Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
J Virol ; 85(12): 5764-72, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21490100

RESUMEN

We constructed vaccine vectors based on live recombinant vesicular stomatitis virus (VSV) and a Semliki Forest virus (SFV) replicon (SFVG) that propagates through expression of the VSV glycoprotein (G). These vectors expressing simian immunodeficiency virus (SIV) Gag and Env proteins were used to vaccinate rhesus macaques with a new heterologous prime-boost regimen designed to optimize induction of antibody. Six vaccinated animals and six controls were then given a high-dose mucosal challenge with the diverse SIVsmE660 quasispecies. All control animals became infected and had peak viral RNA loads of 10(6) to 10(8) copies/ml. In contrast, four of the vaccinees showed significant (P = 0.03) apparent sterilizing immunity and no detectable viral loads. Subsequent CD8(+) T cell depletion confirmed the absence of SIV infection in these animals. The two other vaccinees had peak viral loads of 7 × 10(5) and 8 × 10(3) copies/ml, levels below those of all of the controls, and showed undetectable virus loads by day 42 postchallenge. The vaccine regimen induced high-titer prechallenge serum neutralizing antibodies (nAbs) to some cloned SIVsmE660 Env proteins, but antibodies able to neutralize the challenge virus swarm were not detected. The cellular immune responses induced by the vaccine were generally weak and did not correlate with protection. Although the immune correlates of protection are not yet clear, the heterologous VSV/SFVG prime-boost is clearly a potent vaccine regimen for inducing virus nAbs and protection against a heterogeneous viral swarm.


Asunto(s)
Anticuerpos Antivirales/sangre , Vectores Genéticos/inmunología , Esquemas de Inmunización , Vacunas contra el SIDAS/administración & dosificación , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Productos del Gen env/genética , Productos del Gen env/inmunología , Productos del Gen env/metabolismo , Productos del Gen gag/genética , Productos del Gen gag/inmunología , Productos del Gen gag/metabolismo , Vectores Genéticos/administración & dosificación , Inmunización , Inmunización Secundaria , Macaca mulatta , Pruebas de Neutralización , Vacunas contra el SIDAS/genética , Vacunas contra el SIDAS/inmunología , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/metabolismo , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Virus de la Estomatitis Vesicular Indiana/genética , Virus de la Estomatitis Vesicular Indiana/metabolismo , Carga Viral
2.
Proc Natl Acad Sci U S A ; 105(15): 5850-5, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18413606

RESUMEN

Persistent infection with the high-risk subset of genitotropic human papillomavirus (HPV) genotypes is a necessary cause of cervical cancer. Given the global burden of cervical cancer, a low-cost, broadly protective vaccine is needed. RG-1 is a cross-neutralizing and protective monoclonal antibody that recognizes residues 17-36 of HPV16 minor capsid protein L2. Because this epitope is highly conserved in divergent HPV types, we determined whether vaccination with HPV16 L2 17-36 peptide is broadly protective. The peptide was administered to BALB/c mice three times at monthly intervals, either alone or in the context of a synthetic lipopeptide vaccine candidate (P25-P2C-HPV) produced by linkage of the HPV peptide with a broadly recognized T helper epitope (P25) and the Toll-like receptor-2 (TLR2) ligand dipalmitoyl-S-glyceryl cysteine (P2C). In contrast to vaccination with the L2 17-36 peptide or P25-P2C alone, a potent L2-specific antibody response was generated to the P25-P2C-HPV lipopeptide when delivered either s.c. or intranasally. Sera from mice vaccinated with the P25-P2C-HPV lipopeptide neutralized not only HPV16 pseudovirions but also other evolutionarily divergent oncogenic genital (HPV18, HPV45) and cutaneous (HPV5, BPV1) types. The L2-specific antibody response depended on MHC class II, CD40, and MyD88 signaling. Additionally, vaccination with the P25-P2C-HPV lipopeptide protected mice from homologous challenge with HPV16 pseudovirions at cutaneous and genital sites and heterologous challenge with HPV45 pseudovirions. If provided in the appropriate context, therefore, HPV16 L2 17-36 might be used in a totally synthetic cross-protective HPV vaccine.


Asunto(s)
Proteínas de la Cápside/inmunología , Reacciones Cruzadas/inmunología , Proteínas Oncogénicas Virales/inmunología , Vacunas contra Papillomavirus/química , Animales , Anticuerpos Antivirales , Formación de Anticuerpos , Antígenos Virales , Proteínas de la Cápside/uso terapéutico , Homólogo de la Proteína Chromobox 5 , Epítopos , Humanos , Lipoproteínas/uso terapéutico , Ratones , Pruebas de Neutralización , Proteínas Oncogénicas Virales/uso terapéutico , Vacunas contra Papillomavirus/inmunología , Fragmentos de Péptidos/uso terapéutico
3.
Am J Pathol ; 174(1): 136-43, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19095951

RESUMEN

The expression pattern of human papillomavirus (HPV) capsid antigen L2 is poorly described, and the significance of its localization with both promyelocytic leukemia protein (PML) and Daxx in a subnuclear domain, nuclear domain 10 (ND-10), when ectopically expressed in tissue culture cells is controversial. To address whether ND-10 localization of L2 occurs in natural cervical lesions, we used a HPV16 and HPV18 L2-specific monoclonal antibody (RG-1), in addition to rabbit antiserum to HPV6 L2, to localize L2. Immunohistochemical staining with RG-1 produced diffuse staining in the nuclei of some cells located within the superficial epithelial layers in eight of nine cases of HPV16/18(+) cervical intraepithelial neoplasia grade 1 (CIN1); however, no staining was observed in HPV16/18(+) high-grade CIN (0 of 8 cases), normal cervical epithelium (0 of 20 cases), cervical squamous cell carcinoma (0 of 102 cases), adenocarcinoma (0 of 51 cases), or adenosquamous carcinoma (0 of 6 cases). HPV16/18(+) cervical lesions that express L2 exhibit higher HPV16/18 genome copies per cell compared with those that do not positively stain with RG-1 (P = 0.04). RG-1 staining of HeLa cells transfected with L2 expression constructs was frequently concentrated in the ND-10, particularly in cells expressing high levels of L2, and co-localized with the cellular markers of ND-10, PML, and Daxx. In contrast, L2 was primarily diffuse within the nucleus and distinct from ND-10 as defined by PML immunofluorescent staining in CIN lesions, condylomata, and HPV16-transduced organotypic cultures.


Asunto(s)
Proteínas de la Cápside/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Anticuerpos Monoclonales , Western Blotting , Proteínas Co-Represoras , Femenino , Técnica del Anticuerpo Fluorescente , Prepucio , Células HeLa , Humanos , Inmunohistoquímica , Masculino , Chaperonas Moleculares , Proteínas Nucleares/metabolismo , Técnicas de Cultivo de Órganos , Proteína de la Leucemia Promielocítica , Transporte de Proteínas , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
4.
J Virol ; 82(9): 4638-46, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18305047

RESUMEN

Pseudovirions of human papillomavirus type 16 (HPV16), the principal etiologic agent in 50% of cervical cancers, were used as a model system to investigate the cell surface interactions involved in the exposure of the broadly cross-neutralizing papillomavirus L2 epitopes. These neutralizing epitopes were exposed only after cell surface binding and a subsequent change in capsid conformation that permitted cleavage by the cellular protease furin at a specific highly conserved site in L2 that is immediately upstream of the cross-neutralizing epitopes. Unexpectedly, binding of L2 antibodies led to the release of the capsid/antibody complexes from the cell surface and their accumulation on the extracellular matrix. Study of the dynamics of exposure of the L2 epitopes further revealed that representatives of the apparently dominant class of L1-specific neutralizing antibodies induced by virus-like particle vaccination prevent infection, not by preventing cell surface binding but rather by preventing the conformation change involved in exposure of the L2 neutralizing epitope. These findings suggest a dynamic model of virion-cell surface interactions that has implications for both evolution of viral serotypes and the efficacy of current and future HPV vaccines.


Asunto(s)
Anticuerpos Antivirales/inmunología , Reacciones Cruzadas/inmunología , Papillomavirus Humano 16/inmunología , Pruebas de Neutralización , Anticuerpos Monoclonales , Antígenos Virales , Proteínas de la Cápside/química , Proteínas de la Cápside/inmunología , Línea Celular , Membrana Celular/metabolismo , Epítopos , Matriz Extracelular , Humanos , Vacunas contra Papillomavirus/inmunología , Conformación Proteica , Vacunas
5.
Virol J ; 6: 176, 2009 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-19860897

RESUMEN

Vaccination of mice with minor capsid protein L2 or passive transfer with the L2-specific neutralizing monoclonal antibody RG-1 protects against human papillomavirus type 16 (HPV16) challenge. Here we explored the nature of the RG-1 epitope and its contribution to viral infectivity. RG-1 bound equivalently HPV16 L2 residues 17-36 with or without an intact C22-C28 disulphide bridge. HPV16 L2 mutations K20A, C22A, C22S, C28A, C28S, or P29A prevented RG-1 binding, whereas Y19A, K23A or Q24A had no impact. Mutation of either C22 or C28 to alanine or serine compromises HPV16 pseudoviral infectivity both in vitro and in the murine vaginal tract, but does not impact pseudovirion assembly. Despite their lack of infectivity, HPV16 pseudovirions containing C22S or C28S mutant L2 bind to cell surfaces, are taken up, and expose the 17-36 region on the virion surface as for wild type HPV16 pseudovirions suggesting normal furin cleavage of L2. Mutation of the second cysteine residue in Bovine papillomavirus type 1 (BPV1) L2 to serine (C25S) dramatically reduced the infectivity of BPV1 pseudovirions. Surprisingly, in contrast to the double mutation in HPV16 L2, the BPV1 L2 C19S, C25S double mutation reduced BPV1 pseudovirion infectivity of 293TT cells by only half.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Proteínas de la Cápside/antagonistas & inhibidores , Proteínas de la Cápside/fisiología , Epítopos/inmunología , Papillomavirus Humano 16/patogenicidad , Proteínas Oncogénicas Virales/antagonistas & inhibidores , Proteínas Oncogénicas Virales/fisiología , Sustitución de Aminoácidos/genética , Animales , Papillomavirus Bovino 1/genética , Papillomavirus Bovino 1/patogenicidad , Proteínas de la Cápside/inmunología , Cisteína/genética , Femenino , Células HeLa , Humanos , Ratones , Mutagénesis Sitio-Dirigida , Proteínas Oncogénicas Virales/inmunología , Infecciones por Papillomavirus/virología , Virulencia
6.
J Virol ; 81(24): 13927-31, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17928339

RESUMEN

We generated a monoclonal antibody, RG-1, that binds to highly conserved L2 residues 17 to 36 and neutralizes human papillomavirus 16 (HPV16) and HPV18. Passive immunotherapy with RG-1 was protective in mice. Antiserum to the HPV16 L2 peptide comprising residues 17 to 36 (peptide 17-36) neutralized pseudoviruses HPV5, HPV6, HPV16, HPV 18, HPV31, HPV 45, HPV 52, HPV 58, bovine papillomavirus 1, and HPV11 native virions. Depletion of HPV16 L2 peptide 17-36-reactive antibodies from cross-neutralizing rabbit and human L2-specific sera abolished cross-neutralization and drastically reduced neutralization of the cognate type. This cross-neutralization of diverse HPVs associated with cervical cancer, genital warts, and epidermodysplasia verruciformis suggests the possibility of a broadly protective, peptide-based vaccine.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Proteínas de la Cápside/inmunología , Proteínas Oncogénicas Virales/inmunología , Papillomaviridae/clasificación , Papillomaviridae/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Antivirales/administración & dosificación , Anticuerpos Antivirales/biosíntesis , Reacciones Cruzadas , Papillomavirus Humano 16/inmunología , Papillomavirus Humano 16/patogenicidad , Humanos , Inmunización Pasiva , Ratones , Datos de Secuencia Molecular , Pruebas de Neutralización , Infecciones por Papillomavirus/prevención & control
7.
Cancer Res ; 66(23): 11120-4, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17145854

RESUMEN

Oncogenic human papillomavirus (HPV) infection is a necessary cause of cervical cancer. Therefore, vaccination to prevent or eliminate HPV infection could reduce the incidence of cervical cancer. A fusion protein comprising HPV16 L2, E6, and E7 is a candidate combination preventive and therapeutic HPV vaccine. The L1- and L2-specific and neutralizing serum antibody titers and peripheral blood mononucleocyte antigen-specific proliferative responses generated by vaccination thrice at monthly intervals with HPV16 L2E7E6 were compared in two studies: a phase I randomized double-blind placebo controlled dose escalation trial in 40 healthy volunteers and a phase II trial of HPV16 L2E7E6 at the maximum dose in 29 women with high-grade anogenital intraepithelial neoplasia (AGIN). Vaccination of healthy volunteers induced L2-specific serum antibodies that were detected 1 month after the final vaccination (P(binomial) < 0.001). There was a significant trend to seroconversion for HPV16 and HPV18 neutralizing antibodies with increasing vaccine dose (P = 0.006 and P = 0.03, respectively). Seroconversion for HPV18 neutralizing antibodies showed a significant positive trend with increasing dose (P = 0.03) and was associated with seroconversion for HPV16 neutralizing antibodies (P(exact) = 0.04). The antigen-specific proliferative response of vaccinated healthy volunteers also showed a significant trend with increasing vaccine dose (P = 0.04). However, AGIN patients responded less effectively to vaccination than healthy patients for induction of HPV16 L2-specific antibody (P < 0.001) and proliferative responses (P < 0.001). Vaccination of healthy volunteers thrice with 533-mug HPV16 L2E7E6 at monthly intervals induced L2-specific serum antibodies that neutralized across papillomavirus species. Responses in AGIN patients were infrequent.


Asunto(s)
Anticuerpos Antivirales/sangre , Proteínas de la Cápside/inmunología , Proteínas Oncogénicas Virales/inmunología , Vacunas contra Papillomavirus/administración & dosificación , Proteínas Represoras/inmunología , Adulto , Neoplasias del Ano/sangre , Neoplasias del Ano/inmunología , Neoplasias del Ano/prevención & control , Proteínas de la Cápside/genética , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Reacciones Cruzadas/inmunología , Método Doble Ciego , Ensayo de Inmunoadsorción Enzimática , Femenino , Neoplasias de los Genitales Femeninos/sangre , Neoplasias de los Genitales Femeninos/inmunología , Neoplasias de los Genitales Femeninos/prevención & control , Papillomavirus Humano 16/inmunología , Papillomavirus Humano 18/inmunología , Humanos , Masculino , Persona de Mediana Edad , Pruebas de Neutralización , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus/sangre , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/prevención & control , Ensayos Clínicos Controlados Aleatorios como Asunto , Proteínas Recombinantes de Fusión/inmunología , Proteínas Represoras/genética , Factores de Tiempo , Vacunación/métodos
8.
Virology ; 476: 405-412, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25591175

RESUMEN

We reported previously on a vaccine approach that conferred apparent sterilizing immunity to SIVsmE660. The vaccine regimen employed a prime-boost using vectors based on recombinant vesicular stomatitis virus (VSV) and an alphavirus replicon expressing either SIV Gag or SIV Env. In the current study, we tested the ability of vectors expressing only the SIVsmE660 Env protein to protect macaques against the same high-dose mucosal challenge. Animals developed neutralizing antibody levels comparable to or greater than seen in the previous vaccine study. When the vaccinated animals were challenged with the same high-dose of SIVsmE660, all became infected. While average peak viral loads in animals were slightly lower than those of previous controls, the viral set points were not significantly different. These data indicate that Gag, or the combination of Gag and Env are required for the generation of apparent sterilizing immunity to the SIVsmE660 challenge.


Asunto(s)
Vacunas contra el SIDA/inmunología , Productos del Gen env/inmunología , Productos del Gen gag/inmunología , Membrana Mucosa/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Modelos Animales de Enfermedad , Productos del Gen env/administración & dosificación , Productos del Gen env/genética , Productos del Gen gag/administración & dosificación , Productos del Gen gag/genética , VIH/genética , VIH/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , Humanos , Macaca mulatta , Membrana Mucosa/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética
9.
Clin Vaccine Immunol ; 21(9): 1224-31, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24990902

RESUMEN

Immunization with human papillomavirus (HPV) L1 virus-like particles (VLPs) prevents infection with HPV. However, the expense and logistical demands of current VLP vaccines will limit their widespread use in resource-limited settings, where most HPV-induced cervical cancer occurs. Live oral adenovirus vaccines have properties that are well-suited for use in such settings. We have described a live recombinant adenovirus vaccine prototype that produces abundant HPV16 L1 protein from the adenovirus major late transcriptional unit and directs the assembly of HPV16 VLPs in tissue culture. Recombinant-derived VLPs potently elicit neutralizing antibodies in mice. Here, we characterize the immune response to the recombinant after dual oral and intranasal immunization of pigtail macaques, in which the virus replicates as it would in immunized humans. The immunization of macaques induced vigorous humoral responses to adenovirus capsid and nonstructural proteins, although, surprisingly, not against HPV L1. In contrast, immunization elicited strong T-cell responses to HPV VLPs as well as adenovirus virions. T-cell responses arose immediately after the primary immunization and were boosted by a second immunization with recombinant virus. T-cell immunity contributes to protection against a wide variety of pathogens, including many viruses. The induction of a strong cellular response by the recombinant indicates that live adenovirus recombinants have potential as vaccines for those agents. These studies encourage and will inform the continued development of viable recombinant adenovirus vaccines.


Asunto(s)
Adenovirus Humanos/genética , Portadores de Fármacos , Papillomavirus Humano 16/inmunología , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Administración Intranasal , Administración Oral , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Femenino , Papillomavirus Humano 16/genética , Macaca nemestrina , Infecciones por Papillomavirus/inmunología , Vacunas contra Papillomavirus/administración & dosificación , Vacunas contra Papillomavirus/genética , Linfocitos T/inmunología , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación , Vacunas de Partículas Similares a Virus/genética
10.
PLoS One ; 9(10): e109678, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25360552

RESUMEN

Identification of transmitted/founder simian immunodeficiency virus (SIV) envelope sequences responsible for infection may prove critical for understanding HIV/SIV mucosal transmission. We used single genome amplification and phylogenetic analyses to characterize transmitted/founder SIVs both in the inoculum and in immunized-infected rhesus monkeys. Single genome amplification of the SIVsmE660 inoculum revealed a maximum diversity of 1.4%. We also noted that the consensus sequence of the challenge stock differed from the vaccine construct in 10 amino acids including 3 changes in the V4 loop. Viral env was prepared from rhesus plasma in 3 groups of 6 immunized with vesicular stomatitis virus (VSV) vectors and boosted with Semliki forest virus (SFV) replicons expressing (a) SIVsmE660 gag-env (b) SIVsmE660 gag-env plus rhesus GM-CSF and (c) control influenza hemagglutinin protein. Macaques were immunized twice with VSV-vectors and once with SFV vector and challenged intrarectally with 4000 TCID50. Single genome amplification characterized the infections of 2 unprotected animals in the gag-env immunized group, both of which had reduced acute plasma viral loads that ended as transient infections indicating partial immune control. Four of 6 rhesus were infected in the gag-env + GM-CSF group which demonstrated that GM-CSF abrogated protection. All 6 animals from the control group were infected having high plasma viral loads. We obtained 246 full-length envelope sequences from SIVsmE660 infected macaques at the peak of infection and determined the number of transmitted/founder variants per animal. Our analysis found that 2 of 2 gag-env vaccinated but infected macaques exhibited single but distinct virus envelope lineages whereas rhesus vaccinated with gag-env-GM-CSF or HA control exhibited both single and multiple env lineages. Because there were only 2 infected animals in the gag-env vaccinated rhesus compared to 10 infected rhesus in the other 2 groups, the significance of finding single env variants in the gag-env vaccinated group could not be established.


Asunto(s)
Macaca mulatta/virología , Filogenia , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Proteínas del Envoltorio Viral/genética , Animales , Productos del Gen env/genética , Productos del Gen env/inmunología , Productos del Gen gag/genética , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Inmunización , Macaca mulatta/inmunología , Datos de Secuencia Molecular , Vacunas contra el SIDAS/inmunología , Virus de los Bosques Semliki/genética , Virus de los Bosques Semliki/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/transmisión , Virus de la Inmunodeficiencia de los Simios/genética , Virus de la Inmunodeficiencia de los Simios/inmunología , Estomatitis Vesicular/virología , Proteínas del Envoltorio Viral/inmunología , Carga Viral
11.
J Gen Virol ; 90(Pt 1): 136-43, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19088282

RESUMEN

Evidence of a possible association of cutaneous human papillomavirus (HPV) types, especially members of the genus Betapapillomavirus, and the development of non-melanoma skin cancer (NMSC) is accumulating. Vaccination with virus-like particles (VLPs) consisting of self-assembled L1, the major capsid protein, has been introduced to control anogenital HPV infection. This study examined the serological relationship between betapapillomavirus (beta-PV) types 5 and 8 and the new type HPV-92, which has recently been isolated from a basal cell carcinoma containing a high number of viral genomes. Following expression by recombinant baculoviruses, the L1 protein of HPV-92 self-assembled into VLPs that elicited high-titre antibodies after immunization, similar to VLPs from HPV-5 and -8. Haemagglutination inhibition (HAI) assays were used as a surrogate method for the detection of virion-neutralizing antibodies, which correlates with protection from infection. Antisera raised against HPV-5 and -8 VLPs displayed HAI activity not only against the homologous type, but also against heterologous HPV types 5, 8 and 92, whereas HAI activity of antisera against HPV-92 VLP was restricted to the homologous type. The results of neutralization assays using HPV-5 pseudovirions were consistent with those from HAI assays. Cross-neutralizing immune responses by VLP vaccination against heterologous HPV types may provide broader protection against the multiplicity of HPV types detected in NMSC. If a close link to HPV infection can be conclusively established, these results may provide a basis for further evaluation of VLPs of beta-PVs as candidates for a prophylactic skin-type HPV vaccine, aimed at reducing the incidence of NMSC.


Asunto(s)
Anticuerpos Antivirales/inmunología , Betapapillomavirus/inmunología , Proteínas de la Cápside/inmunología , Proteínas Oncogénicas Virales/inmunología , Betapapillomavirus/aislamiento & purificación , Reacciones Cruzadas , Pruebas de Inhibición de Hemaglutinación , Humanos , Pruebas de Neutralización , Virosomas/inmunología
12.
Vaccine ; 27(7): 1040-9, 2009 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-19095032

RESUMEN

A vaccine comprising human papillomavirus type 16 (HPV16) L2, E6 and E7 in a single tandem fusion protein (termed TA-CIN) has the potential advantages of both broad cross-protection against HPV transmission through induction of L2 antibodies able to cross neutralize different HPV types and of therapy by stimulating T cell responses targeting HPV16 early proteins. However, patients vaccinated with TA-CIN alone develop weak HPV neutralizing antibody and E6/E7-specific T cell responses. Here we test TA-CIN formulated along with the adjuvant GPI-0100, a semi-synthetic quillaja saponin analog that was developed to promote both humoral and cellular immune responses. Subcutaneous administration to mice of TA-CIN (20 microg) with 50microg GPI-0100, three times at biweekly intervals, elicited high titer HPV16 neutralizing serum antibody, robust neutralizing titers for other HPV16-related types, including HPV31 and HPV58, and neutralized to a lesser extent other genital mucosatropic papillomaviruses like HPV18, HPV45, HPV6 and HPV11. Notably, vaccination with TA-CIN in GPI-0100 protected mice from cutaneous HPV16 challenge as effectively as HPV16 L1 VLP without adjuvant. Formulation of TA-CIN with GPI-0100 enhanced the production of E7-specific, interferon gamma producing CD8(+) T cell precursors by 20-fold. Vaccination with TA-CIN in GPI-0100 also completely prevented tumor growth after challenge with 5x10(4) HPV16-transformed TC-1 tumor cells, whereas vaccination with TA-CIN alone delayed tumor growth. Furthermore, three monthly vaccinations with 125 microg of TA-CIN and 1000 microg GPI-0100 were well tolerated by pigtail macaques and induced both HPV16 E6/E7-specific T cell responses and serum antibodies that neutralized all HPV types tested.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Proteínas de la Cápside/inmunología , Proteínas Oncogénicas Virales/inmunología , Vacunas contra Papillomavirus/inmunología , Proteínas Recombinantes de Fusión/inmunología , Proteínas Represoras/inmunología , Saponinas/farmacología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antivirales/sangre , Linfocitos T CD8-positivos/inmunología , Proteínas de la Cápside/genética , Citocinas/metabolismo , Femenino , Humanos , Inmunización Secundaria/métodos , Inyecciones Subcutáneas , Macaca , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/efectos adversos , Vacunas contra Papillomavirus/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/efectos adversos , Proteínas Recombinantes de Fusión/genética , Proteínas Represoras/genética , Saponinas/administración & dosificación
13.
J Natl Cancer Inst ; 101(11): 782-92, 2009 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-19470949

RESUMEN

BACKGROUND: Vaccination with minor capsid protein L2 induces antibodies that cross-neutralize diverse papillomavirus types. However, neutralizing antibody titers against the papillomavirus type from which the L2 vaccine was derived are generally higher than the titers against heterologous types, which could limit effectiveness against heterologous types. We hypothesized that vaccination with concatenated multitype L2 fusion proteins derived from known cross-protective epitopes of several divergent human papillomavirus (HPV) types might enhance immunity across clinically relevant HPV genotypes. METHODS: Antibody responses of mice (n = 120) and rabbits (n = 23) to vaccination with HPV-16 amino-terminal L2 polypeptides or multitype L2 fusion proteins, namely, 11-200 x 3 (HPV types 6, 16, 18), 11-88 x 5 (HPV types 1, 5, 6, 16, 18), or 17-36 x 22 (five cutaneous, two mucosal low-risk, and 15 oncogenic types), that were formulated alone or in GPI-0100, alum, or 1018 ISS adjuvants were compared with vaccination with L1 virus-like particles (VLPs), including Gardasil, a licensed quadrivalent HPV L1 vaccine, and a negative control. Mice were challenged with HPV-16 pseudovirions 4 months after vaccination. Statistical tests were two-sided. RESULTS: The HPV-16 L2 polypeptides generated robust HPV-16-neutralizing antibody responses, albeit lower than those to HPV-16 L1 VLPs, and lower responses against other HPVs. In contrast, vaccination with the multitype L2 fusion proteins 11-200 x 3 and 11-88 x 5 induced high serum neutralizing antibody titers against all heterologous HPVs tested. 11-200 x 3 formulated in GPI-0100 adjuvant or alum with 1018 ISS protected mice against HPV-16 challenge (reduction in HPV-16 infection vs phosphate-buffered saline control, P < .001) 4 months after vaccination as well as HPV-16 L1 VLPs, but 11-200 x 3 alone or formulated with either alum or 1018 ISS was less effective (reduction in HPV-16 infection, P < .001). CONCLUSION: Concatenated multitype L2 proteins in adjuvant have potential as pan-oncogenic HPV vaccines.


Asunto(s)
Anticuerpos Antivirales/inmunología , Linfocitos B/inmunología , Proteínas de la Cápside/inmunología , Papillomavirus Humano 16/inmunología , Pruebas de Neutralización , Proteínas Oncogénicas Virales/inmunología , Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus/inmunología , Análisis de Varianza , Animales , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/sangre , Proteínas de la Cápside/uso terapéutico , Ensayo de Inmunoadsorción Enzimática , Epítopos/inmunología , Femenino , Vacuna Tetravalente Recombinante contra el Virus del Papiloma Humano Tipos 6, 11 , 16, 18 , Humanos , Sueros Inmunes , Ratones , Ratones Endogámicos BALB C , Pruebas de Neutralización/métodos , Proteínas Oncogénicas Virales/uso terapéutico , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/virología , Vacunas contra Papillomavirus/uso terapéutico , Conejos , Factores de Tiempo , Virión
14.
Vaccine ; 26(3): 351-60, 2008 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-18096279

RESUMEN

Cervical cancer is one of the most common cancers in women worldwide. Persistent infection with human papillomavirus (HPV) is considered to be the etiological factor for cervical cancer. Therefore, an effective vaccine against HPV infections may lead to the control of cervical cancer. An ideal HPV vaccine should aim to generate both humoral immune response to prevent new infections as well as cell-mediated immunity to eliminate established infection or HPV-related disease. In the current study, we have generated a potential preventive and therapeutic HPV DNA vaccine using human calreticulin (CRT) linked to HPV16 early proteins, E6 and E7 and the late protein L2 (hCRTE6E7L2). We found that vaccination with hCRTE6E7L2 DNA vaccine induced a potent E6/E7-specific CD8+ T cell immune response, resulting in a significant therapeutic effect against E6/E7 expressing tumor cells. In addition, vaccination with hCRTE6E7L2 DNA generated significant L2-specific neutralizing antibody responses, protecting against pseudovirion infection. Thus, the hCRTE6E7L2 DNA vaccines are capable of generating potent preventive and therapeutic effects in vaccinated mice. Our data has significant clinical implications.


Asunto(s)
Infecciones por Papillomavirus/prevención & control , Vacunas contra Papillomavirus , Infecciones Tumorales por Virus/prevención & control , Vacunas de ADN , Animales , Anticuerpos Antivirales/sangre , Biolística , Linfocitos T CD8-positivos/inmunología , Calreticulina/genética , Calreticulina/inmunología , Proteínas de la Cápside/genética , Proteínas de la Cápside/inmunología , Línea Celular , Cricetinae , Diseño de Fármacos , Femenino , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/inmunología , Papillomaviridae/genética , Papillomaviridae/inmunología , Proteínas E7 de Papillomavirus , Infecciones por Papillomavirus/inmunología , Vacunas contra Papillomavirus/administración & dosificación , Vacunas contra Papillomavirus/genética , Vacunas contra Papillomavirus/inmunología , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Infecciones Tumorales por Virus/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Vacunas de ADN/inmunología
15.
Vaccine ; 25(17): 3501-10, 2007 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-16914239

RESUMEN

Immunization against human papillomavirus (HPV) infection promises to reduce the worldwide burden of cervical cancer. To evaluate the potential of live recombinant adenoviruses for induction of HPV infection-blocking immunity, we prepared viable adenovirus recombinants that express the HPV16 L1 gene from the adenovirus major late transcriptional unit. Adenovirus-produced HPV16 L1 assembles into virus-like particles (VLPs) in infected cells in culture. Purified HPV16 VLPs are recognized by HPV16 neutralizing antibodies and induce high neutralizing titers when injected intraperitoneally into mice. Canine oral papillomavirus VLPs derived from previously described recombinants also induce strong antibody responses in mice. These data support our suggestion that viable adenovirus recombinants will be able to induce protective immunity to papillomavirus infection during replication in human vaccinees.


Asunto(s)
Anticuerpos Antivirales/sangre , Proteínas de la Cápside/inmunología , Proteínas Oncogénicas Virales/inmunología , Vacunas contra Papillomavirus/inmunología , Vacunas Sintéticas/inmunología , Adenoviridae/genética , Animales , Proteínas de la Cápside/genética , Femenino , Inmunización , Ratones , Ratones Endogámicos BALB C , Proteínas Oncogénicas Virales/genética , Proteínas Recombinantes/inmunología , Virión/inmunología
16.
J Virol ; 81(21): 11585-92, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17715230

RESUMEN

Current L1 virus-like particle (VLP) vaccines provide type-restricted protection against a small subset of the human papillomavirus (HPV) genotypes associated with cervical cancer, necessitating continued cytologic screening of vaccinees. Cervical cancer is most problematic in countries that lack the resources for screening or highly multivalent HPV VLP vaccines, suggesting the need for a low-cost, broadly protective vaccinogen. Here, N-terminal L2 polypeptides comprising residues 1 to 88 or 11 to 200 derived from HPV16, bovine papillomavirus type 1 (BPV1), or cottontail rabbit papillomavirus (CRPV) were produced in bacteria. Rabbits were immunized with these N-terminal L2 polypeptides and concurrently challenged with CRPV and rabbit oral papillomavirus (ROPV). Vaccination with either N-terminal L2 polypeptides of CRPV effectively protected rabbits from CRPV challenge but not from papillomas induced by cutaneous challenge with CRPV genomic DNA. Furthermore, papillomas induced by CRPV genomic DNA deficient for L2 expression grew at the same rate as those induced by wild-type CRPV genomic DNA, further suggesting that the L2 polypeptide vaccines lack therapeutic activity. Neutralizing serum antibody titers of >15 correlated with protection (P < 0.001), a finding consistent with neutralizing antibody-mediated protection. Surprisingly, a remarkable degree of protection against heterologous papillomavirus types was observed after vaccination with N-terminal L2 polypeptides. Notably, vaccination with HPV16 L2 11-200 protected against cutaneous and mucosal challenge with CRPV and ROPV, respectively, papillomaviruses that are evolutionarily divergent from HPV16. Further, vaccination with HPV16 L2 11-200 generates broadly cross-neutralizing serum antibody, suggesting the potential of L2 as a second-generation preventive HPV vaccine antigen.


Asunto(s)
Proteínas de la Cápside/química , Proteínas Oncogénicas Virales/química , Vacunas Virales/química , Animales , Proteínas de la Cápside/metabolismo , ADN Viral/genética , ADN Viral/metabolismo , Femenino , Genotipo , Pruebas de Neutralización , Proteínas Oncogénicas Virales/metabolismo , Papiloma/prevención & control , Papiloma/virología , Papillomaviridae/metabolismo , Péptidos/química , Estructura Terciaria de Proteína , Conejos , Piel/virología , Neoplasias del Cuello Uterino/prevención & control , Neoplasias del Cuello Uterino/virología
17.
Vaccine ; 25(11): 2001-10, 2007 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-17239496

RESUMEN

Peptides of the papillomavirus L2 minor capsid protein can induce antibodies (Ab) that neutralize a broad range of human papillomavirus (HPV) genotypes. Unfortunately, L2 is antigenically subdominant to L1 in the virus capsid. To induce a strong anti-L2 Ab response with cross-neutralizing activity to other mucosal types, chimeric virus-like particles (VLP) were generated in which HPV16 L2 neutralization epitopes (comprising L2 residues 69-81 or 108-120) are inserted within an immunodominant surface loop (between residues 133 and 134) of the L1 major capsid protein of bovine papillomavirus type 1 (BPV1). These chimeras self-assembled into pentameric capsomers, or complete VLP similar to wild type (wt) L1 protein. Immunization of rabbits with assembled particle preparations induced L2-specific serum Ab with titers 10-fold higher than those induced by cognate synthetic L2 peptides coupled to KLH. Antisera to both chimeric proteins partially neutralized HPV16 pseudovirions, confirming that both HPV16 L2 peptides define neutralization epitopes. When analyzed for the ability to cross-neutralize infection by authentic HPV11 virions, using detection of early viral RNA by RT-PCR-assays as the readout, immune serum to chimeric protein comprising L2 residues 69-81, but not 108-120, was partially neutralizing. In addition, mouse-antiserum induced by vaccinations with synthetic L2 peptide 108-120, but not 69-81, was partially neutralizing in this assay. Induction of cross-neutralization Ab by L2 epitopes displayed on chimeric VLP represents a possible strategy for the generation of broad-spectrum vaccines to protect against relevant mucosal HPV and associated neoplasia.


Asunto(s)
Proteínas de la Cápside/inmunología , Epítopos/inmunología , Papillomavirus Humano 11/inmunología , Papillomavirus Humano 16/inmunología , Proteínas Oncogénicas Virales/inmunología , Vacunas contra Papillomavirus/inmunología , Animales , Anticuerpos Antivirales/sangre , Papillomavirus Bovino 1/genética , Proteínas de la Cápside/genética , Reacciones Cruzadas , Epítopos/genética , Vectores Genéticos , Humanos , Ratones , Modelos Animales , Pruebas de Neutralización , Proteínas Oncogénicas Virales/genética , Vacunas contra Papillomavirus/genética , Conejos , Vacunas de Virosoma/inmunología
18.
Vaccine ; 25(17): 3302-10, 2007 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-17291642

RESUMEN

The E6 and E7 oncoproteins of the high-risk HPV type16 represent ideal targets for HPV vaccine development, they being consistently expressed in cervical cancer lesions. Since HPV-16 is primarily transmitted through genital mucosal route, mucosal immune responses constitute an essential feature for vaccination strategies against HPV-associated lesions. We present here evidence showing that mucosal immunization of mice by the intranasal route with a mixture of peptides E7(44-62) and E6(43-57) from the E7 and E6 oncoproteins of HPV-16, respectively, using a mutant cholera toxin adjuvant (CT-2*), primed strong antigen-specific cellular immune responses in systemic and mucosal tissues. Significant levels of IFN-gamma production by both CD4 and CD8 cells were observed along with CTL responses that were effective against both peptide-pulsed targets as well as syngeneic tumor cells (TC-1) expressing the cognate E6 and E7 proteins. Furthermore, mice immunized with the peptide mixture and CT-2* effectively resisted TC-1 tumor challenge. These results together with our earlier observations that T cell responses to these peptides correlate with recurrence-free survival in women after ablative treatment for HPV-associated cervical intraepithelial neoplasia, support the potential of these E6 and E7 peptides for inclusion in vaccine formulations.


Asunto(s)
Proteínas Oncogénicas Virales/inmunología , Vacunas contra Papillomavirus/inmunología , Fragmentos de Péptidos/inmunología , Proteínas Represoras/inmunología , Neoplasias del Cuello Uterino/prevención & control , Vacunas Sintéticas/inmunología , Administración Intranasal , Secuencia de Aminoácidos , Animales , Toxina del Cólera/administración & dosificación , Femenino , Inmunidad Mucosa , Interferón gamma/biosíntesis , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Proteínas E7 de Papillomavirus , Vacunas contra Papillomavirus/administración & dosificación , Linfocitos T Citotóxicos/inmunología
19.
J Virol ; 79(11): 6723-31, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15890910

RESUMEN

The papillomavirus capsid mediates binding to the cell surface and passage of the virion to the perinuclear region during infection. To better understand how the virus traffics across the cell, we sought to identify cellular proteins that bind to the minor capsid protein L2. We have identified syntaxin 18 as a protein that interacts with bovine papillomavirus type 1 (BPV1) L2. Syntaxin 18 is a target membrane-associated soluble N-ethylmaleimide-sensitive factor-attachment protein receptor (tSNARE) that resides in the endoplasmic reticulum (ER). The ectopic expression of FLAG-tagged syntaxin 18, which disrupts ER trafficking, blocked BPV1 pseudovirion infection. Furthermore, the expression of FLAG-syntaxin 18 prevented the passage of BPV1 pseudovirions to the perinuclear region that is consistent with the ER. Genetic studies identified a highly conserved L2 domain, DKILK, comprising residues 40 to 44 that mediated BPV1 trafficking through the ER during infection via an interaction with the tSNARE syntaxin 18. Mutations within the DKILK motif of L2 that did not significantly impact virion morphogenesis or binding at the cell surface prevented the L2 interaction with syntaxin 18 and disrupted BPV1 infection.


Asunto(s)
Papillomavirus Bovino 1/fisiología , Papillomavirus Bovino 1/patogenicidad , Proteínas de la Cápside/fisiología , Proteínas de la Membrana/fisiología , Infecciones por Papillomavirus/etiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Sitios de Unión/genética , Papillomavirus Bovino 1/genética , Papillomavirus Bovino 1/ultraestructura , Células COS , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Bovinos , Línea Celular , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Humanos , Técnicas In Vitro , Proteínas de la Membrana/genética , Microscopía Electrónica , Datos de Secuencia Molecular , Mutagénesis Sitio-Dirigida , Infecciones por Papillomavirus/virología , Unión Proteica , Proteínas Qa-SNARE , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección , Virulencia , Ensamble de Virus
20.
Virology ; 337(2): 365-72, 2005 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-15885736

RESUMEN

Vaccination with papillomavirus L2 has been shown to induce neutralizing antibodies that protect against homologous type infection and cross-neutralize a limited number of genital HPVs. Surprisingly, we found that antibodies to bovine papillomavirus (BPV1) L2 amino acids 1-88 induced similar titers of neutralizing antibodies against Human papillomavirus (HPV)16 and 18 and BPV1 pseudoviruses and also neutralized HPV11 native virions. These antibodies also neutralized each of the other pseudovirus types tested, HPV31, HPV6 and Cottontail rabbit papillomavirus (CRPV) pseudoviruses, albeit with lower titers. HPV16, HPV18, HPV31, HPV6 and CRPV L2 anti-sera also displayed some cross-neutralization, but the titers were lower and did not encompass all pseudoviruses tested. This study demonstrates the presence of broadly cross-neutralizing epitopes at the N-terminus of L2 that are shared by cutaneous and mucosal types and by types that infect divergent species. BPV1 L2 was exceptionally effective at inducing cross-neutralizing antibodies to these shared epitopes.


Asunto(s)
Proteínas de la Cápside/genética , Proteínas Oncogénicas Virales/genética , Papillomaviridae/genética , Papillomaviridae/inmunología , Infecciones por Papillomavirus/inmunología , Secuencia de Aminoácidos , Proteínas de la Cápside/inmunología , Humanos , Datos de Secuencia Molecular , Pruebas de Neutralización , Proteínas Oncogénicas Virales/inmunología , Sistemas de Lectura Abierta , Alineación de Secuencia , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA