Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
J Virol ; 91(11)2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28331098

RESUMEN

There are currently no approved therapeutics or vaccines to treat or protect against the severe hemorrhagic fever and death caused by Ebola virus (EBOV). Ebola virus-like particles (EBOV VLPs) consisting of the matrix protein VP40, the glycoprotein (GP), and the nucleoprotein (NP) are highly immunogenic and protective in nonhuman primates against Ebola virus disease (EVD). We have constructed a modified vaccinia virus Ankara-Bavarian Nordic (MVA-BN) recombinant coexpressing VP40 and GP of EBOV Mayinga and the NP of Taï Forest virus (TAFV) (MVA-BN-EBOV-VLP) to launch noninfectious EBOV VLPs as a second vaccine modality in the MVA-BN-EBOV-VLP-vaccinated organism. Human cells infected with either MVA-BN-EBOV-VLP or MVA-BN-EBOV-GP showed comparable GP expression levels and transport of complex N-glycosylated GP to the cell surface. Human cells infected with MVA-BN-EBOV-VLP produced large amounts of EBOV VLPs that were decorated with GP spikes but excluded the poxviral membrane protein B5, thus resembling authentic EBOV particles. The heterologous TAFV NP enhanced EBOV VP40-driven VLP formation with efficiency similar to that of the homologous EBOV NP in a transient-expression assay, and both NPs were incorporated into EBOV VLPs. EBOV GP-specific CD8 T cell responses were comparable between MVA-BN-EBOV-VLP- and MVA-BN-EBOV-GP-immunized mice. The levels of EBOV GP-specific neutralizing and binding antibodies, as well as GP-specific IgG1/IgG2a ratios induced by the two constructs, in mice were also similar, raising the question whether the quality rather than the quantity of the GP-specific antibody response might be altered by an EBOV VLP-generating MVA recombinant.IMPORTANCE The recent outbreak of Ebola virus (EBOV), claiming more than 11,000 lives, has underscored the need to advance the development of safe and effective filovirus vaccines. Virus-like particles (VLPs), as well as recombinant viral vectors, have proved to be promising vaccine candidates. Modified vaccinia virus Ankara-Bavarian Nordic (MVA-BN) is a safe and immunogenic vaccine vector with a large capacity to accommodate multiple foreign genes. In this study, we combined the advantages of VLPs and the MVA platform by generating a recombinant MVA-BN-EBOV-VLP that would produce noninfectious EBOV VLPs in the vaccinated individual. Our results show that human cells infected with MVA-BN-EBOV-VLP indeed formed and released EBOV VLPs, thus producing a highly authentic immunogen. MVA-BN-EBOV-VLP efficiently induced EBOV-specific humoral and cellular immune responses in vaccinated mice. These results are the basis for future advancements, e.g., by including antigens from various filoviral species to develop multivalent VLP-producing MVA-based filovirus vaccines.


Asunto(s)
Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/aislamiento & purificación , Glicoproteínas/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Virus Vaccinia/genética , Virión/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Virus del Ébola/genética , Ebolavirus/genética , Ebolavirus/inmunología , Ebolavirus/fisiología , Glicoproteínas/genética , Humanos , Inmunoglobulina G/sangre , Ratones , Nucleoproteínas/genética , Nucleoproteínas/inmunología , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/inmunología , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/inmunología , Virión/fisiología
2.
Cancer Immunol Immunother ; 61(1): 19-29, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21822917

RESUMEN

MVA-BN®-HER2 is a new candidate immunotherapy designed for the treatment of HER-2-positive breast cancer. Here, we demonstrate that a single treatment with MVA-BN®-HER2 exerts potent anti-tumor efficacy in a murine model of experimental pulmonary metastasis. This anti-tumor efficacy occurred despite a strong tumor-mediated immunosuppressive environment characterized by a high frequency of regulatory T cells (T(reg)) in the lungs of tumor-bearing mice. Immunogenicity studies showed that treatment with MVA-BN®-HER2 induced strongly Th1-dominated HER-2-specific antibody and T-cell responses. MVA-BN®-HER2-induced anti-tumor activity was characterized by an increased infiltration of lungs with highly activated, HER-2-specific, CD8+CD11c+ T cells accompanied by a decrease in the frequency of T(reg) cells in the lung, resulting in a significantly increased ratio of effector T cells to T(reg) cells. In contrast, administration of HER2 protein formulated in Complete Freund's Adjuvant (CFA) induced a strongly Th2-biased immune response to HER-2. However, this did not lead to significant infiltration of the tumor-bearing lungs by CD8+ T cells or the decrease in the frequency of T(reg) cells nor did it result in anti-tumor efficacy. In vivo depletion of CD8+ cells confirmed that CD8 T cells were required for the anti-tumor activity of MVA-BN®-HER2. Furthermore, depletion of CD4+ or CD25+ cells demonstrated that tumor-induced T(reg) cells promoted tumor growth and that CD4 effector cells also contribute to MVA-BN®-HER2-mediated anti-tumor efficacy. Taken together, our data demonstrate that treatment with MVA-BN®-HER2 controls tumor growth through mechanisms including the induction of Th1-biased HER-2-specific immune responses and the control of tumor-mediated immunosuppression.


Asunto(s)
Adenocarcinoma/terapia , Subgrupos de Linfocitos B/inmunología , Vacunas contra el Cáncer/farmacología , Neoplasias del Colon/terapia , Inmunoterapia/métodos , Receptor ErbB-2/inmunología , Linfocitos T Reguladores/inmunología , Adenocarcinoma/inmunología , Adenocarcinoma/patología , Animales , Subgrupos de Linfocitos B/enzimología , Subgrupos de Linfocitos B/patología , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunofenotipificación , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Linfocitos T Reguladores/enzimología , Linfocitos T Reguladores/patología
3.
J Virol ; 84(19): 9907-19, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20668072

RESUMEN

Modified vaccinia virus Ankara (MVA) has a highly restricted host range in cell culture and is apathogenic in vivo. MVA was derived from the parental chorioallantois vaccinia virus Ankara (CVA) by more than 570 passages in chicken embryo fibroblast (CEF) cells. During CEF cell passaging, six major deletions comprising 24,668 nucleotides occurred in the CVA genome. We have cloned both the MVA and the parental CVA genome as bacterial artificial chromosomes (BACs) and have sequentially introduced the six major MVA deletions into the cloned CVA genome. Reconstituted mutant CVA viruses containing up to six major MVA deletions showed no detectable replication restriction in 12 of 14 mammalian cell lines tested; the exceptions were rabbit cell lines RK13 and SIRC. In mice, CVA mutants with up to three deletions showed slightly enhanced virulence, suggesting that gene deletion in replicating vaccinia virus (VACV) can result in gain of fitness in vivo. CVA mutants containing five or all six deletions were still pathogenic, with a moderate degree of attenuation. Deletion V was mainly responsible for the attenuated phenotype of these mutants. In conclusion, loss or truncation of all 31 open reading frames in the six major deletions is not sufficient to reproduce the specific MVA phenotype of strong attenuation and highly restricted host range. Mutations in viral genes outside or in association with the six major deletions appear to contribute significantly to this phenotype. Host range restriction and avirulence of MVA are most likely a cooperative effect of gene deletions and mutations involving the major deletions.


Asunto(s)
Eliminación de Gen , Genoma Viral , Virus Vaccinia/genética , Virus Vaccinia/patogenicidad , Animales , Línea Celular , Embrión de Pollo , Cromosomas Artificiales Bacterianos/genética , Efecto Citopatogénico Viral , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Fenotipo , Conejos , Recombinación Genética , Vaccinia/etiología , Vaccinia/virología , Virus Vaccinia/fisiología , Virulencia/genética , Cultivo de Virus , Replicación Viral
4.
Eur J Pharm Biopharm ; 129: 215-221, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29870747

RESUMEN

The stability profile of a vaccine has important implications for storage, cold chain management and field deployment. The heterologous prime-boost Janssen Ebola vaccine regimen demonstrated an acceptable safety profile and durability of Ebola-specific immune responses in Phase I studies in healthy adults. Potency (infectious titre) of both components of the Ad26.ZEBOV/MVA-BN-Filo regimen were assessed using qPCR-based potency assay and flow cytometry during real-time and accelerated stability studies, conducted between -80 °C and 25 °C. Additionally, vaccine potency was assessed following agitation, temperature cycling, freeze-thawing and while in the injection system. Ad26.ZEBOV remained stable for 24 months when frozen and at 2-8 °C; MVA-BN-Filo remained stable for 24 months frozen and 12 months at 2-8 °C. Potency of both vaccines was maintained during temperature cycling, agitation and freeze-thawing. When exposed to high temperatures (up to 40 °C) in a syringe/needle both vaccines remained stable for at least 6 h. The vaccines are expected to maintain potency for 36 months when frozen (based on extrapolation of observed stability). The findings of this study indicate that the stability of the Ad26.ZEBOV/MVA-BN-Filo is likely suitable for field deployment in regions at risk of Ebola outbreaks, where cold chain maintenance is challenging owing to infrastructure and resource limitations.


Asunto(s)
Brotes de Enfermedades/prevención & control , Composición de Medicamentos/métodos , Vacunas contra el Virus del Ébola/farmacología , Fiebre Hemorrágica Ebola/prevención & control , Antígenos Virales/química , Antígenos Virales/inmunología , Estabilidad de Medicamentos , Almacenaje de Medicamentos , Vacunas contra el Virus del Ébola/química , Vacunas contra el Virus del Ébola/inmunología , Vacunas contra el Virus del Ébola/uso terapéutico , Congelación , Fiebre Hemorrágica Ebola/epidemiología , Humanos , Temperatura
5.
PLoS One ; 13(2): e0192312, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29462200

RESUMEN

The search for a universal filovirus vaccine that provides protection against multiple filovirus species has been prompted by sporadic but highly lethal outbreaks of Ebolavirus and Marburgvirus infections. A good prophylactic vaccine should be able to provide protection to all known filovirus species and as an upside potentially protect from newly emerging virus strains. We investigated the immunogenicity and protection elicited by multivalent vaccines expressing glycoproteins (GP) from Ebola virus (EBOV), Sudan virus (SUDV), Taï Forest virus (TAFV) and Marburg virus (MARV). Immune responses against filovirus GP have been associated with protection from disease. The GP antigens were expressed by adenovirus serotypes 26 and 35 (Ad26 and Ad35) and modified Vaccinia virus Ankara (MVA) vectors, all selected for their strong immunogenicity and good safety profile. Using fully lethal NHP intramuscular challenge models, we assessed different vaccination regimens for immunogenicity and protection from filovirus disease. Heterologous multivalent Ad26-Ad35 prime-boost vaccination regimens could give full protection against MARV (range 75%-100% protection) and EBOV (range 50% to 100%) challenge, and partial protection (75%) against SUDV challenge. Heterologous multivalent Ad26-MVA prime-boost immunization gave full protection against EBOV challenge in a small cohort study. The use of such multivalent vaccines did not show overt immune interference in comparison with monovalent vaccines. Multivalent vaccines induced GP-specific antibody responses and cellular IFNγ responses to each GP expressed by the vaccine, and cross-reactivity to TAFV GP was detected in a trivalent vaccine expressing GP from EBOV, SUDV and MARV. In the EBOV challenge studies, higher humoral EBOV GP-specific immune responses (p = 0.0004) were associated with survival from EBOV challenge and less so for cellular immune responses (p = 0.0320). These results demonstrate that it is feasible to generate a multivalent filovirus vaccine that can protect against lethal infection by multiple members of the filovirus family.


Asunto(s)
Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Enfermedad del Virus de Marburg/prevención & control , Marburgvirus/inmunología , Vacunas Virales/inmunología , Animales , Femenino , Macaca fascicularis , Masculino
7.
Clin Cancer Res ; 23(22): 6833-6845, 2017 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-28855356

RESUMEN

Purpose: The transcription factor brachyury has been shown in preclinical studies to be a driver of the epithelial-to-mesenchymal transition (EMT) and resistance to therapy of human tumor cells. This study describes the characterization of a Modified Vaccinia Ankara (MVA) vector-based vaccine expressing the transgenes for brachyury and three human costimulatory molecules (B7.1, ICAM-1, and LFA-3, designated TRICOM) and a phase I study with this vaccine.Experimental Design: Human dendritic cells (DC) were infected with MVA-brachyury-TRICOM to define their ability to activate brachyury-specific T cells. A dose-escalation phase I study (NCT02179515) was conducted in advanced cancer patients (n = 38) to define safety and to identify brachyury-specific T-cell responses.Results: MVA-brachyury-TRICOM-infected human DCs activated CD8+ and CD4+ T cells specific against the self-antigen brachyury in vitro No dose-limiting toxicities were observed due to vaccine in cancer patients at any of the three dose levels. One transient grade 3 adverse event (AE) possibly related to vaccine (diarrhea) resolved without intervention and did not recur with subsequent vaccine. All other AEs related to vaccine were transient and ≤grade 2. Brachyury-specific T-cell responses were observed at all dose levels and in most patients.Conclusions: The MVA-brachyury-TRICOM vaccine directed against a transcription factor known to mediate EMT can be administered safely in patients with advanced cancer and can activate brachyury-specific T cells in vitro and in patients. Further studies of this vaccine in combination therapies are warranted and planned. Clin Cancer Res; 23(22); 6833-45. ©2017 AACR.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Proteínas Fetales/inmunología , Vectores Genéticos , Neoplasias/inmunología , Neoplasias/terapia , Proteínas de Dominio T Box/inmunología , Virus Vaccinia , Adulto , Anciano , Anciano de 80 o más Años , Antígeno B7-1/genética , Biomarcadores de Tumor , Antígenos CD58/genética , Vacunas contra el Cáncer/genética , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Proteínas Fetales/genética , Vectores Genéticos/genética , Humanos , Molécula 1 de Adhesión Intercelular/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Persona de Mediana Edad , Neoplasias/genética , Neoplasias/mortalidad , Proteínas de Dominio T Box/genética , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transgenes , Resultado del Tratamiento , Virus Vaccinia/genética
8.
Oncogene ; 24(10): 1711-7, 2005 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-15674340

RESUMEN

Latent membrane protein 1 (LMP1), an oncoprotein encoded by Epstein-Barr virus (EBV), is an integral membrane protein, which acts like a constitutively active receptor. LMP1 is critical for some facet of EBV's induction and maintenance of proliferation of infected B cells. It, in part, mimics signaling by the CD40 receptor and has been implicated in regulating proliferation, survival, or both properties of EBV-infected cells. We established a conditional LMP1 allele in the context of the intact EBV genome to define the immediate-early cellular target genes regulated by LMP1 in order to assess its contributions to infected human B cells. The functional analysis of this conditional system indicated that LMP1 specifically induces mitogenic B-cell activation through c-myc and Jun/AP1 family members and confirms its direct role in upregulating expression of multiple genes with opposing activities involved in cell survival. LMP1's signals were found to be essential for the G1/S transition in human B cells; cells lacking LMP1's signals are cell cycle arrested and survive quiescently. LMP1's activities are therefore not required to maintain survival in nonproliferating cells. LMP1 does induce both pro- and antiapoptotic genes whose balance seems to permit survival during LMP1's induction and maintenance of proliferation.


Asunto(s)
Apoptosis , Linfocitos B/inmunología , Proliferación Celular , Proteínas de la Matriz Viral/fisiología , Linfocitos B/virología , Fase G1 , Regulación de la Expresión Génica , Humanos , Activación de Linfocitos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fase S , Transducción de Señal
9.
Cancer Res ; 63(11): 2982-9, 2003 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-12782607

RESUMEN

The EBV latent membrane protein 1 (LMP1) is an integral membrane protein that acts like a constitutively activated receptor. LMP1 interacts with members of the tumor necrosis factor receptor-associated factor family, as well as with tumor necrosis factor receptor-associated death domain, resulting in induction of nuclear factor-kappaB, the p38 mitogen-activated protein kinase pathway, and the c-Jun NH(2)-terminal kinase activator protein 1-signaling cascade. The binding of Janus kinase 3 results in activation of signal transducers and activators of transcription. The domain structure of LMP1 has been mapped extensively, but the quantitative contribution of distinct LMP1 domains to the efficiency of B-cell proliferation by EBV has not been determined. On the basis of the maxi-EBV system, which allows us to introduce and study mutations in the context of the complete EBV genome, a panel of 10 EBV mutants with alterations in the LMP1 gene locus was established. The mutant EBVs were tested for their efficiency to induce and maintain proliferation of clonal B-cell lines in vitro. Surprisingly and with reduced frequency, EBV mutants which deleted LMP1's COOH terminus, transmembrane domains, or the entire open reading frame were able to generate proliferating B-cell clones that were dependent on the presence of human fibroblast feeder cells. A B-cell clone carrying the LMP1-null mutant EBV genome was also analyzed for oncogenicity in severe combined immunodeficiency mice. Our results demonstrate that LMP1 is critical but not mandatory for the generation of proliferating B cells in vitro. LMP1 functions greatly contribute to EBV's transformation potential and appear essential for its oncogenicity in severe combined immunodeficiency mice.


Asunto(s)
Linfocitos B/virología , Transformación Celular Viral/genética , Herpesvirus Humano 4/genética , Proteínas de la Matriz Viral/fisiología , Alelos , Animales , Linfocitos B/patología , Linfoma de Burkitt/patología , Linfoma de Burkitt/virología , División Celular/genética , División Celular/fisiología , Humanos , Ratones , Ratones SCID , Mutación , Células Tumorales Cultivadas , Proteínas de la Matriz Viral/genética
10.
Oncotarget ; 6(29): 28194-210, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26317648

RESUMEN

Several transcription factors play a role in the alteration of gene expression that occurs during cancer metastasis. Twist expression has been shown to be associated with the hallmarks of the metastatic process, as well as poor prognosis and drug resistance in many tumor types. However, primarily due to their location within the cell and the lack of a hydrophobic groove required for drug attachment, transcription factors such as Twist are difficult to target with conventional therapies. An alternative therapeutic strategy is a vaccine comprised of a Modified vaccinia Ankara (MVA), incorporating the Twist transgene and a TRIad of COstimulatory Molecules (B7-1, ICAM-1, LFA-3; TRICOM). Here we characterize an MVA-TWIST/TRICOM vaccine that induced both CD4+ and CD8+ Twist-specific T-cell responses in vivo. In addition, administration of this vaccine reduced both the primary tumor growth and metastasis in the 4T1 model of metastatic breast cancer. In the TRAMP transgenic model of spontaneous prostate cancer, MVA-TWIST/TRICOM alone significantly improved survival, and when combined with the androgen receptor antagonist enzalutamide, the vaccine further improved survival. These studies thus provide a rationale for the use of active immunotherapy targeting transcription factors involved in the metastatic process and for the combination of cancer vaccines with androgen deprivation.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Neoplasias Mamarias Animales/inmunología , Neoplasias de la Próstata/inmunología , Proteína 1 Relacionada con Twist/inmunología , Virus Vaccinia/inmunología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/metabolismo , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica/inmunología , Masculino , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/prevención & control , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Metástasis de la Neoplasia , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/prevención & control , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Supervivencia , Resultado del Tratamiento , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Carga Tumoral/inmunología , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo , Vacunación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/metabolismo , Virus Vaccinia/genética , Virus Vaccinia/metabolismo
11.
PLoS One ; 8(8): e73511, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23951355

RESUMEN

Modified vaccinia virus Ankara (MVA) has been shown to be suitable for the generation of experimental vaccines against cancer and infectious diseases, eliciting strong humoral and cellular immune responses. In viral vectored vaccines, strong recombinant antigen expression and timing of expression influence the quantity and quality of the immune response. Screening of synthetic and native poxvirus promoters for strong protein expression in vitro and potent immune responses in vivo led to the identification of the MVA13.5L promoter, a unique and novel naturally occurring tandem promoter in MVA composed of two 44 nucleotide long repeated motifs, each containing an early promoter element. The MVA13.5L gene is highly conserved across orthopoxviruses, yet its function is unknown. The unique structure of its promoter is not found for any other gene in the MVA genome and is also conserved in other orthopoxviruses. Comparison of the MVA13.5L promoter activity with synthetic poxviral promoters revealed that the MVA13.5L promoter produced higher levels of protein early during infection in HeLa cells and particularly in MDBK cells, a cell line in which MVA replication stops at an early stage before the expression of late genes. Finally, a recombinant antigen expressed under the control of this novel promoter induced high antibody titers and increased CD8 T cell responses in homologous prime-boost immunization compared to commonly used promoters. In particular, the recombinant antigen specific CD8 T cell responses dominated over the immunodominant B8R vector-specific responses after three vaccinations and even more during the memory phase. These results have identified the native MVA13.5L promoter as a new potent promoter for use in MVA vectored preventive and therapeutic vaccines.


Asunto(s)
Vectores Genéticos/genética , Vectores Genéticos/inmunología , Regiones Promotoras Genéticas , Virus Vaccinia/genética , Virus Vaccinia/inmunología , Animales , Anticuerpos Antivirales/inmunología , Antígenos/inmunología , Secuencia de Bases , Linfocitos T CD8-positivos/inmunología , Línea Celular , Embrión de Pollo , Femenino , Expresión Génica , Orden Génico , Vectores Genéticos/administración & dosificación , Humanos , Inmunidad Celular , Inmunidad Humoral , Memoria Inmunológica , Ratones , Datos de Secuencia Molecular , Vacunas Virales/genética , Vacunas Virales/inmunología
12.
Cancer Res ; 71(15): 5235-44, 2011 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-21670078

RESUMEN

MVA-BN-PRO (BN ImmunoTherapeutics) is a candidate immunotherapy product for the treatment of prostate cancer. It encodes 2 tumor-associated antigens, prostate-specific antigen (PSA), and prostatic acid phosphatase (PAP), and is derived from the highly attenuated modified vaccinia Ankara (MVA) virus stock known as MVA-BN. Past work has shown that the immunogenicity of antigens can be improved by targeting their localization to exosomes, which are small, 50- to 100-nm diameter vesicles secreted by most cell types. Exosome targeting is achieved by fusing the antigen to the C1C2 domain of the lactadherin protein. To test whether exosome targeting would improve the immunogenicity of PSA and PAP, 2 additional versions of MVA-BN-PRO were produced, targeting either PSA (MVA-BN-PSA-C1C2) or PAP (MVA-BN-PAP-C1C2) to exosomes, while leaving the second transgene untargeted. Treatment of mice with MVA-BN-PAP-C1C2 led to a striking increase in the immune response against PAP. Anti-PAP antibody titers developed more rapidly and reached levels that were 10- to 100-fold higher than those for mice treated with MVA-BN-PRO. Furthermore, treatment with MVA-BN-PAP-C1C2 increased the frequency of PAP-specific T cells 5-fold compared with mice treated with MVA-BN-PRO. These improvements translated into a greater frequency of tumor rejection in a PAP-expressing solid tumor model. Likewise, treatment with MVA-BN-PSA-C1C2 increased the antigenicity of PSA compared with treatment with MVA-BN-PRO and resulted in a trend of improved antitumor efficacy in a PSA-expressing tumor model. These experiments confirm that targeting antigen localization to exosomes is a viable approach for improving the therapeutic potential of MVA-BN-PRO in humans.


Asunto(s)
Adenocarcinoma/inmunología , Anticuerpos Antineoplásicos/biosíntesis , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Exosomas/inmunología , Inmunoterapia Activa/métodos , Neoplasias de la Próstata/inmunología , Proteínas Tirosina Fosfatasas/inmunología , Fosfatasa Ácida , Adenocarcinoma/patología , Adenocarcinoma/terapia , Animales , Antígenos de Superficie/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/uso terapéutico , Sistemas de Liberación de Medicamentos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Proteínas de la Leche/inmunología , Proteínas de la Leche/farmacocinética , Antígeno Prostático Específico/administración & dosificación , Antígeno Prostático Específico/inmunología , Neoplasias de la Próstata/patología , Neoplasias de la Próstata/terapia , Estructura Terciaria de Proteína , Células TH1/inmunología , Vacunas Atenuadas/inmunología , Virus Vaccinia/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Gen Virol ; 85(Pt 8): 2347-2356, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15269376

RESUMEN

Influenza virus still poses a major threat to human health. Despite widespread vaccination programmes and the development of drugs targeting essential viral proteins, the extremely high mutation rate of influenza virus still leads to the emergence of new pathogenic virus strains. Therefore, it has been suggested that cellular cofactors that are essential for influenza virus infection might be better targets for antiviral therapy. It has previously been reported that influenza virus efficiently infects Epstein-Barr virus-immortalized B cells, whereas Burkitt's lymphoma cells are virtually resistant to infection. Using this cellular system, it has been shown here that an active NF-kappaB signalling pathway is a general prerequisite for influenza virus infection of human cells. Cells with low NF-kappaB activity were resistant to influenza virus infection, but became susceptible upon activation of NF-kappaB. In addition, blocking of NF-kappaB activation severely impaired influenza virus infection of otherwise highly susceptible cells, including the human lung carcinoma cell lines A549 and U1752 and primary human cells. On the other hand, infection with vaccinia virus was not dependent on an active NF-kappaB signalling pathway, demonstrating the specificity of this pathway for influenza virus infection. These results might be of major importance for both the development of new antiviral therapies and the understanding of influenza virus biology.


Asunto(s)
FN-kappa B/fisiología , Orthomyxoviridae/fisiología , Transducción de Señal , Linfocitos B/virología , Antígenos CD40/fisiología , Línea Celular , Herpesvirus Humano 4/genética , Humanos , Proteínas de la Matriz Viral/fisiología
14.
J Virol ; 77(15): 8290-8, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12857898

RESUMEN

Epstein-Barr virus (EBV) infects human resting B cells and transforms them in vitro into continuously growing lymphoblastoid cell lines (LCLs). EBV nuclear antigen 2 (EBNA2) is one of the first viral proteins expressed after infection. It is able to transactivate viral as well as cellular target genes by interaction with cellular transcription factors. EBNA2 target genes can be studied easily by using an LCL (ER/EB2-5) in which wild-type EBNA2 is replaced by an estrogen-inducible EBNA2. Since the cell surface molecule CD83, a member of the immunoglobulin superfamily and a marker for mature dendritic cells, appeared on the surface of ER/EB2-5 cells within 3 h after the addition of estrogen, we analyzed the regulation of CD83 induction by EBV in more detail. Despite its rapid induction, CD83 turned out to be an indirect target gene of EBNA2. We could show that the viral latent membrane protein 1 (LMP1) is responsible for the induction of CD83 by using an LCL expressing a ligand- or antibody-inducible recombinant nerve growth factor receptor-LMP1 fusion protein. The inducibility of the CD83 promoter by LMP1 was mediated by the activation of NF-kappaB, as seen by use of luciferase reporter assays using the CD83 promoter and LMP1 mutants. Additionally, fusion constructs of the transmembrane domain of LMP1 and the intracellular signaling domain of CD40, TNF-R1, and TNF-R2 likewise transactivated the CD83 promoter via NF-kappaB. Our studies show that CD83 is also a target of the NF-kappaB signaling pathway in B cells.


Asunto(s)
Herpesvirus Humano 4/fisiología , Inmunoglobulinas/metabolismo , Glicoproteínas de Membrana/metabolismo , FN-kappa B/metabolismo , Transducción de Señal , Regulación hacia Arriba , Proteínas de la Matriz Viral/metabolismo , Antígenos CD , Linfocitos B , Antígenos CD40/metabolismo , Línea Celular Transformada , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Estrógenos/farmacología , Eliminación de Gen , Humanos , Inmunoglobulinas/genética , Glicoproteínas de Membrana/genética , Regiones Promotoras Genéticas , Receptores del Factor de Necrosis Tumoral/metabolismo , Proteínas de la Matriz Viral/genética , Proteínas Virales , Antígeno CD83
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA