hepatitis virus (WHV) core antigen (WHcAg) in mice. In the present study, we first examined whether this new prime-boost immunization could induce WHcAg-specific T-cell responses and effectively control WHV replication in the WHV-transgenic mouse model. Secondly, we evaluated the therapeutic effect of this new vaccination strategy in chronically WHV-infected woodchucks in combination with a potent antiviral treatment. Immunization of WHV-transgenic mice by DNA prime-AdV boost regimen elicited potent and functional WHcAg-specific CD8+ T-cell response that consequently resulted in the reduction of the WHV load below the detection limit in more than 70% of animals. The combination therapy of entecavir (ETV) treatment and DNA prime-AdV boost immunization in chronic WHV carriers resulted in WHsAg- and WHcAg-specific CD4+ and CD8+ T-cell responses, which were not detectable in ETV-only treated controls. Woodchucks receiving the combination therapy showed a prolonged suppression of WHV replication and lower WHsAg levels compared to controls. Moreover, two of four immunized carriers remained WHV negative after the end of ETV treatment and developed anti-WHs antibodies. These results demonstrate that the combined antiviral and vaccination approach efficiently elicited sustained immunological control of chronic hepadnaviral infection in woodchucks and may be a new promising therapeutic strategy in patients.


Asunto(s)
Adenoviridae , Vacunas contra Hepatitis B/farmacología , Hepatitis B Crónica/prevención & control , Inmunidad Celular/efectos de los fármacos , Inmunización Secundaria , Vacunas de ADN/microbiología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Vacunas contra Hepatitis B/genética , Vacunas contra Hepatitis B/inmunología , Hepatitis B Crónica/genética , Hepatitis B Crónica/inmunología , Humanos , Inmunidad Celular/genética , Inmunidad Celular/inmunología , Marmota , Ratones , Ratones Transgénicos , Vacunas de ADN/genética
2.
J Virol ; 87(13): 7708-16, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23637419

RESUMEN

Hepatitis D virus (HDV) superinfection of hepatitis B virus (HBV) carriers causes severe liver disease and a high rate of chronicity. Therefore, a vaccine protecting HBV carriers from HDV superinfection is needed. To protect from HDV infection an induction of virus-specific T cells is required, as antibodies to the two proteins of HDV, p24 and p27, do not neutralize the HBV-derived envelope of HDV. In mice, HDV-specific CD8(+) and CD4(+) T cell responses were induced by a DNA vaccine expressing HDV p27. In subsequent experiments, seven naive woodchucks were immunized with a DNA prime and adenoviral boost regimen prior to simultaneous woodchuck hepatitis virus (WHV) and HDV infection. Five of seven HDV-immunized woodchucks were protected against HDV infection, while acute self-limiting WHV infection occurred as expected. The two animals with the breakthrough had a shorter HDV viremia than the unvaccinated controls. The DNA prime and adenoviral vector boost vaccination protected woodchucks against HDV infection in the setting of simultaneous infection with WHV and HDV. In future experiments, the efficacy of this protocol to protect from HDV infection in the setting of HDV superinfection will need to be proven.


Asunto(s)
Virus de la Hepatitis B de la Marmota/inmunología , Hepatitis B/complicaciones , Hepatitis D/prevención & control , Virus de la Hepatitis Delta/inmunología , Sobreinfección/prevención & control , Linfocitos T/inmunología , Adenoviridae , Animales , Anticuerpos Antivirales/sangre , Aspartato Aminotransferasas/metabolismo , Western Blotting , Cartilla de ADN/genética , Ensayo de Inmunoadsorción Enzimática , Femenino , Vectores Genéticos/inmunología , Hepatitis D/complicaciones , Inmunización Secundaria , Marmota , Ratones , Plásmidos/genética , Sobreinfección/virología , Vacunas Virales/genética , Vacunas Virales/inmunología
3.
J Virol ; 86(17): 9297-310, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22718818

RESUMEN

Induction of hepatitis B virus (HBV)-specific cytotoxic T cells by therapeutic immunization may be a strategy to treat chronic hepatitis B. In the HBV animal model, woodchucks, the application of DNA vaccine expressing woodchuck hepatitis virus (WHV) core antigen (WHcAg) in combination with antivirals led to the prolonged control of viral replication. However, it became clear that the use of more potent vaccines is required to overcome WHV persistence. Therefore, we asked whether stronger and more functional T-cell responses could be achieved using the modified vaccines and an optimized prime-boost vaccination regimen. We developed a new DNA plasmid (pCGWHc) and recombinant adenoviruses (AdVs) showing high expression levels of WHcAg. Mice vaccinated with the improved plasmid pCGWHc elicited a stronger WHcAg-specific CD8(+) T-cell response than with the previously used vaccines. Using multicolor flow cytometry and an in vivo cytotoxicity assay, we showed that immunization in a DNA prime-AdV boost regimen resulted in an even more vigorous and functional T-cell response than immunization with the new plasmid alone. Immunization of naïve woodchucks with pCGWHc plasmid or AdVs induced a significant WHcAg-specific degranulation response prior to the challenge, this response had not been previously detected. Consistently, this response led to a rapid control of infection after the challenge. Our results demonstrate that high antigen expression levels and the DNA prime-AdV boost immunization improved the T-cell response in mice and induced significant T-cell responses in woodchucks. Therefore, this new vaccination strategy may be a candidate for a therapeutic vaccine against chronic HBV infection.


Asunto(s)
Adenoviridae/inmunología , Antígenos del Núcleo de la Hepatitis B/inmunología , Vacunas contra Hepatitis B/inmunología , Virus de la Hepatitis B/inmunología , Hepatitis B/inmunología , Linfocitos T/inmunología , Vacunas de ADN/inmunología , Adenoviridae/genética , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Hepatitis B/virología , Antígenos del Núcleo de la Hepatitis B/administración & dosificación , Antígenos del Núcleo de la Hepatitis B/genética , Vacunas contra Hepatitis B/administración & dosificación , Vacunas contra Hepatitis B/genética , Virus de la Hepatitis B/genética , Humanos , Inmunización Secundaria , Marmota , Ratones , Ratones Endogámicos C57BL , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética
4.
J Virol ; 86(3): 1706-16, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22090142

RESUMEN

Processing and presentation of vaccine antigens by professional antigen-presenting cells (APCs) is of great importance for the efficient induction of protective immunity. We analyzed whether the efficacy of an adenovirus-based retroviral vaccine can be enhanced by coadministration of adenovirus-encoded chemokines that attract and stimulate APCs. In the Friend retrovirus (FV) mouse model we coexpressed CCL3, CCL20, CCL21, or CXCL14 from adenoviral vectors, together with FV Gag and Env antigens, and then analyzed immune responses and protection from pathogenic FV infection. Although most tested chemokines did not improve protection against FV challenge, mice that received adenoviral vectors encoding CCL3 together with FV antigens showed significantly better control over viral loads and FV-induced disease than mice immunized with the viral antigens only. Improved protection correlated with enhanced virus-specific CD4+ T cell responses and higher neutralizing antibody titers. To apply these results to an HIV vaccine, mice were immunized with adenoviral vectors encoding the HIV antigens Env and Gag-Pol and coadministered vectors encoding CCL3. Again, this combination vaccine induced higher virus-specific antibody titers and CD4+ T cell responses than did the HIV antigens alone. These results indicate that coexpression of the chemokine CCL3 by adenovirus-based vectors may be a promising tool to improve antiretroviral vaccination strategies.


Asunto(s)
Adenoviridae/inmunología , Quimiocina CCL3/administración & dosificación , Infecciones por Retroviridae/prevención & control , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Línea Celular , Humanos , Ratones
5.
Virol J ; 10: 108, 2013 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-23560981

RESUMEN

BACKGROUND: Inert nanoparticles are attracting attention as carriers for protein-based vaccines. Here we evaluate the immunogenicity of the model antigen ovalbumin delivered on polystyrene particles and directly compare particulate delivery with adenovirus-based immunization. FINDINGS: Mice were vaccinated with soluble ovalbumin, ovalbumin-coated polystyrene particles of different sizes, or an adenovirus-based expression-display vector that encodes and displays a pIX-ovalbumin fusion protein. Antibody responses were clearly higher when ovalbumin was administered on polystyrene particles compared to soluble protein administration, regardless of the particle size. Compared to adenovirus-based immunization, antibody levels were lower if an equivalent amount of protein was delivered, and no cellular immune response was detectable. CONCLUSIONS: We demonstrate in a side-by-side comparison that inert nanoparticles allow for the reduction of the administered antigen amount compared to immunization with soluble protein and induce strongly enhanced antibody responses, but responses are lower compared to adenovirus-based immunization.


Asunto(s)
Adenoviridae/química , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/farmacocinética , Nanopartículas/química , Vacunación/métodos , Vacunas/administración & dosificación , Vacunas/inmunología , Animales , Antígenos/administración & dosificación , Antígenos/inmunología , Portadores de Fármacos/química , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Poliestirenos
6.
Clin Transl Sci ; 16(9): 1569-1581, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37408165

RESUMEN

Consensus of regulatory decisions on the same Marketing Authorization Application (MAA) are critical for stakeholders. In this context, regulatory decision patterns from the Swissmedic (SMC), the US Food and Drug Administration (FDA), and the European Medicines Agency (EMA) were analyzed for hemato-oncology products (OP) and non-oncology products (NOP). We compared 336 SMC regulatory decisions between 2009 and 2018 on new active substances with the EMA and the FDA for OP (n = 77) and NOP (n = 259) regarding approval rates, consensus, and divergent decisions. For OP MAA, we analyzed the underlying reasons for divergent decisions; for consensus decisions, the similarity and strictness of labeling. For OP, the approval rate for the SMC was 88.4%, the EMA 91.3%, and the FDA 95.7%. For NOP, the SMC had an approval rate of 86.2%, the EMA of 93.8%, and the FDA of 88.8%. The consensus decision rate among agencies was 88.4% for OP and 84.4% for NOP. The main clinical driver for divergent decisions for OP was nonrandomized trial design and low patient numbers. Comparing the approved indication wordings, the highest similarity was between the SMC and the EMA, and lowest for the FDA and the EMA. Investigating label strictness, the FDA numerically had the highest but not-statistically significant number of strict labels. The approval rate stratified by disease area (OP and NOP) differed among the SMC, the EMA, and the FDA. High concordance in regulatory decisions was observed between agencies for OP as well as NOP. Reasons for divergent decisions regarding OP were mainly due to scientific uncertainties. Comparing strictness of indications, numerical but no statistically significant differences were observed between agencies.


Asunto(s)
Aprobación de Drogas , Estados Unidos , Humanos , United States Food and Drug Administration , Incertidumbre , Europa (Continente)
7.
Retrovirology ; 8: 75, 2011 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-21943056

RESUMEN

BACKGROUND: Type I interferons (IFNs) exhibit direct antiviral effects, but also distinct immunomodulatory properties. In this study, we analyzed type I IFN subtypes for their effect on prophylactic adenovirus-based anti-retroviral vaccination of mice against Friend retrovirus (FV) or HIV. RESULTS: Mice were vaccinated with adenoviral vectors encoding FV Env and Gag proteins alone or in combination with vectors encoding IFNα1, IFNα2, IFNα4, IFNα5, IFNα6, IFNα9 or IFNß. Only the co-administration of adenoviral vectors encoding IFNα2, IFNα4, IFNα6 and IFNα9 resulted in strongly improved immune protection of vaccinated mice from subsequent FV challenge infection with high control over FV-induced splenomegaly and reduced viral loads. The level of protection correlated with augmented virus-specific CD4(+) T cell responses and enhanced antibody titers. Similar results were obtained when mice were vaccinated against HIV with adenoviral vectors encoding HIV Env and Gag-Pol in combination with various type I IFN encoding vectors. Here mainly CD4(+) T cell responses were enhanced by IFNα subtypes. CONCLUSIONS: Our results indicate that certain IFNα subtypes have the potential to improve the protective effect of adenovirus-based vaccines against retroviruses. This correlated with augmented virus-specific CD4(+) T cell and antibody responses. Thus, co-expression of select type I IFNs may be a valuable tool for the development of anti-retroviral vaccines.


Asunto(s)
Antígenos Virales/inmunología , Infecciones por VIH/prevención & control , Interferón Tipo I/inmunología , Infecciones por Retroviridae/prevención & control , Vacunas Virales/inmunología , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Anticuerpos Antivirales/inmunología , Antígenos Virales/administración & dosificación , Antígenos Virales/genética , Línea Celular , Femenino , Virus de la Leucemia Murina de Friend/genética , Virus de la Leucemia Murina de Friend/inmunología , Virus de la Leucemia Murina de Friend/fisiología , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , VIH/genética , VIH/inmunología , VIH/fisiología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Humanos , Interferón Tipo I/administración & dosificación , Interferón Tipo I/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/virología , Carga Viral , Vacunas Virales/administración & dosificación , Vacunas Virales/genética
8.
J Virol ; 84(4): 1967-76, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20007267

RESUMEN

We present a new type of adenoviral vector that both encodes and displays a vaccine antigen on the capsid, thus combining in itself gene-based and protein vaccination; this vector resulted in an improved vaccination outcome in the Friend virus (FV) model. For presentation of the envelope protein gp70 of Friend murine leukemia virus on the adenoviral capsid, gp70 was fused to the adenovirus capsid protein IX. When compared to vaccination with conventional FV Env- and Gag-encoding adenoviral vectors, vaccination with the adenoviral vector that encodes and displays pIX-gp70 combined with an FV Gag-encoding vector resulted in significantly improved protection against systemic FV challenge infection, with highly controlled viral loads in plasma and spleen. This improved protection correlated with improved neutralizing antibody titers and stronger CD4(+) T-cell responses. Using a vector that displays gp70 without encoding it, we found that while the antigen display on the capsid alone was sufficient to induce high levels of binding antibodies, in vivo expression was necessary for the induction of neutralizing antibodies. This new type of adenovirus-based vaccine could be a valuable tool for vaccination.


Asunto(s)
Adenoviridae/genética , Adenoviridae/inmunología , Antígenos Virales/genética , Virus de la Leucemia Murina de Friend/genética , Virus de la Leucemia Murina de Friend/inmunología , Vacunación , Animales , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Presentación de Antígeno , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Femenino , Vectores Genéticos , Humanos , Leucemia Experimental/inmunología , Leucemia Experimental/prevención & control , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Retroviridae/inmunología , Infecciones por Retroviridae/prevención & control , Infecciones Tumorales por Virus/inmunología , Infecciones Tumorales por Virus/prevención & control , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología
9.
World J Gastroenterol ; 14(12): 1842-50, 2008 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-18350621

RESUMEN

AIM: To promote the development of improved tumor vaccination strategies relying on the intratumoral expression of viral fusogenic membrane proteins, we elucidated whether the size of syncytia or the way tumor cells die has an effect on the therapeutic outcome. METHODS: In two syngeneic subcutaneous murine colon cancer models we assessed the anti-neoplastic effect on vector-treated and contralateral untreated tumors. RESULTS: Intratumoral injection of a replication-defective adenovirus encoding respiratory syncytial virus fusion protein (RSV-F) alone (Ad.RSV-F) or together with the attachment glycoprotein RSV-G (Ad.RSV-F/G) led to a significant growth reduction of the vector-treated and contralateral untreated tumors. The treatment response was associated with a strong tumor-specific CTL response and significantly improved survival with medians of 46 d and 44 d, respectively. Intratumoral injection of Ad.RSV-G or a soluble RSV-F encoding adenovirus (Ad.RSV-F(sol)) had no significant anti-neoplastic effect. The median survival of these treatment groups and of Ad.Null-treated control animals was about 30 d. CONCLUSION: Although in vitro transduction of colon cancer cell lines with Ad.RSV-F/G resulted in about 8-fold larger syncytia than with Ad.RSV-F, the in vivo outcome was not significantly different. Transduction of murine colon cancer cell lines with Ad.RSV-F or Ad.RSV-F/G caused apoptotic cell death, in contrast to transduction with Ad.RSV-G or Ad.RSV-F(sol), suggesting an importance of the mode of cell death. Overall, these findings provide insight into improved tumor vaccination strategies relying on the intratumoral expression of viral fusogenic membrane proteins.


Asunto(s)
Apoptosis/fisiología , Células Gigantes , Virus Sincitiales Respiratorios/metabolismo , Proteínas del Envoltorio Viral/inmunología , Proteínas Virales de Fusión/inmunología , Adenoviridae/genética , Adenoviridae/metabolismo , Animales , Antineoplásicos/metabolismo , Fusión Celular , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Células Gigantes/citología , Células Gigantes/fisiología , Humanos , Estimación de Kaplan-Meier , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas del Envoltorio Viral/genética , Proteínas Virales de Fusión/genética
10.
Mol Cancer Ther ; 6(7): 1942-50, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17620425

RESUMEN

Although cancers can naturally elicit immune responses, immune ignorance is a common observation preventing immune-mediated elimination of tumor cells. We assessed whether intratumoral expression of respiratory syncytial virus fusion (RSV-F) protein, encoded by a replication-defective adenovirus vector (Ad.RSV-F), alone or in combination with local coexpression of cytokines can induce tumor-specific immune responses in a syngeneic murine colon cancer model. We confirmed in vitro by dye colocalization that transduction of murine cells with Ad.RSV-F induces cell-cell fusion. In vivo, we showed in a bilateral syngeneic s.c. colon cancer model in C57BL/6 and BALB/c mice that intratumoral injection of Ad.RSV-F leads to a significant volume reduction not only of the directly vector-treated tumor but also of the contralateral not directly vector-treated tumor. The intratumoral administration of Ad.RSV-F in combination with adenovirus vectors encoding interleukin (IL)-2, IL-12, IL-18, IL-21, or granulocyte macrophage colony-stimulating factor significantly enhanced the antitumor effect on the directly vector-treated tumor and also on the contralateral tumor. The antineoplastic efficacy of this combined treatment was significantly higher than that of the individual treatment components and was associated with the induction of a tumor-specific CTL response and increased infiltration of the tumors by natural killer cells and macrophages. Intratumoral coexpression of RSV-F and IL-21 resulted in the highest tumor growth inhibition and improved survival. Our experimental data indicate that intratumoral expression of RSV-F in combination with cytokines is a promising novel tool for the development of in situ tumor vaccination approaches.


Asunto(s)
Adenoviridae/genética , Vacunas contra el Cáncer/inmunología , Neoplasias Colorrectales/prevención & control , Citocinas/genética , Virus Sincitiales Respiratorios , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Animales , Antineoplásicos/farmacología , Vacunas contra el Cáncer/genética , Fusión Celular , Neoplasias Colorrectales/inmunología , Citocinas/inmunología , Femenino , Vectores Genéticos/genética , Células HT29 , Humanos , Células Asesinas Naturales/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Esplenomegalia/patología , Análisis de Supervivencia , Linfocitos T Citotóxicos/efectos de los fármacos
11.
Hum Gene Ther ; 18(5): 435-50, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17518612

RESUMEN

We evaluated whether the expression of measles virus fusogenic membrane glycoproteins H and F (MV-FMG), encoded by a herpes simplex virus type 1 (HSV-1) amplicon vector, can serve with or without viral oncolysis (G47Delta) and facultative irinotecan chemotherapy, alone or in combination with the monoclonal epidermal growth factor receptor (EGFR) inhibitory antibody cetuximab, as a platform for inducing tumor-specific immune responses against colon cancer. We demonstrated in vitro that MV-FMG expression in murine cells resulted in cell-cell fusion and synergistically enhanced the cytotoxicity of irinotecan alone or in combination with cetuximab. In a bilateral syngeneic subcutaneous MC38 and Colon26 tumor model in C57BL/6 and BALB/c mice we assessed both the effect on directly vector-treated tumors and the effect on contralateral, not directly vector-treated tumors. We demonstrated that the combination of three treatment components with or without cetuximab resulted in the best volume reduction of both directly vector-treated and not directly vector-treated tumors as well as pronounced infiltration of both tumor types with natural killer cells, macrophages, and T cells. T cells of these animals exhibited strong ex vivo cytotoxic activity against the tumor cells, indicating that the antineoplastic effect on untreated tumors was mediated by an antitumor immune response. Preexisting immunity against HSV-1 or measles virus had no detrimental effect on overall treatment efficacy. Our data indicate that MV-FMG expression in combination with viral oncolysis with or without clinically relevant chemotherapy for colon cancer treatment warrants further investigation.


Asunto(s)
Neoplasias del Colon/terapia , Virus del Sarampión/genética , Viroterapia Oncolítica/métodos , Proteínas Virales de Fusión/genética , Animales , Antineoplásicos/uso terapéutico , Camptotecina/análogos & derivados , Camptotecina/uso terapéutico , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/uso terapéutico , Fusión Celular , Línea Celular Tumoral , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Terapia Combinada , Terapia Genética/métodos , Humanos , Irinotecán , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T Citotóxicos/inmunología
12.
Hum Gene Ther ; 18(1): 51-62, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17184155

RESUMEN

The clinical course of sarcoma warrants the development of new therapeutic options, such as gene therapy. However, the lack of coxsackievirus-adenovirus receptor (CAR) on sarcoma cells limits the efficacy of adenovirus type 5 (Ad5)-based gene therapy. In this study we evaluated 20 different adenoviral types and 1 Ad5 vector with RGD-containing fiber for their internalization efficiency in sarcoma cells. We demonstrated that adenovirus types 35, 3, 7, 11, 9, and 22 and Ad5lucRGD virions (ranked in descending order) have significantly higher internalization efficiency in the tested sarcoma cells when compared with Ad5. On the basis of these results we developed a conditionally replication-competent adenoviral vector, Ad5Delta24.Ki.COX, and compared its oncolytic efficacy with that of Ad5/35Delta24.Ki.COX, an Ad5-based vector with the Ad35 fiber shaft and knob domains. Because both vectors differed only in the fiber, we were able to assess whether the adenoviral type with the most efficient internalization resulted also in enhanced treatment efficacy. We evaluated the antineoplastic activity of the oncolytic adenoviral vectors alone or in combination with the expression of measles virus fusogenic membrane glycoproteins and/or ifosfamide. The findings of our xenograft model were as follows: animals that received Ad5/35-based therapy had significantly smaller tumors than animals treated with the homologous Ad5-based vectors. In addition, we demonstrated that the combination of virotherapy, intratumoral expression of fusogenic membrane glycoproteins, and ifosfamide was clearly superior compared with treatment with individual components alone or as combinations of two components. In conclusion, Ad35-based vectors are promising for the treatment of sarcoma.


Asunto(s)
Adenoviridae , Terapia Genética , Vectores Genéticos , Sarcoma/terapia , Transducción Genética , Internalización del Virus , Adenoviridae/genética , Animales , Línea Celular Tumoral , Femenino , Humanos , Virus del Sarampión/genética , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Neoplasias Experimentales/genética , Neoplasias Experimentales/terapia , Oligopéptidos/genética , Sarcoma/genética , Trasplante Heterólogo , Replicación Viral/genética
13.
Cancer Biol Ther ; 6(4): 510-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17374986

RESUMEN

Previously we demonstrated that the expression of fusogenic membrane proteins (FMG) of measles virus (MV-H/F) can synergistically enhance chemotherapy. In this study, we used median-effect analysis to evaluate whether the expression of respiratory syncytial virus (RSV-F), as well as vesicular stomatitis virus (VSV-G) can also synergistically enhance chemotherapy. Furthermore we elucidated by western blot analysis some molecular pathways that might be responsible for this effect. We showed in colorectal cancer cell lines that the expression of MV-H/F, but also of RSV-F, as well as VSV-G can synergistically enhance p53-independent clinically relevant chemotherapy (FOLFOX) over most of the cytotoxic dose range. In a subcutaneous HT-29 colorectal xenograft model, we demonstrated that the administration of replication-deficient adenovirus vectors encoding MV-H/F, RSV-F or VSV-G in combination with FOLFOX significantly enhanced treatment outcome when compared to the treatment with each compound individually. The anti-neoplastic efficacy of RSV-F was somewhat better than that of MV-H/F and both were statistically significantly more efficacious than VSV-G alone or in combination with chemotherapy. Treatment efficacy was further significantly improved when the replication-deficient FMG encoding vectors were trans-complemented for replication with a replication-restricted oncolytic adenovirus to improve tumor transduction efficiency. The combination of FMG expression, chemotherapy and trans-complementing oncolytic vectors resulted in a significantly better treatment efficacy than treatment with its components as single- or double-agent therapy. Our data indicates that FMG expression (i.e., RSV-F and MV-H/F) in combination with chemotherapy and viral oncolysis warrants further investigations.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Colorrectales/terapia , Terapia Genética , Virus Sincitiales Respiratorios/genética , Vesiculovirus/genética , Proteínas Virales de Fusión/genética , Animales , Anexina A5/metabolismo , Apoptosis , Caspasas/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/tratamiento farmacológico , Terapia Combinada , Femenino , Fluorouracilo/uso terapéutico , Humanos , Leucovorina/uso terapéutico , Fusión de Membrana , Ratones , Ratones Desnudos , Mitocondrias/enzimología , Compuestos Organoplatinos/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Int J Mol Med ; 20(5): 673-81, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17912460

RESUMEN

We assessed whether intratumoral expression of the fusogenic membrane protein of vesicular stomatitis virus (VSV-G), encoded by a replication-defective adenovirus vector (Ad.VSV-G), alone or in combination with local coexpression of cytokines induces tumor-specific immune responses in a syngeneic murine colon cancer model. We confirmed in vitro by dye colocalization that transduction of murine cells with Ad.VSV-G induces cell-cell fusion. In a bilateral syngeneic subcutaneous colon cancer model in C57BL/6 and BALB/c mice, we demonstrated that intratumoral injection of Ad.VSV-G leads to a significant growth reduction of the directly vector-treated tumor, but also of the contralateral not directly vector-treated tumor. When compared to monotherapy, the anti-neoplastic efficacy was significantly enhanced when intratumoral Ad.VSV-G administration was combined with adenovirus vectors encoding IL-2, IL-12, IL-18, IL-21, or GM-CSF. The anti-tumor effects of the first three cytokines in combination with VSV-G expression were somewhat greater than those of the latter two. However, the differences did not reach statistical significance. The combination therapy resulted also in a significantly enhanced survival when compared to monotherapy. In addition, we demonstrated that intratumoral expression of VSV-G in combination with the tested cytokines induced a strong tumor-specific cytotoxic T lymphocyte (CTL) response and infiltration of tumors with macrophages. The effects of the combination therapy were clearly greater than those of the monotherapy. Our experimental data indicate that intratumoral expression of VSV-G, particularly in combination with cytokines, is a promising novel tool for the development of in situ tumor vaccination approaches.


Asunto(s)
Adenoviridae/metabolismo , Citocinas/metabolismo , Vectores Genéticos/metabolismo , Inmunoterapia , Glicoproteínas de Membrana/metabolismo , Neoplasias/inmunología , Neoplasias/terapia , Proteínas del Envoltorio Viral/metabolismo , Animales , Antineoplásicos/farmacología , Vacunas contra el Cáncer , Fusión Celular , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Colon/efectos de los fármacos , Colon/patología , Femenino , Células Gigantes/efectos de los fármacos , Células Gigantes/patología , Humanos , Macrófagos/citología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Esplenomegalia/patología , Análisis de Supervivencia , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
15.
World J Gastroenterol ; 13(22): 3063-70, 2007 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-17589921

RESUMEN

AIM: To evaluate whether intratumoral expression of measles virus fusogenic membrane glycoproteins H and F (MV-FMG), encoded by an adenovirus vector Ad.MV-H/F, alone or in combination with local coexpression of cytokines (IL-2, IL-12, IL-18, IL-21 or GM-CSF), can serve as a platform for inducing tumor-specific immune responses in colon cancer. METHODS: We used confocal laser scanning microscopy and flow cytometry to analyze cell-cell fusion after expression of MV-FMG by dye colocalization. In a syngeneic bilateral subcutaneous MC38 and Colon26 colon cancer model in C57BL/6 and BALB/c mice, we assessed the effect on both the directly vector-treated tumor as well as the contralateral, not directly vector-treated tumor. We assessed the induction of a tumor-specific cytotoxic T lymphocyte (CTL) response with a lactate dehydrogenase (LDH) release assay. RESULTS: We demonstrated in vitro that transduction of MC38 and Colon26 cells with Ad.MV-H/F resulted in dye colocalization, indicative of cell-cell fusion. In addition, in the syngeneic bilateral tumor model we demonstrated a significant regression of the directly vector-inoculated tumor upon intratumoral expression of MV-FMG alone or in combination with the tested cytokines. We observed the highest anti-neoplastic efficacy with MV-FMG and IL-21 coexpression. The degree of tumor regression of the not directly vector-treated tumor correlated with the anti-neoplastic response of the directly vector-treated tumor. This regression was mediated by a tumor-specific CTL response. CONCLUSION: Our data indicate that intratumoral expression of measles virus fusogenic membrane glycoproteins is a promising tool both for direct tumor treatment as well as for tumor vaccination approaches that can be further enhanced by cytokine coexpression.


Asunto(s)
Adenocarcinoma/prevención & control , Adenoviridae/genética , Vacunas contra el Cáncer/uso terapéutico , Neoplasias del Colon/prevención & control , Citocinas/metabolismo , Vectores Genéticos/uso terapéutico , Proteínas Virales de Fusión/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Fusión Celular , Línea Celular Tumoral , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Citocinas/genética , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Regulación Viral de la Expresión Génica , Células HT29 , Humanos , Inmunoterapia/métodos , Interleucinas/genética , Interleucinas/metabolismo , Macrófagos/patología , Virus del Sarampión/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/patología , Proteínas Virales de Fusión/genética
16.
Mol Cancer Ther ; 5(8): 2013-22, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16928822

RESUMEN

Pancreatic cancer has a poor prognosis with an annual mortality rate close to the annual incidence rate. We evaluated whether the expression of measles virus fusogenic membrane glycoproteins (FMG) H and F will enhance chemotherapy. Using Chou-Talalay analysis, we showed in vitro in pancreatic cancer cells that the expression of FMG often synergistically enhances clinically relevant chemotherapy. Furthermore, cell fusion in combination with chemotherapy resulted in strongly enhanced Annexin V binding, an early marker for apoptosis, when compared with single treatment. We showed in an i.p. and s.c. pancreatic xenograft model that the administration of a replication-defective adenoviral vector Ad.H/F encoding tumor-restricted FMG in combination with gemcitabine significantly enhanced treatment outcome when compared with treatment with each compound individually. To improve tumor transduction efficiency, the Ad.H/F vector was also transcomplemented with an oncolytic replication-restricted adenovirus (Ad.COX*MK), resulting in significantly improved treatment efficacy. We assessed treatment efficacy by survival analysis or measuring growth, respectively. In the i.p. model, on day 120, three of eight animals treated with this novel triple therapy consisting of Ad.H/F, gemcitabine, and Ad.COX*MK were alive and tumor free. Treatment with Ad.H/F and Ad.COX*MK resulted in one long-term survivor. In all other treatment groups, there were no long-term survivors. The significantly improved therapeutic outcome of animals receiving the triple therapy was attributed to multiple factors, including most likely improved FMG expression throughout the tumor and enhanced sensitivity of the tumor cells to gemcitabine by adenoviral gene products but also FMG expression. Qualitatively similar results were obtained in a s.c. pancreatic xenograft model.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de la Fusión de la Membrana/genética , Glicoproteínas de Membrana/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Adenoviridae/genética , Animales , Apoptosis , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacología , Femenino , Vectores Genéticos , Humanos , Proteínas de la Fusión de la Membrana/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Viroterapia Oncolítica/métodos , Neoplasias Pancreáticas/mortalidad , Tasa de Supervivencia , Transducción Genética , Células Tumorales Cultivadas , Gemcitabina
17.
Mol Cancer Ther ; 5(2): 374-81, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16505112

RESUMEN

In our current study, we developed oncolytic adenoviruses which preferentially lyse pancreatic and colon cancer cells by replacing viral E1 and/or E4 promoter with the tumor/tissue-specific promoters, cyclooxygenase-2 (COX-2), midkine (MK), or the cell cycle-dependent promoter, E2F1. We generated three sets of recombinant adenoviral vectors. In the first set, only the native E1A promoter was replaced by the COX-2, MK, or E2F1 promoter, respectively. In the second set, the viral E4 promoter was substituted by these heterologous promoters and the viral E1A promoter was substituted by the ubiquitously active cytomegalovirus-IE promoter. In the third set, we substituted the viral E1A and E4 promoters with the COX-2, MK, or E2F1 promoter, respectively. In our system, transcriptional targeting of solitary viral E1A resulted in 50% enhanced restricted vector replication when compared with an unrestricted replication-competent adenovirus. Furthermore, a targeted expression of the viral E1A gene products had a greater effect on restricted adenoviral replication than that of the E4 region. With our vectors, Ad.COX.MK and Ad.MK.COX, using two different heterologous promoters to control E1A and E4 expression, we showed enhanced viral replication specificity when compared with Ad.COX.COX or Ad.MK.MK, respectively. In a s.c. xenograft tumor model, there was no significant difference in the antineoplastic efficacy of the double heterologous promoter-controlled vectors when compared with our unrestricted replication-competent control adenovirus or vectors with only E1A transcriptionally driven by a heterologous promoter.


Asunto(s)
Adenoviridae/fisiología , Neoplasias Colorrectales/terapia , Genes Relacionados con las Neoplasias , Viroterapia Oncolítica , Neoplasias Pancreáticas/terapia , Regiones Promotoras Genéticas , Replicación Viral/genética , Adenoviridae/genética , Neoplasias Colorrectales/genética , Ciclooxigenasa 2/genética , Factor de Transcripción E2F1/genética , Regulación Neoplásica de la Expresión Génica , Regulación Viral de la Expresión Génica , Vectores Genéticos/genética , Vectores Genéticos/fisiología , Humanos , Proteínas de la Membrana/genética , Neoplasias Pancreáticas/genética , Transcripción Genética , Células Tumorales Cultivadas
18.
BMC Biotechnol ; 6: 36, 2006 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-16887042

RESUMEN

BACKGROUND: Oncolytic adenoviruses are promising agents for the multimodal treatment of cancer. However, tumor-selectivity is crucial for their applicability in patients. Recent studies by several groups demonstrated that oncolytic adenoviruses with tumor-/tissue-specific expression of the E1 and E4 genes, which are pivotal for adenoviral replication, have a specificity profile that is superior to viruses that solely target the expression of E1 or E4 genes. Presently the E1 and E4 regions are modified in a time consuming sequential fashion. RESULTS: Based on the widely used adenoviral cloning system AdEasy we generated a novel transfer vector that allows efficient and rapid generation of conditionally replication-competent adenovirus type 5 based vectors with the viral E1 and E4 genes under the transcriptional control of heterologous promoters. For insertion of the promoters of interest our transfer vector has two unique multiple cloning sites. Additionally, our shuttle plasmid allows encoding of a transgene within the E1A transcription unit. The modifications, including E1 mutations, are introduced into the adenoviral genome by a single homologous recombination step in Escherichia coli. Subsequently infectious viruses are rescued from plasmids. As a proof-of-concept we generated two conditionally replication-competent adenoviruses Ad.Ki x COX and Ad.COX x Ki with the promoters of the Ki-67 protein and the cyclooxygenase-2 (COX-2) driving E1 and E4 and vice versa. CONCLUSION: We demonstrated with our cloning system efficient generation of double heterologous promoter controlled oncolytic adenoviral vectors by a single homologous recombination step in bacteria. The generated viruses showed preferential replication in tumor cells and in a subcutaneous HT-29 colon cancer xenograft model the viruses demonstrated significant oncolytic activity comparable with dl327.


Asunto(s)
Adenovirus Humanos/genética , Escherichia coli/genética , Ingeniería Genética/métodos , Vectores Genéticos/genética , Virus Oncolíticos/genética , Regiones Promotoras Genéticas/genética , Recombinación Genética/genética , Proteínas E1 de Adenovirus/genética , Proteínas E1 de Adenovirus/metabolismo , Proteínas E4 de Adenovirus/genética , Proteínas E4 de Adenovirus/metabolismo , Adenovirus Humanos/fisiología , Animales , Niño , Ciclooxigenasa 2/genética , Efecto Citopatogénico Viral , ADN Recombinante/genética , Células HT29 , Humanos , Antígeno Ki-67/genética , Ratones , Ratones Endogámicos BALB C , Mutación/genética , Virus Oncolíticos/fisiología , Especificidad de Órganos , Replicación Viral , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Retrovirology ; 2: 2, 2005 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-15656908

RESUMEN

BACKGROUND: The antibacterial activity of host defense peptides (HDP) is largely mediated by permeabilization of bacterial membranes. The lipid membrane of enveloped viruses might also be a target of antimicrobial peptides. Therefore, we screened a panel of naturally occurring HDPs representing different classes for inhibition of early, Env-independent steps in the HIV replication cycle. A lentiviral vector-based screening assay was used to determine the inhibitory effect of HDPs on early steps in the replication cycle and on cell metabolism. RESULTS: Human LL37 and porcine Protegrin-1 specifically reduced lentiviral vector infectivity, whereas the reduction of luciferase activities observed at high concentrations of the other HDPs is primarily due to modulation of cellular activity and/ or cytotoxicity rather than antiviral activity. A retroviral vector was inhibited by LL37 and Protegrin-1 to similar extent, while no specific inhibition of adenoviral vector mediated gene transfer was observed. Specific inhibitory effects of Protegrin-1 were confirmed for wild type HIV-1. CONCLUSION: Although Protegrin-1 apparently inhibits an early step in the HIV-replication cycle, cytotoxic effects might limit its use as an antiviral agent unless the specificity for the virus can be improved.


Asunto(s)
Antiinfecciosos/farmacología , Péptidos Catiónicos Antimicrobianos/farmacología , VIH-1/efectos de los fármacos , Lentivirus/efectos de los fármacos , Proteínas/farmacología , Replicación Viral/efectos de los fármacos , Animales , Antiinfecciosos/toxicidad , Péptidos Catiónicos Antimicrobianos/toxicidad , Catelicidinas , Línea Celular , Vectores Genéticos , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/crecimiento & desarrollo , Cocos Grampositivos/efectos de los fármacos , Cocos Grampositivos/crecimiento & desarrollo , VIH-1/fisiología , Humanos , Lentivirus/fisiología , Luciferasas/genética , Luciferasas/metabolismo , Péptidos/farmacología , Péptidos/toxicidad , Proteínas/toxicidad , Transducción Genética
20.
Hum Gene Ther ; 13(1): 101-12, 2002 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-11779414

RESUMEN

Since human adenoviruses replicate only in human cells, toxicology studies with adenoviral vectors are hampered by the lack of a permissive nonhuman host. Before a replication-competent adenoviral vector expressing HSV-tk (Ad.OW34) can be used in clinical studies for intratumoral injections in patients with cutaneous lesions of head and neck cancer or intralesional injection for in situ vaccination strategy in advanced metastatic melanoma patients, risks have to be estimated in animal studies. In an attempt to assess potential toxicology, dissemination, persistence and shedding, we injected Ad.OW34 subcutaneously into cotton rats. (Sigmodon hispidus), which are considered a semi-permissive host for human adenoviruses. The animals underwent one or two subcutaneous injection cycles with 2.3 x 10(12) viral particles/kg each or a single course with 6.9 x 10(13) viral particles/kg and were analyzed at defined time points for histopathological changes in the brain, heart, lungs, spleen, liver, kidneys, ovaries, and skin. Additionally, these tissues as well as urine, feces, mouth, and skin swabs were analyzed at multiple time points by real-time quantitative polymerase chain reaction for the presence of vector sequences. The only significant treatment-related histopathologic finding was dermatitis with mild acanthosis at the site of vector injection. All other tissues evaluated were within normal limits or showed changes that were most likely incidental or spontaneous in nature. Vector sequences were detected in the skin at the injection site and to a lesser extent in the liver, spleen, and lungs. In addition, small amounts of vector DNA were detected in the ovaries. The vector sequences were rapidly cleared and the absence of viral sequences in the excreta and swabs of the majority of animals suggest that there was no significant replication of the vector in this host. The administration of Ad.OW34 was also associated with mild hyperamylasemia, lymphocytosis, and granulocytosis; however, we did not observe any clinical signs of illness or death in the experimental animals over the course of the study.


Asunto(s)
Adenovirus Humanos/genética , Vectores Genéticos/administración & dosificación , Herpes Simple/enzimología , Sigmodontinae/virología , Timidina Quinasa/genética , Animales , Femenino , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Humanos , Inyecciones Subcutáneas , Recuento de Leucocitos , Microscopía Electrónica , Reacción en Cadena de la Polimerasa , Distribución Tisular , Replicación Viral , Esparcimiento de Virus
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA