Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Prep Biochem Biotechnol ; 53(2): 148-156, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-35302435

RESUMEN

Cervical cancer caused by Human papillomavirus (HPV) is one of the most common causes of cancer death in women worldwide. Even though the disease can be avoided by immunization, the expensive price of HPV vaccines makes it hard to be accessed by women in middle-low-income countries. Thus, the development of generic HPV vaccines is needed to address inequalities in life-saving access. This study aimed to develop the HPV52 L1 VLP-based recombinant vaccine using Pichia pastoris expression system. The l1 gene was codon-optimized based on P. pastoris codon usage resulting CAI value of 0.804. The gene was inserted into the pD902 plasmid under the regulation of the AOX1 promoter. The linear plasmid was transformed into P. pastoris BG10 genome and screened in YPD medium containing zeocin antibiotic. Colony of transformant that grown on highest zeocin concentration was characterized by genomic PCR and sequencing. The positive clone was selected and expressed using BMGY/BMMY medium induced with various methanol concentrations. The SDS-PAGE and Western blot analyses showed that 55 kDa L1 protein was successfully expressed using an optimum concentration of 1% methanol. The self-assembly of HPV52 L1 protein was also proven using TEM analysis. Moreover, we also analyzed the B-cell epitope of HPV52 L1 protein based on several criteria, including antigenicity, surface accessibility, flexibility, and hydrophilicity. We assumed that epitope 476GLQARPKLKRPASSAPRTSTKKKKV500 could be developed as an epitope-based vaccine with a neutralizing antibody response toward HPV52 infection. Finally, our study provided the alternative for developing low-cost HPV vaccines, either VLP or epitope-based.


Asunto(s)
Virus del Papiloma Humano , Vacunas contra Papillomavirus , Femenino , Humanos , Metanol/metabolismo , Proteínas de la Cápside/genética , Pichia/genética , Pichia/metabolismo , Vacunas contra Papillomavirus/genética , Vacunas contra Papillomavirus/metabolismo , Epítopos/metabolismo , Codón/metabolismo
2.
Viruses ; 13(7)2021 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-34372545

RESUMEN

Human papillomavirus (HPV) imposes an increased risk of developing cervical, anal and oropharyngeal cancer. In the Western world, HPV infection is currently the major cause of oropharyngeal cancer. The effectiveness of HPV vaccines for oral or oropharyngeal HPV infection is yet to be determined. This study conducted a systematic literature search in Pubmed and Embase. Studies investigating the impact of HPV vaccines on oral or oropharyngeal HPV infection were enrolled. This review reports the relative prevention percentage (RPP), including a risk of bias assessment as well as a quality assessment study. Nine studies were included (48,777 participants): five cross-sectional studies; one randomized community trial study (RCT); one longitudinal cohort study; and two case-control studies. A significant mean RPP of 83.9% (66.6-97.8%) was calculated from the cross-sectional studies, 82.4% in the included RCT and 83% in the longitudinal cohort study. Further, two case-control studies that measured antibody response in participants immunized with HPV vaccines were included. Respectively, 100% and 93.2% of participants developed HPV-16 Immunoglobulin G (IgG) antibodies in oral fluids post-vaccination. Analysis of the studies identified a significant decrease in vaccine-type oral or oropharyngeal HPV infections in study participants immunized with HPV vaccines across study designs and heterogenous populations. Further, a significant percentage of participants developed IgG antibodies in oral fluid post-vaccination.


Asunto(s)
Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/farmacología , Alphapapillomavirus/patogenicidad , Estudios de Casos y Controles , Estudios Transversales , Femenino , Humanos , Estudios Longitudinales , Masculino , Boca/virología , Neoplasias de la Boca/prevención & control , Neoplasias de la Boca/virología , Neoplasias Orofaríngeas/prevención & control , Neoplasias Orofaríngeas/virología , Orofaringe/virología , Papillomaviridae/inmunología , Papillomaviridae/patogenicidad , Infecciones por Papillomavirus/metabolismo , Vacunas contra Papillomavirus/metabolismo , Profilaxis Pre-Exposición/métodos , Vacunación
3.
Biotechnol Lett ; 43(9): 1933-1944, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34313864

RESUMEN

OBJECTIVES: Human papillomavirus infection (HPV) is the most common viral infection which is causes of cervical, penal, vulvar, anal and, oropharyngeal cancer. E7 protein of HPV is a suitable target for induction of T cell responses and controlling HPV-related cancer. The aim of the current study was to designed and evaluated a novel fusion protein containing the different E7 proteins of the HPV 16, 18, 6 and 11, linked to the cell-penetrating peptide HIV-1 Tat 49-57, in order to improve cytotoxic immune responses in in-vitro and in-vivo. RESULTS: In this study whole sequence of HPV16,18,6,11 E7-Tat (47-57) and HPV16,18,6,11 E7 cloned into the vector and expressed in E. coli (BL21). The purified protein was confirmed by SDS page and western blotting and then injected into the C57BL/6 mice. The efficiency of the fusion protein vaccine was assessed by antibody response assay, cytokine assay (IL-4 and IFN-γ), CD + 8 cytotoxicity assay and tumor challenge experiment. Result showed that fusion proteins containing Adjuvant (IFA,CFA) could express higher titer of antibody. Also, we showed that vaccination with E7-Tat and, E7-Tat-ADJ induced high frequencies of E7-specific CD8 + T cells and CD107a expression as well as IFN-γ level and enhanced long-term survival in the therapeutic animal models. CONCLUSION: Our finding suggested that this novel fusion protein vaccine was able to induce therapeutic efficacy and immunogenicity by improving CD8 + T cell in TC-1 tumor bearing mice; so this vaccine may be appreciated for research against HPV and tumor immunotherapies.


Asunto(s)
Alphapapillomavirus/metabolismo , VIH-1/genética , Neoplasias Pulmonares/virología , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/administración & dosificación , Fragmentos de Péptidos/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Alphapapillomavirus/genética , Alphapapillomavirus/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Vacunas contra el Cáncer/metabolismo , Clonación Molecular , Escherichia coli/genética , Escherichia coli/crecimiento & desarrollo , Femenino , VIH-1/metabolismo , Papillomavirus Humano 11/genética , Papillomavirus Humano 11/metabolismo , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Papillomavirus Humano 18/genética , Papillomavirus Humano 18/metabolismo , Papillomavirus Humano 6/genética , Papillomavirus Humano 6/metabolismo , Humanos , Neoplasias Pulmonares/prevención & control , Ratones , Ratones Endogámicos C57BL , Proteínas E7 de Papillomavirus/genética , Vacunas contra Papillomavirus/inmunología , Vacunas contra Papillomavirus/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Vacunación , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Vacunas Sintéticas/metabolismo
4.
Int Immunopharmacol ; 69: 279-288, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30743204

RESUMEN

No licensed therapeutic human papillomavirus (HPV) vaccine is currently available, so it remains a high priority to develop a therapeutic HPV vaccine or prophylactic/therapeutic HPV vaccine for cervical cancer. In this current study, we designed an HPV vaccine including CpG oligodeoxynucleotides 1826 as an adjuvant and HPV16 E7 43-77 peptide as antigen, which contains a CD8 T cell epitope (E7 49-57), and two CD4 T cell epitopes (E7 43-77 and E7 50-62). The prophylactic and therapeutic effect on cervical cancer induced by a single administration of vaccine, were comprehensively evaluated by examining the tumor size and the percentage of tumor-free/bearing mice. The cellular immunity and modulation of immunosuppressive cells induced by the vaccine were evaluated by examining intracellular cytokine staining (ICS) of splenocytes and FCM, respectively. Antigen-specific cytotoxic T-lymphocyte (CTL) responses were investigated using in vivo cytolytic assay. The results showed that the single administration of vaccine elicited significant prophylactic as well as therapeutic effect on cervical cancer. The increased cellular immunity mediated by CD4 + IFN-γ + T cells and CD8 + IFN-γ + T cells, and the decreased numbers of immunosuppressive cells including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) were induced by the vaccine. Antigen-specific CTL response was also induced by vaccination. These findings suggested that significant anti-tumor effect of the vaccine may result from the induction of increased cellular immunity and decreased immunosuppressive cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Epítopos de Linfocito T/metabolismo , Papillomavirus Humano 16/fisiología , Células Supresoras de Origen Mieloide/inmunología , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/inmunología , Vacunas contra Papillomavirus/inmunología , Péptidos/metabolismo , Linfocitos T Reguladores/inmunología , Neoplasias del Cuello Uterino/inmunología , Adyuvantes Inmunológicos , Animales , Células Cultivadas , Citotoxicidad Inmunológica , Femenino , Humanos , Interferón gamma/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/inmunología , Vacunas contra Papillomavirus/metabolismo , Vacunación , Vacunas de Subunidad
5.
Appl Microbiol Biotechnol ; 100(3): 1231-1240, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26446387

RESUMEN

Here, we describe a process for expression, purification, and characterization of truncated human papillomavirus type-6 (HPV-6) L1 virus-like particles (VLPs). The scalable cultivation process in a WAVE Bioreactor at the 10-L scale was optimized to express HPV-6 L1 VLPs using the baculovirus insect expression system. A hollow fiber membrane system was used for the integrated operation, including concentration, diafiltration, extraction, and clarification. The HPV-6 L1 protein was further purified by anion-exchange chromatography and hydrophobic chromatography. The HPV-6 L1 protein could self-assemble into VLPs with a diameter of approximately 50-60 nm after removal of the reductant dithiothreitol (DTT). The final purified HPV-6 L1 VLPs product was characterized to estimate yield and purity, and exceeds the requirements for pharmaceutical-grade VLP vaccine. Immunization of mice demonstrated that the vaccine could elicit high titer neutralizing antibodies in vivo. This study confirms the feasibility of producing pharmaceutical-grade HPV type-6 L1 VLPs on an industrial scale for clinical trials.


Asunto(s)
Proteínas de la Cápside/metabolismo , Microbiología Industrial/métodos , Infecciones por Papillomavirus/virología , Vacunas contra Papillomavirus/metabolismo , Animales , Baculoviridae/genética , Baculoviridae/metabolismo , Reactores Biológicos , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Proteínas de la Cápside/aislamiento & purificación , Femenino , Humanos , Inmunización , Microbiología Industrial/instrumentación , Ratones , Ratones Endogámicos BALB C , Vacunas contra Papillomavirus/química , Vacunas contra Papillomavirus/genética , Vacunas contra Papillomavirus/aislamiento & purificación , Spodoptera
6.
Gene Ther ; 22(7): 560-7, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25756550

RESUMEN

Cellular immunity against cancer can be achieved with viral vector- and DNA-based immunizations. In preclinical studies, cancer vaccines are very potent, but in clinical trials these potencies are not achieved yet. Thus, a rational approach to improve cancer vaccines is warranted. We previously demonstrated that the relatively low intrinsic immunogenicity of DNA vaccines could be enhanced by inclusion of endoplasmic reticulum (ER) targeting and universal helper epitopes within the vaccine. We now evaluated whether an optimal antigen format, as defined in DNA vaccines, can further enhance the effectiveness of recombinant Semliki Forest virus (rSFV) vaccines. To this purpose, we generated, characterized and evaluated the efficacy of rSFV replicon particles expressing human papillomavirus E6 and/or E7 proteins fused to several helper T-cell epitopes and an ER targeting signal. Here, we show that inclusion of a helper cassette and an ER targeting signal enhanced protein stability and markedly augmented the frequencies of human papillomavirus-specific T cells. Even at an immunization dose of as low as 10(5) replicon particles, this novel vaccine achieved tumor regression and protection. Thus, even highly effective viral vector vaccines can benefit from an improved antigen format, based on the inclusion of defined helper epitopes and ER targeting.


Asunto(s)
Antígenos Virales/inmunología , Vacunas contra el Cáncer/inmunología , Proteínas Oncogénicas Virales/inmunología , Proteínas E7 de Papillomavirus/inmunología , Vacunas contra Papillomavirus/inmunología , Animales , Antígenos Virales/administración & dosificación , Antígenos Virales/genética , Vacunas contra el Cáncer/administración & dosificación , Cricetinae , Epítopos de Linfocito T/química , Femenino , Humanos , Riñón/citología , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/administración & dosificación , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/administración & dosificación , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Vacunas contra Papillomavirus/genética , Vacunas contra Papillomavirus/metabolismo , Virus de los Bosques Semliki/genética , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Neoplasias del Cuello Uterino/prevención & control , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología
7.
PLoS One ; 9(7): e101576, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24999962

RESUMEN

Antibodies specific for neutralizing epitopes in either Human papillomavirus (HPV) capsid protein L1 or L2 can mediate protection from viral challenge and thus their accurate and sensitive measurement at high throughput is likely informative for monitoring response to prophylactic vaccination. Here we compare measurement of L1 and L2-specific neutralizing antibodies in human sera using the standard Pseudovirion-Based Neutralization Assay (L1-PBNA) with the newer Furin-Cleaved Pseudovirion-Based Neutralization Assay (FC-PBNA), a modification of the L1-PBNA intended to improve sensitivity towards L2-specific neutralizing antibodies without compromising assay of L1-specific responses. For detection of L1-specific neutralizing antibodies in human sera, the FC- PBNA and L1-PBNA assays showed similar sensitivity and a high level of correlation using WHO standard sera (n = 2), and sera from patients vaccinated with Gardasil (n = 30) or an experimental human papillomavirus type 16 (HPV16) L1 VLP vaccine (n = 70). The detection of L1-specific cross-neutralizing antibodies in these sera using pseudovirions of types phylogenetically-related to those targeted by the L1 virus-like particle (VLP) vaccines was also consistent between the two assays. However, for sera from patients (n = 17) vaccinated with an L2-based immunogen (TA-CIN), the FC-PBNA was more sensitive than the L1-PBNA in detecting L2-specific neutralizing antibodies. Further, the neutralizing antibody titers measured with the FC-PBNA correlated with those determined with the L2-PBNA, another modification of the L1-PBNA that spacio-temporally separates primary and secondary receptor engagement, as well as the protective titers measured using passive transfer studies in the murine genital-challenge model. In sum, the FC-PBNA provided sensitive measurement for both L1 VLP and L2-specific neutralizing antibody in human sera. Vaccination with TA-CIN elicits weak cross-protective antibody in a subset of patients, suggesting the need for an adjuvant.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Furina/metabolismo , Pruebas de Neutralización/métodos , Vacunas contra Papillomavirus/inmunología , Vacunas contra Papillomavirus/metabolismo , Vacunación , Virión/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Especificidad de Anticuerpos , Proteínas de la Cápside/inmunología , Proteínas de la Cápside/metabolismo , Vacuna Tetravalente Recombinante contra el Virus del Papiloma Humano Tipos 6, 11 , 16, 18 , Humanos , Pruebas de Neutralización/normas , Proteínas Oncogénicas Virales/inmunología , Proteínas Oncogénicas Virales/metabolismo , Papillomaviridae/inmunología , Proteolisis , Estándares de Referencia
8.
Curr Med Chem ; 21(7): 932-40, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24083601

RESUMEN

Gardasil® is a quadrivalent human papillomavirus (HPV) protein-based vaccine containing genotype-specific L1 capsid proteins of HPV-16, HPV-18, HPV-6 and HPV-11 in the form of virus-like-particles (VLPs) as the active ingredient. The VLPs are produced by a DNA recombinant technology. It is uncertain if the residual HPV L1 gene DNA fragments in the vaccine products are considered contaminants or excipients of the Gardasil® vaccine. Because naked viral DNA fragments, if present in the vaccine, may bind to the insoluble amorphous aluminum hydroxyphosphate sulfate (AAHS) adjuvant which may help deliver the foreign DNA into macrophages, causing unintended pathophysiologic effects, experiments were undertaken to develop tests for HPV L1 gene DNA fragments in the final products of Gardasil® by polymerase chain reaction (PCR) and direct DNA sequencing. The results showed that while the HPV-11 and HPV-18 L1 gene DNA fragments in Gardasil® were readily amplified by the common GP6/MY11 degenerate consensus primers, the HPV-16 L1 gene DNA may need specially designed non-degenerate PCR primers for amplification at different regions of the L1 gene and different stringency conditions for detection. These variable melting profiles of HPV DNA in the insoluble fraction of the Gardasil® vaccine suggest that the HPV DNA fragments are firmly bound to the aluminum AAHS adjuvant. All methods developed for detecting residual HPV DNA in the vaccine Gardasil® for quality assurance must take into consideration the variable melting profiles of the DNA to avoid false negative results.


Asunto(s)
ADN/análisis , Vacunas contra Papillomavirus/genética , Adyuvantes Inmunológicos/química , Secuencia de Bases , Proteínas de la Cápside/biosíntesis , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , ADN/metabolismo , Cartilla de ADN/metabolismo , Vacuna Tetravalente Recombinante contra el Virus del Papiloma Humano Tipos 6, 11 , 16, 18 , Humanos , Desnaturalización de Ácido Nucleico , Vacunas contra Papillomavirus/inmunología , Vacunas contra Papillomavirus/metabolismo , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN
9.
PLoS One ; 7(11): e50296, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23209698

RESUMEN

Human papillomavirus is known to be the major pathogen of cervical cancer. Here, we report the efficacy of a bivalent human papillomavirus type 16 and 18 DNA vaccine system following repeated dosing in mice and pigs using a recombinant baculovirus bearing human endogenous retrovirus envelope protein (AcHERV) as a vector. The intramuscular administration of AcHERV-based HPV16L1 and HPV18L1 DNA vaccines induced antigen-specific serum IgG, vaginal IgA, and neutralizing antibodies to levels comparable to those achieved using the commercially marketed vaccine Cervarix. Similar to Cervarix, AcHERV-based bivalent vaccinations completely blocked subsequent vaginal challenge with HPV type-specific pseudovirions. However, AcHERV-based bivalent vaccinations induced significantly higher cell-mediated immune responses than Cervarix, promoting 4.5- (HPV16L1) and 3.9-(HPV18L1) fold higher interferon-γ production in splenocytes upon stimulation with antigen type-specific pseudovirions. Repeated dosing did not affect the immunogenicity of AcHERV DNA vaccines. Three sequential immunizations with AcHERV-HP18L1 DNA vaccine followed by three repeated dosing with AcHERV-HP16L1 over 11 weeks induced an initial production of anti-HPV18L1 antibody followed by subsequent induction of anti-HPV16L1 antibody. Finally, AcHERV-based bivalent DNA vaccination induced antigen-specific serum IgG immune responses in pigs. These results support the further development of AcHERV as a bivalent human papillomavirus DNA vaccine system for use in preventing the viral infection as well as treating the infected women by inducing both humoral and cell-mediated immune responses. Moreover, the possibility of repeated dosing indicates the utility of AcHERV system for reusable vectors of other viral pathogen vaccines.


Asunto(s)
Retrovirus Endógenos/genética , Vacunas contra Papillomavirus/inmunología , Retroviridae/genética , Vacunas de ADN/metabolismo , Animales , Baculoviridae/genética , Retrovirus Endógenos/metabolismo , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Vectores Genéticos , Proteínas Fluorescentes Verdes/metabolismo , Papillomavirus Humano 16/metabolismo , Papillomavirus Humano 18/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Vacunas contra Papillomavirus/metabolismo , Porcinos
10.
Artículo en Inglés | MEDLINE | ID: mdl-22134037

RESUMEN

Several studies indicated that biopharmaceuticals based on the recombinant protein E7 of human papillomavirus (HPV) can serve as therapeutic vaccines preventing the development of cancer in women infected with high-risk types of HPV such as HPV16. Here, we report effective extraction and purification of a plant-produced E7GGG-lichenase fusion protein, an HPV16 subunit vaccine candidate, from Nicotiana benthamiana plants, to a high yield. The target contains the modified HPV16 E7 protein internally fused to the surface loop of a truncated, hexa-His- and KDEL-tagged variant of bacterial lichenase, and has been previously shown to possess anti-cancer activity in an animal model. We purified the protein using a combination of immobilized metal-ion affinity chromatography and gel filtration. The achieved purity of the final product was 99% as confirmed by Coomassie or SYPRO Ruby staining after sodium dodecyl sulfate-polyacrylamide gel electrophoresis and by analytical size exclusion chromatography coupled with multi-angle laser light scattering. The overall yield was 50% corresponding to 0.1g of protein per 1 kg plant biomass. Only slight changes in these parameters were observed during the process scale-up from 50 g to 1 kg of processed leaf biomass.


Asunto(s)
Papillomavirus Humano 16/química , Proteínas E7 de Papillomavirus/química , Vacunas contra Papillomavirus/química , Proteínas Recombinantes de Fusión/química , Western Blotting , Tampones (Química) , Cromatografía en Gel , Cromatografía por Intercambio Iónico , Electroforesis en Gel de Poliacrilamida , Glicósido Hidrolasas/metabolismo , Papillomavirus Humano 16/inmunología , Peso Molecular , Proteínas E7 de Papillomavirus/inmunología , Proteínas E7 de Papillomavirus/aislamiento & purificación , Vacunas contra Papillomavirus/metabolismo , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/aislamiento & purificación , Nicotiana/química , Nicotiana/virología , Vacunas de Subunidad/química , Vacunas de Subunidad/inmunología
11.
Plant Biotechnol J ; 9(6): 651-60, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21447051

RESUMEN

Human papillomavirus (HPV) causes cervical cancer in women worldwide, which is currently prevented by vaccines based on virus-like particles (VLPs). However, these vaccines have certain limitations in their availability to developing countries, largely due to elevated costs. Concerning the highest burden of disease in resource-poor countries, development of an improved mucosal and cost-effective vaccine is a necessity. As an alternative to VLPs, capsomeres have been shown to be highly immunogenic and can be used as vaccine candidate. Furthermore, coupling of an adjuvant like Escherichia coli heat-labile enterotoxin subunit B (LTB) to an antigen can increase its immunogenicity and reduce the costs related to separate co-administration of adjuvants. Our study demonstrates the expression of two pentameric proteins: the modified HPV-16 L1 (L1_2xCysM) and LTB as a fusion protein in tobacco chloroplasts. Homoplasmy of the transplastomic plants was confirmed by Southern blotting. Western blot analysis showed that the LTB-L1 fusion protein was properly expressed in the plastids and the recombinant protein was estimated to accumulate up to 2% of total soluble protein. Proper folding and display of conformational epitopes for both LTB and L1 in the fusion protein was confirmed by GM1-ganglioside binding assay and antigen capture ELISA, respectively. However, all transplastomic lines showed chlorosis, male sterility and growth retardation, which persisted in the ensuing four generations studied. Nevertheless, plants reached maturity and produced seeds by pollination with wild-type plants. Taken together, these results pave the way for the possible development of a low-cost adjuvant-coupled vaccine with potentially improved immunogenicity against cervical cancer.


Asunto(s)
Proteínas de la Cápside/inmunología , Proteínas Oncogénicas Virales/inmunología , Vacunas contra Papillomavirus/genética , Adyuvantes Inmunológicos/genética , Antígenos Virales/genética , Toxinas Bacterianas/química , Toxinas Bacterianas/genética , Toxinas Bacterianas/metabolismo , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Cloroplastos/genética , Cloroplastos/metabolismo , Enterotoxinas/química , Enterotoxinas/genética , Enterotoxinas/metabolismo , Epítopos , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Femenino , Gangliósido G(M1)/metabolismo , Humanos , Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/genética , Vacunas contra Papillomavirus/inmunología , Vacunas contra Papillomavirus/metabolismo , Fenotipo , Infertilidad Vegetal/genética , Plantas Modificadas Genéticamente/genética , Plastidios/genética , Polinización , Conformación Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes de Fusión/metabolismo , Semillas/fisiología , Nicotiana/genética , Nicotiana/crecimiento & desarrollo , Nicotiana/metabolismo , Neoplasias del Cuello Uterino/inmunología
12.
Arch Virol ; 156(4): 587-95, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21234770

RESUMEN

We investigated the potential of Nicotiana benthamiana to express the E7 protein of human papillomavirus 8 (HPV-8), a paradigm genotype among cutaneous HPVs. The protein, modified in its putative pRb-binding domain (E7(QGD)), was transiently expressed in leaves following infiltration with agrobacteria carrying either a binary vector combined with silencing suppressor constructs or replicating tobacco mosaic virus (TMV)-based vectors with different targeting signals. HPV-8 E7(QGD) yields ranged from 250 ng to 4.6 mg per gram of fresh leaf tissue. The highest yields were obtained with TMV-based vectors targeting the antigen to the apoplast. HPV8-CER (H2(q)) mice transformed with the complete early region of HPV-8 showed a delay in the onset of skin papillomatous lesions and produced E7-specific immunoglobulins G when inoculated subcutaneously with leaf extracts expressing E7(QGD). Furthermore, we demonstrated that the plant-made HPV-8 E7(QGD) induced a specific cytotoxic response in C57BL/6 (H2(b)) mice.


Asunto(s)
Proteínas Oncogénicas Virales/inmunología , Papiloma/prevención & control , Vacunas contra Papillomavirus/inmunología , Neoplasias Cutáneas/prevención & control , Animales , Anticuerpos Antivirales/sangre , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Inmunoglobulina G/sangre , Inyecciones Subcutáneas , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Papiloma/inmunología , Vacunas contra Papillomavirus/genética , Vacunas contra Papillomavirus/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/metabolismo , Piel , Neoplasias Cutáneas/inmunología , Nicotiana/genética , Nicotiana/metabolismo
13.
Curr Opin Mol Ther ; 12(5): 598-606, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20886392

RESUMEN

Commercially available prophylactic HPV vaccines for cervical cancer prevention have limited use in women with previous viral exposure. Therefore, a therapeutic HPV vaccine would benefit patients with HPV-associated genital diseases. Being developed by Cancer Research Technology Ltd, under license from Xenova Group plc, TA-CIN (Tissue Antigen - Cervical Intraepithelial Neoplasia) is a fusion protein vaccine comprising the HPV16 viral proteins L2, E6 and E7 for the treatment of HPV16-associated genital diseases. In mouse models, TA-CIN induced dose-dependent HPV16-specific CD4 and CD8 T-cell responses, which were enhanced when boosted with the vaccinia-based vector vaccine TA-HPV (Therapeutic Antigen - HPV). A phase I clinical trial of TA-CIN in healthy volunteers reported no serious adverse events and HPV16-specific cellular immune responses. Phase II trials in patients with anogenital and vulval intraepithelial neoplasia investigated heterologous prime/boost strategies with TA-CIN/TA-HPV and TA-HPV/TA-CIN, but neither of the regimens offered advantages over single-agent TA-HPV. A recent phase II trial investigating imiquimod/TA-CIN in patients with vulval intraepithelial neoplasia demonstrated significant infiltration of CD4 and CD8 T-cells in lesion responders and complete lesion regression in 63% of patients. More comprehensive case-controlled trials are needed to define responders to immunotherapy with TA-CIN and verify its prophylactic and therapeutic properties.


Asunto(s)
Enfermedades de los Genitales Femeninos/tratamiento farmacológico , Enfermedades de los Genitales Femeninos/virología , Enfermedades de los Genitales Masculinos/tratamiento farmacológico , Enfermedades de los Genitales Masculinos/virología , Infecciones por Papillomavirus/tratamiento farmacológico , Vacunas contra Papillomavirus/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Femenino , Enfermedades de los Genitales Femeninos/inmunología , Enfermedades de los Genitales Masculinos/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/virología , Vacunas contra Papillomavirus/efectos adversos , Vacunas contra Papillomavirus/genética , Vacunas contra Papillomavirus/metabolismo , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
14.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 26(1): 38-40, 2010 Jan.
Artículo en Chino | MEDLINE | ID: mdl-20056086

RESUMEN

AIM: To develop a recombinant protein vaccine with epitopes on L1 protein for prevention of HPV16 infection. METHODS: One B cell epitope and one T cell epitope from HPV16 L1 protein were chosen to construct genes encoding different recombinant proteins. Then all the 3D structures of the proteins were predicted by Geno3D in internet. An optimal protein named RH was chosen in which three B epitopes' location simulates that in the real HPV16 L1. Gene X (consisting of B+T+T) and gene Y (consisting of B+T) were constructed by multiple PCR. RH gene (consisting of X+X+X+X+Y+Y+Y) was constructed, sub-cloned into pFastbac1, and expressed in Sf9. RESULTS: RH protein with epitopes on HPV16 L1 was designed and predicted by Geno3D. RH gene was constructed and sub cloned into pFastbac1. Finally, it was successfully expressed in baculovirus expression system. CONCLUSION: A HPV16 recombinant vaccine is successfully constructed and expressed.


Asunto(s)
Proteínas de la Cápside/genética , Diseño Asistido por Computadora , Proteínas Oncogénicas Virales/genética , Vacunas contra Papillomavirus/genética , Ingeniería de Proteínas , Animales , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Línea Celular , Expresión Génica , Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/metabolismo , Vacunas contra Papillomavirus/química , Vacunas contra Papillomavirus/metabolismo , Estructura Terciaria de Proteína
15.
J Virol ; 83(19): 10085-95, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19640991

RESUMEN

The amino (N) terminus of the human papillomavirus (HPV) minor capsid protein L2 can induce low-titer, cross-neutralizing antibodies. The aim of this study was to improve immunogenicity of L2 peptides by surface display on highly ordered, self-assembled virus-like particles (VLP) of major capsid protein L1, and to more completely characterize neutralization epitopes of L2. Overlapping peptides comprising amino acids (aa) 2 to 22 (hereafter, chimera or peptide 2-22), 13 to 107, 18 to 31, 17 to 36, 35 to 75, 75 to 112, 115 to 154, 149 to 175, and 172 to 200 of HPV type 16 (HPV16) L2 were genetically engineered into the DE surface loop of bovine papillomavirus type 1 L1 VLP. Except for chimeras 35-75 and 13-107, recombinant fusion proteins assembled into VLP. Vaccination of rabbits with Freund's adjuvanted native VLP induced higher L2-specific antibody titers than vaccination with corresponding sodium dodecyl sulfate-denatured proteins. Immune sera to epitopes within residues 13 to 154 neutralized HPV16 in pseudovirion neutralization assays, whereas chimera 17-36 induced additional cross-neutralization to divergent high-risk HPV18, -31, -45, -52, and -58; low-risk HPV11; and beta-type HPV5 (titers of 50 to 10,000). Aluminum hydroxide-monophosphoryl lipid A (Alum-MPL)-adjuvanted VLP induced similar patterns of neutralization in both rabbits and mice, albeit with 100-fold-lower titers than Freund's adjuvant. Importantly, Alum-MPL-adjuvanted immunization with chimeric HPV16L1-HPV16L2 (peptide 17-36) VLP induced neutralization or cross-neutralization of HPV16, -18, -31, -45, -52, and -58; HPV6 and -11; and HPV5 (titers of 50 to 100,000). Immunization with HPV16 L1-HPV16 L2 (chimera 17-36) VLP in adjuvant applicable for human use induces broad-spectrum neutralizing antibodies against HPV types evolutionarily divergent to HPV16 and thus may protect against infection with mucosal high-risk, low-risk, and beta HPV types and associated disease.


Asunto(s)
Baculoviridae/metabolismo , Proteínas de la Cápside/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Vacunas contra Papillomavirus/metabolismo , Animales , Cápside/química , Epítopos/química , Ratones , Ratones Endogámicos BALB C , Microscopía Electrónica de Transmisión/métodos , Péptidos/química , Estructura Terciaria de Proteína , Conejos , Proteínas Recombinantes de Fusión/química , Riesgo
16.
Vaccine ; 27(32): 4363-9, 2009 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-19481843

RESUMEN

DNA vaccines have emerged as a potential alternative to current strategies to control cancer for their safety, stability and ease of preparation. We have previously demonstrated that a DNA vaccine encoding calreticulin (CRT) linked to human papillomavirus type 16 (HPV-16) E7 antigen (CRT/E7) can generate significant E7-specific immune responses and antitumor effects in vaccinated mice, thus enhancing DNA vaccine potency. Another strategy to improve DNA vaccine potency is by enhancing the level of expression of the antigen encoded in the vaccine. DNA methylation has been shown to lead to silencing of the genes that would affect the expression of the encoded antigen of the DNA vaccines. In the current study, we reasoned that CRT/E7 DNA vaccination combined with demethylating agent, 5-aza-2'-deoxycytidine (DAC) would lead to upregulation of CRT/E7 expression, resulting in improved DNA vaccine potency. We found that pre-treatment with DAC led to increased CRT/E7 DNA expression, leading to enhanced E7-specific CD8+ T cell immune responses as well as the antitumor effects generated by the CRT/E7 DNA vaccine. Thus, our data suggest that combination of CRT/E7 DNA vaccination with DAC treatment may represent a potentially promising approach to control HPV-associated malignancies. The clinical implications of this study are discussed.


Asunto(s)
Azacitidina/análogos & derivados , Vacunas contra Papillomavirus/inmunología , Vacunas contra Papillomavirus/metabolismo , Vacunas de ADN/inmunología , Vacunas de ADN/metabolismo , Animales , Azacitidina/metabolismo , Linfocitos T CD8-positivos/inmunología , Decitabina , Femenino , Expresión Génica , Humanos , Ratones , Análisis de Supervivencia
17.
J Biomed Sci ; 16: 49, 2009 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-19473507

RESUMEN

BACKGROUND: It is important to develop innovative therapies for advanced stage cancers in addition to the conventional therapies including chemotherapy, radiation and surgery. Antigen-specific immunotherapy has emerged as a novel alternate therapy for advanced stage cancers, which may be employed in conjunction with conventional therapies. METHODS: In the current study, we tested the effect of treatment with the chemotherapeutic agent, apigenin in combination with DNA vaccines encoding the HPV-16 E7 antigen linked to heat shock protein 70 (HSP70) in the control of the E7-expressing tumor, TC-1. RESULTS: We observed that treatment with apigenin rendered the TC-1 tumor cells more susceptible to lysis by E7-specific cytotoxic CD8+ T cells. Furthermore, treatment of TC-1 tumor cells with apigenin was found to enhance apoptotic tumor cell death in vitro in a dose-dependant manner. We showed that TC-1 tumor-bearing mice treated with apigenin combined with E7-HSP70 DNA generate highest frequency of primary and memory E7-specific CD8+ T cells, leading to potent therapeutic anti-tumor effects against E7-expressing tumors. CONCLUSION: Thus, apigenin represents a promising chemotherapeutic agent, which may be used in combination with immunotherapy for the treatment of advanced stage cancers. The clinical implications of the current strategy are discussed.


Asunto(s)
Antineoplásicos/administración & dosificación , Apigenina/uso terapéutico , Inmunoterapia/métodos , Animales , Antígenos de Neoplasias/metabolismo , Apoptosis , Terapia Combinada/métodos , Femenino , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus , Vacunas contra Papillomavirus/metabolismo , Vacunas de ADN/uso terapéutico
18.
Wei Sheng Wu Xue Bao ; 49(11): 1527-33, 2009 Nov.
Artículo en Chino | MEDLINE | ID: mdl-20112683

RESUMEN

OBJECTIVE: To produce human papillomavirus type 11 virus-like particles (HPV11 VLPs) from Escherichia coli and to investigate its immunogenicity and type cross neutralization nature. METHODS: We expressed the major capsid protein of HPV11 (HPV11-L1) in Escherichia coli ER2566 in non fusion fashion and purified by amino sulfate precipitation, ion-exchange chromatography and hydrophobic interaction chromatography, sequentially. Then we removed the reductant DTT to have the purified HPV11-L1 self-assemble into VLPs in vitro. We investigated the morphology of these VLPs with dynamic light scattering and transmission electron microscopy. We assayed the immunogenicity of the resultant HPV11 VLPs by vaccinations on mice and evaluated by HPV6/11/16/18 pseudovirion neutralization cell models. RESULTS: We expressed HPV11 L1 in Escherichia coli with two forms, soluble and inclusion body. The soluble HPV11 L1 with over 95% purity can self assemble to VLPs in high efficiency. Morphologically, these VLPs were globular, homogeneous and with a diameter of - 50 nm, which is quite similar with native HPV11 virions. The half effective dosage (ED50) of HPV11 VLPs is 0.031 microg, and the maximum titer of neutralizing antibody elicited is averaged to 10(6). The cross neutralization activity (against HPV6/16/18) of the anti-HPV11 serum was found to have exact correlation to the inter-type homology in amino acid alignment. CONCLUSION: We can provide HPV11 VLPs with highly immunogenicity from prokaryote expression system, which may pave a new way for research and development of prophylactic vaccine for HPV11.


Asunto(s)
Proteínas de la Cápside/inmunología , Proteínas de la Cápside/aislamiento & purificación , Expresión Génica , Papillomavirus Humano 11/inmunología , Infecciones por Papillomavirus/inmunología , Virión/inmunología , Animales , Anticuerpos Antivirales/inmunología , Proteínas de la Cápside/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Femenino , Papillomavirus Humano 11/clasificación , Papillomavirus Humano 11/genética , Humanos , Ratones , Ratones Endogámicos BALB C , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/virología , Vacunas contra Papillomavirus/genética , Vacunas contra Papillomavirus/inmunología , Vacunas contra Papillomavirus/aislamiento & purificación , Vacunas contra Papillomavirus/metabolismo , Filogenia , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Virión/clasificación , Virión/genética
19.
Acta Biochim Biophys Sin (Shanghai) ; 40(5): 437-42, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18465029

RESUMEN

To improve the existing human papillomavirus type 16 (HPV16) virus-like particle (VLP) preparation, the Drosophila inducible/secreted expression system, a highly efficient, economical method, was used to produce HPV16 VLPs. Drosophila Schneider-2 cells were cotransfected with pMT/BiP/V5-His expression vector containing the target gene encoding HPV16L1 protein without nucleus localization sequence and the selection vector pCoHygro plasmids at the ratio of 4:1. The stabled hygromycin-resistant cell line was obtained 1 month later, and the protein expression was induced by copper sulfate. The molecular mass of expressed HPV16L1 protein was 66 kDa, as revealed by SDS-PAGE, and confirmed by Western blot analysis. The yield of HPV16L1 protein was 0.554 mg per 1x10(7) cells. The characteristics of HPV16L1 protein were further analyzed by mouse erythrocyte hemagglutination assay, hemagglutination inhibition assay, and transmission electron microscopy. Results showed that the truncated protein was as biologically active as natural HPVL1 protein, inducing murine erythrocyte agglutination and VLP formation. These findings indicate that the Drosophila inducible/secreted expression system is promising as a convenient and economical method for the preparation of HPV16 VLP vaccine.


Asunto(s)
Drosophila/metabolismo , Papillomavirus Humano 16/metabolismo , Vacunas contra Papillomavirus/metabolismo , Ingeniería de Proteínas/métodos , Transfección/métodos , Animales , Línea Celular , Drosophila/genética , Papillomavirus Humano 16/genética , Vacunas contra Papillomavirus/genética , Proteínas Recombinantes/química
20.
Clin Cancer Res ; 13(1): 341-9, 2007 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-17200373

RESUMEN

PURPOSE: The goal of this study was to investigate the therapeutic potentials of combining chemotherapy with human papillomavirus (HPV) E7 subunit vaccines in an animal tumor model and to determine the underlying therapeutic mechanisms. EXPERIMENTAL DESIGN: Animals bearing HPV E6/E7-expressing tumors were treated intratumorally with a selected cytotoxic drug, cisplatin, twice at 1-week interval and s.c. with E7 subunit vaccines thrice at 1-week interval. Tumor chemoimmunoresponse was measured by tumor size. Ag-specific CTL activities and tumor histology were checked in mice under treatments. Apoptosis, in vivo T-cell subset depletion, adoptive CTL transfer, and tumor regression were used to determine the mechanisms for antitumor therapeutic effects. RESULTS: Combined therapy using cisplatin plus E7 subunit vaccines improved cure and recurrence rates of tumors and long-term antitumor immunity dramatically more than single therapy alone. In particular, both components of E7 subunit vaccines were required for induction of Ag-specific CTL as well as therapeutic synergy when combined with cisplatin. This therapeutic synergy was abrogated by depletion of CD8(+) T cells in vivo and was concomitant with histologic changes (such as heavy infiltration of lymphocytes and reduced tumor cell density). Finally, the increased sensitivity of cisplatin-treated tumors to CTL-mediated killing was found to be responsible for therapeutic synergy. CONCLUSIONS: E7 subunit vaccines plus cisplatin mediate antitumor therapeutic synergy through the increased sensitivity of cisplatin-treated tumors to CTL-mediated killing. Moreover, E7-based therapeutic vaccines have the potential to improve chemotherapy in patients with cervical cancer.


Asunto(s)
Vacunas contra el Cáncer , Cisplatino/administración & dosificación , Terapia Combinada , Proteínas E7 de Papillomavirus/química , Vacunas contra Papillomavirus/metabolismo , Linfocitos T Citotóxicos/citología , Animales , Antineoplásicos/farmacología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Femenino , Inmunoterapia/métodos , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA