Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Pharm Sci ; 105(7): 2032-41, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27233688

RESUMEN

A recombinant Clostridium difficile expression system was used to produce genetically engineered toxoids A and B as immunogens for a prophylactic vaccine against C. difficile-associated disease. Although all known enzymatic activities responsible for cytotoxicity were genetically abrogated, the toxoids exhibited residual cytotoxic activity as measured in an in vitro cell-based cytotoxicity assay. The residual cytotoxicity was eliminated by treating the toxoids with 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) and N-hydroxysuccinimide. Mass spectrometry and amino acid analysis of the EDC-inactivated toxoids identified crosslinks, glycine adducts, and ß-alanine adducts. Surface plasmon resonance analysis demonstrated that modifications resulting from the chemical treatment did not appreciably affect recognition of epitopes by both toxin A- and B-specific neutralizing monoclonal antibodies. Compared to formaldehyde-inactivated toxoids, the EDC/N-hydroxysuccinimide-inactivated toxoids exhibited superior stability in solution with respect to reversion of cytotoxic activity.


Asunto(s)
Clostridioides difficile/química , Clostridioides difficile/genética , Ingeniería de Proteínas/métodos , Toxoides/química , Toxoides/genética , Animales , Proteínas Bacterianas/química , Toxinas Bacterianas/química , Vacunas Bacterianas , Supervivencia Celular/efectos de los fármacos , Estabilidad de Medicamentos , Enterotoxinas/química , Epítopos , Etildimetilaminopropil Carbodiimida/química , Inmunización , Mesocricetus , Proteínas Recombinantes , Succinimidas/química , Resonancia por Plasmón de Superficie
2.
J Virol ; 88(12): 6690-701, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24696472

RESUMEN

UNLABELLED: In previous work, a prototypic recombinant vesicular stomatitis virus Indiana serotype (rVSIV) vector expressing simian immunodeficiency virus (SIV) gag and human immunodeficiency virus type 1 (HIV-1) env antigens protected nonhuman primates (NHPs) from disease following challenge with an HIV-1/SIV recombinant (SHIV). However, when tested in a stringent NHP neurovirulence (NV) model, this vector was not adequately attenuated for clinical evaluation. For the work described here, the prototypic rVSIV vector was attenuated by combining specific G protein truncations with either N gene translocations or mutations (M33A and M51A) that ablate expression of subgenic M polypeptides, by incorporation of temperature-sensitive mutations in the N and L genes, and by deletion of the VSIV G gene to generate a replicon that is dependent on trans expression of G protein for in vitro propagation. When evaluated in a series of NHP NV studies, these attenuated rVSIV variants caused no clinical disease and demonstrated a very significant reduction in neuropathology compared to wild-type VSIV and the prototypic rVSIV vaccine vector. In spite of greatly increased in vivo attenuation, some of the rVSIV vectors elicited cell-mediated immune responses that were similar in magnitude to those induced by the much more virulent prototypic vector. These data demonstrate novel approaches to the rational attenuation of VSIV NV while retaining vector immunogenicity and have led to identification of an rVSIV N4CT1gag1 vaccine vector that has now successfully completed phase I clinical evaluation. IMPORTANCE: The work described in this article demonstrates a rational approach to the attenuation of vesicular stomatitis virus neurovirulence. The major attenuation strategy described here will be most likely applicable to other members of the Rhabdoviridae and possibly other families of nonsegmented negative-strand RNA viruses. These studies have also enabled the identification of an attenuated, replication-competent rVSIV vector that has successfully undergone its first clinical evaluation in humans. Therefore, these studies represent a major milestone in the development of attenuated rVSIV, and likely other vesiculoviruses, as a new vaccine platform(s) for use in humans.


Asunto(s)
Vacunas contra el SIDA/inmunología , Sistema Nervioso Central/virología , Vectores Genéticos/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Macaca fascicularis , Virus de la Estomatitis Vesicular Indiana/inmunología , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Animales , Anticuerpos Antivirales/inmunología , Sistema Nervioso Central/inmunología , Modelos Animales de Enfermedad , Vectores Genéticos/genética , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , VIH-1/genética , Humanos , Macaca fascicularis/genética , Macaca fascicularis/inmunología , Macaca fascicularis/virología , Masculino , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Virus de la Estomatitis Vesicular Indiana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/administración & dosificación , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología
3.
J Infect Dis ; 208(2): 319-29, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23596321

RESUMEN

BACKGROUND: We evaluated the immunological responses of African green monkeys immunized with multiple F and G protein-based vaccines and assessed protection against the Memphis 37 strain of respiratory syncytial virus (RSV). METHODS: Monkeys were immunized with F and G proteins adjuvanted with immunostimulatory (CpG) oligodeoxyribonucleotides admixed with either Alhydrogel or ISCOMATRIX adjuvant. Delivery of F and G proteins via replication incompetent recombinant vesicular stomatitis viruses (VSVs) and human adenoviruses was also evaluated. Mucosally or parenterally administered recombinant adenoviruses were used in prime-boost regimens with adjuvanted proteins or recombinant DNA. RESULTS: Animals primed by intranasal delivery of recombinant adenoviruses, and boosted by intramuscular injection of adjuvanted F and G proteins, developed neutralizing antibodies and F/G protein-specific T cells and were protected from RSV infection. Intramuscular injections of Alhydrogel (plus CpG) adjuvanted F and G proteins reduced peak viral loads in the lungs of challenged monkeys. Granulocyte numbers were not significantly elevated, relative to controls, in postchallenge bronchoalveolar lavage samples from vaccinated animals. CONCLUSIONS: This study has validated the use of RSV (Memphis 37) in an African green monkey model of intranasal infection and identified nonreplicating vaccines capable of eliciting protection in this higher species challenge model.


Asunto(s)
Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/farmacología , Virus Sincitiales Respiratorios/inmunología , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Adyuvantes Inmunológicos/farmacología , Animales , Anticuerpos Antivirales/inmunología , Lavado Broncoalveolar/métodos , Chlorocebus aethiops , Granulocitos/inmunología , Granulocitos/virología , Inmunización/métodos , Pulmón/inmunología , Pulmón/virología , Distribución Aleatoria , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Vacunas contra Virus Sincitial Respiratorio/genética , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/genética , Linfocitos T/inmunología , Linfocitos T/virología , Vesiculovirus/genética , Vesiculovirus/inmunología , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología , Carga Viral/inmunología , Proteínas Virales/genética , Proteínas Virales/inmunología , Replicación Viral/genética , Replicación Viral/inmunología
4.
Immunol Lett ; 150(1-2): 134-44, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23261719

RESUMEN

Respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract illness in infants, the elderly, and other high-risk individuals. Despite years of research in this field, there is no effective licensed vaccine to prevent RSV infection. We have generated candidate RSV vaccines using a recombinant vesicular stomatitis virus (rVSV) replicon in which the attachment and fusion domains of the VSV glycoprotein (G) have been deleted (rVSV-Gstem), rendering the virus propagation-defective except in the presence of complementing VSV G provided in trans. A form of this vector encoding the RSV fusion protein (F) gene expressed high levels of F in vitro and elicited durable neutralizing antibody responses as well as complete protection against RSV challenge in vivo. Mice vaccinated with rVSV-Gstem-RSV-F replicons also developed robust cellular responses characterized by both primary and memory Th1-biased CD8+ and CD4+ T cells. Furthermore, a single high dose of the Gstem-RSV-F replicon was effective against challenge with both RSV A and B subgroup viruses. Finally, addition of an RSV glycoprotein (G)-expressing Gstem vector significantly improved the incomplete protection achieved with a single low dose of Gstem-RSV-F vector alone.


Asunto(s)
Vectores Genéticos/genética , Vectores Genéticos/inmunología , Inmunidad Celular , Inmunidad Humoral , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/inmunología , Vesiculovirus/genética , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Femenino , Orden Génico , Vectores Genéticos/administración & dosificación , Humanos , Inmunización , Memoria Inmunológica , Ratones , Infecciones por Virus Sincitial Respiratorio/prevención & control , Células TH1/inmunología , Proteínas Virales de Fusión/genética , Proteínas Virales de Fusión/inmunología
5.
J Virol Methods ; 164(1-2): 43-50, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19941901

RESUMEN

Propagation-defective vesicular stomatitis virus (VSV) vectors that encode a truncated G protein (VSV-Gstem) or lack the G gene entirely (VSV-DeltaG) are attractive vaccine vectors because they are immunogenic, cannot replicate and spread after vaccination, and do not express many of the epitopes that elicit neutralizing anti-VSV immunity. To consider advancing non-propagating VSV vectors towards clinical assessment, scalable technology that is compliant with human vaccine manufacturing must be developed to produce clinical trial material. Accordingly, two propagation methods were developed for VSV-Gstem and VSV-DeltaG vectors encoding HIV gag that have the potential to support large-scale production. One method is based on transient expression of G protein after electroporating plasmid DNA into Vero cells and the second is based on a stable Vero cell line that contains a G gene controlled by a heat shock-inducible transcription unit. Both methods reproducibly supported production of 1 x 10(7) to 1 x 10(8) infectious units (I.U.s) of vaccine vector per milliliter. Results from these studies also showed that optimization of the G gene is necessary for abundant G protein expression from electroporated plasmid DNA or from DNA integrated in the genome of a stable cell line, and that the titers of VSV-Gstem vectors generally exceeded VSV-DeltaG.


Asunto(s)
Vectores Genéticos , Glicoproteínas de Membrana/deficiencia , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/genética , Proteínas del Envoltorio Viral/deficiencia , Animales , Chlorocebus aethiops , Glicoproteínas de Membrana/biosíntesis , Datos de Secuencia Molecular , ARN Viral/genética , Análisis de Secuencia de ADN , Células Vero , Proteínas del Envoltorio Viral/biosíntesis , Cultivo de Virus/métodos , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética
6.
Vaccine ; 27(22): 2930-9, 2009 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-19428903

RESUMEN

Recombinant vesicular stomatitis viruses (rVSVs) are being developed as potential HIV-1 vaccine candidates. To characterize the in vivo replication and dissemination of rVSV vectors in mice, high doses of a highly attenuated vector expressing HIV-1 Gag, rVSV(IN)-N4CT9-Gag1, and a prototypic reference virus, rVSV(IN)-HIVGag5, were delivered intramuscularly (IM), intranasally (IN), or intravenously (IV). We used quantitative, real-time RT-PCR (Q-PCR) and standard plaque assays to measure the temporal dissemination of these viruses to various tissues. Following IM inoculation, both viruses were detected primarily at the injection site as well as in draining lymph nodes; neither virus induced significant weight loss, pathologic signs, or evidence of neuroinvasion. In contrast, following IN inoculation, the prototypic virus was detected in all tissues tested and caused significant weight loss leading to death. IN administration of rVSV(IN)-N4CT9-Gag1 resulted in detection in numerous tissues (brain, lung, nasal turbinates, and lymph nodes) albeit in significantly reduced levels, which caused little or no weight loss nor any mortality. Following IV inoculation, both prototypic and attenuated viruses were detected by Q-PCR in all tissues tested. In contrast to the prototype, rVSV(IN)-N4CT9-Gag1 viral loads were significantly lower in all organs tested, and no infectious virus was detected in the brain following IV inoculation, despite the presence of viral RNA. These studies demonstrated significant differences in the biodistribution patterns of and the associated pathogenicity engendered by the prototypic and attenuated vectors in a highly susceptible host.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/farmacocinética , Vectores Genéticos , Vesiculovirus/crecimiento & desarrollo , Vesiculovirus/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Vacunas contra el SIDA/efectos adversos , Administración Intranasal , Animales , Femenino , Inyecciones Intramusculares , Inyecciones Intravenosas , Ratones , Ratones Endogámicos BALB C , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/farmacocinética , Ensayo de Placa Viral
7.
J Virol ; 82(1): 207-19, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17942549

RESUMEN

Recombinant vesicular stomatitis virus (rVSV) has shown great potential as a new viral vector for vaccination. However, the prototypic rVSV vector described previously was found to be insufficiently attenuated for clinical evaluation when assessed for neurovirulence in nonhuman primates. Here, we describe the attenuation, neurovirulence, and immunogenicity of rVSV vectors expressing human immunodeficiency virus type 1 Gag. These rVSV vectors were attenuated by combinations of the following manipulations: N gene translocations (N4), G gene truncations (CT1 or CT9), noncytopathic M gene mutations (Mncp), and positioning of the gag gene into the first position of the viral genome (gag1). The resulting N4CT1-gag1, N4CT9-gag1, and MncpCT1-gag1 vectors demonstrated dramatically reduced neurovirulence in mice following direct intracranial inoculation. Surprisingly, in spite of a very high level of attenuation, the N4CT1-gag1 and N4CT9-gag1 vectors generated robust Gag-specific immune responses following intramuscular immunization that were equivalent to or greater than immune responses generated by the more virulent prototypic vectors. MncpCT1-gag1 also induced Gag-specific immune responses following intramuscular immunization that were equivalent to immune responses generated by the prototypic rVSV vector. Placement of the gag gene in the first position of the VSV genome was associated with increased in vitro expression of Gag protein, in vivo expression of Gag mRNA, and enhanced immunogenicity of the vector. These findings demonstrate that through directed manipulation of the rVSV genome, vectors that have reduced neurovirulence and enhanced immunogenicity can be made.


Asunto(s)
Vacunas contra el SIDA/inmunología , Vectores Genéticos , VIH-1/genética , Vesiculovirus/genética , Vacunas Virales/inmunología , Vacunas contra el SIDA/genética , Animales , Citocinas/biosíntesis , Anticuerpos Anti-VIH/sangre , Inyecciones Intramusculares , Dosificación Letal Mediana , Ratones , Ratones Endogámicos BALB C , Mutación Puntual , Recombinación Genética , Eliminación de Secuencia , Linfocitos T Citotóxicos/inmunología , Translocación Genética , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Virales/genética , Virulencia , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología
9.
J Virol Methods ; 143(1): 55-64, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17382412

RESUMEN

Assessment of in vivo viral replication of live attenuated recombinant vesicular stomatitis virus (rVSV) vaccine vector candidates encoding HIV gag requires comprehensive preclinical safety studies, and development of sensitive assays to monitor the outcome of vaccination of animals is important. In this study, two 2-step quantitative real-time RT-PCR assays were developed; a singleplex assay to detect VSV genomic RNA from ferrets inoculated intra-cranially (IC) or intra-nasally (IN) with either a wild-type (wt) virus or an attenuated rVSV vector engineered to express HIV gag protein, and a duplex assay to simultaneously detect VSV-N and HIV-gag mRNAs from cynomolgus macaques inoculated intra-thalamically (IT) with the same viruses. Using synthetic oligonucleotides as standards, the lower limit of detection of VSV-N and HIV-gag was 50 copies. Results showed high levels of wt VSV(IN) genomic RNA and mRNA in ferret and macaque tissues, respectively, and significantly lower levels of VSV genomic RNA and VSV-N and HIV-gag mRNAs in tissues from animals inoculated with the attenuated rVSV vector. These assays correlated with both the course of infection for these animals, and the infectious viral load measured by a standard plaque assay, and could be used to determine the safety profile of rVSV vaccine vectors.


Asunto(s)
Vacunas contra el SIDA , Productos del Gen gag/aislamiento & purificación , VIH/genética , ARN Viral/aislamiento & purificación , Virus de la Estomatitis Vesicular Indiana/genética , Vacunas contra el SIDA/genética , Animales , Terapia Antirretroviral Altamente Activa , Hurones , Productos del Gen gag/genética , Vectores Genéticos , Macaca , ARN Viral/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Virus de la Estomatitis Vesicular Indiana/aislamiento & purificación , Carga Viral , Replicación Viral
10.
Virology ; 360(1): 36-49, 2007 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-17098273

RESUMEN

Although vesicular stomatitis virus (VSV) neurovirulence and pathogenicity in rodents have been well studied, little is known about VSV pathogenicity in non-human primates. To address this question, we measured VSV viremia, shedding, and neurovirulence in macaques. Following intranasal inoculation, macaques shed minimal recombinant VSV (rVSV) in nasal washes for 1 day post-inoculation; viremia was not detected. Following intranasal inoculation of macaques, wild type (wt) VSV, rVSV, and two rVSV-HIV vectors showed no evidence of spread to CNS tissues. However, macaques inoculated intrathalamically with wt VSV developed severe neurological disease. One of four macaques receiving rVSV developed clinical and histological signs similar to the wt group, while the remaining three macaques in this group and all of the macaques in the rVSV-HIV vector groups showed no clinical signs of disease and reduced severity of histopathology compared to the wt group. The implications of these findings for rVSV vaccine development are discussed.


Asunto(s)
Enfermedades del Sistema Nervioso Central/virología , Vectores Genéticos , Enfermedades de los Monos/virología , Infecciones por Rhabdoviridae/virología , Virus de la Estomatitis Vesicular Indiana , Vacunas contra el SIDA/administración & dosificación , Vacunas contra el SIDA/genética , Animales , Encéfalo/patología , Encéfalo/virología , Enfermedades del Sistema Nervioso Central/patología , Vectores Genéticos/administración & dosificación , Vectores Genéticos/fisiología , Inflamación/patología , Macaca mulatta , Masculino , Enfermedades de los Monos/patología , Mucosa Nasal/virología , Recombinación Genética , Infecciones por Rhabdoviridae/patología , Médula Espinal/patología , Virus de la Estomatitis Vesicular Indiana/patogenicidad , Virus de la Estomatitis Vesicular Indiana/fisiología , Viremia , Virulencia , Replicación Viral
11.
J Virol ; 81(4): 2056-64, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17151112

RESUMEN

A variety of rational approaches to attenuate growth and virulence of vesicular stomatitis virus (VSV) have been described previously. These include gene shuffling, truncation of the cytoplasmic tail of the G protein, and generation of noncytopathic M gene mutants. When separately introduced into recombinant VSV (rVSV), these mutations gave rise to viruses distinguished from their "wild-type" progenitor by diminished reproductive capacity in cell culture and/or reduced cytopathology and decreased pathogenicity in vivo. However, histopathology data from an exploratory nonhuman primate neurovirulence study indicated that some of these attenuated viruses could still cause significant levels of neurological injury. In this study, additional attenuated rVSV variants were generated by combination of the above-named three distinct classes of mutation. The resulting combination mutants were characterized by plaque size and growth kinetics in cell culture, and virulence was assessed by determination of the intracranial (IC) 50% lethal dose (LD(50)) in mice. Compared to virus having only one type of attenuating mutation, all of the mutation combinations examined gave rise to virus with smaller plaque phenotypes, delayed growth kinetics, and 10- to 500-fold-lower peak titers in cell culture. A similar pattern of attenuation was also observed following IC inoculation of mice, where differences in LD(50) of many orders of magnitude between viruses containing one and two types of attenuating mutation were sometimes seen. The results show synergistic rather than cumulative increases in attenuation and demonstrate a new approach to the attenuation of VSV and possibly other viruses.


Asunto(s)
Glicoproteínas de Membrana/genética , Infecciones por Rhabdoviridae/virología , Virus de la Estomatitis Vesicular Indiana/genética , Proteínas del Envoltorio Viral/genética , Animales , Línea Celular , Chlorocebus aethiops , Cricetinae , Femenino , Eliminación de Gen , Genes Virales/genética , Ratones , Proteínas de la Nucleocápside/genética , Células Vero , Virus de la Estomatitis Vesicular Indiana/crecimiento & desarrollo , Virus de la Estomatitis Vesicular Indiana/patogenicidad , Proteínas de la Matriz Viral/genética , Virulencia , Replicación Viral
12.
J Virol Methods ; 135(1): 91-101, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16569439

RESUMEN

Recovery of recombinant, negative-strand, nonsegmented RNA viruses from a genomic cDNA clone requires a rescue system that promotes de novo assembly of a functional ribonucleoprotein (RNP) complex in the cell cytoplasm. This is accomplished typically by cotransfecting permissive cells with multiple plasmids that encode the positive-sense genomic RNA, the nucleocapsid protein (N or NP), and the two subunits of the viral RNA-dependent RNA polymerase (L and P). The transfected plasmids are transcribed in the cell cytoplasm by phage T7 RNA polymerase (T7 RNAP), which usually is supplied by infection with a recombinant vaccinia virus or through use of a stable cell line that expresses the polymerase. Although both methods of providing T7 RNAP are effective neither is ideal for viral vaccine development for a number of reasons. Therefore, it was necessary to modify existing technology to make it possible to routinely rescue a variety of recombinant viruses when T7 RNAP was provided by a cotransfected expression plasmid. Development of a broadly applicable procedure required optimization of the helper-virus-free methodology, which resulted in several modifications that improved rescue efficiency such as inclusion of plasmids encoding viral glycoproteins and matrix protein, heat shock treatment, and use of electroporation. The combined effect of these enhancements produced several important benefits including: (1) a helper-virus-free methodology capable of rescuing a diverse variety of paramyxoviruses and recombinant vesicular stomatitis virus (rVSV); (2) methodology that functioned effectively when using Vero cells, a suitable substrate for vaccine production; and (3) a method that enabled rescue of highly attenuated recombinant viruses, which had proven refractory to rescue using published procedures.


Asunto(s)
Paramyxovirinae/aislamiento & purificación , Vacunas Atenuadas , Virus de la Estomatitis Vesicular Indiana/aislamiento & purificación , Vacunas Virales/genética , Animales , Chlorocebus aethiops , ADN Recombinante , ADN Viral , ARN Polimerasas Dirigidas por ADN/genética , Virus Helper/genética , Mutación , Paramyxovirinae/genética , Plásmidos/genética , ARN Viral/metabolismo , Transfección , Vacunas Atenuadas/genética , Vacunas Sintéticas/genética , Células Vero , Virus de la Estomatitis Vesicular Indiana/genética , Proteínas Virales/genética , Replicación Viral/genética , Replicación Viral/fisiología
13.
AIDS Res Hum Retroviruses ; 20(9): 989-1004, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15585086

RESUMEN

An experimental AIDS vaccine based on attenuated, recombinant vesicular stomatitis virus (rVSV), when administered by a combination of parenteral and mucosal routes, has proven effective at preventing AIDS in a rhesus macaque model (Rose NF, et al.: Cell 2001;106:539-549). In an effort to determine the optimal route of vaccine administration we evaluated the ability of rVSV-based vaccine vectors expressing HIV-1 Env and SIV Gag proteins, when given either intramuscularly (i.m.) or intranasally (i.n.), to elicit antigen-specific cellular and humoral immune responses, and to protect from a subsequent vaginal challenge with simian-human immunodeficiency virus (SHIV89.6P). Our results demonstrate that macaques vaccinated by the i.n. route developed significantly higher antigen-specific cellular immune responses as determined by MHC class I tetramer staining, IFN-gamma ELISPOT, and cytotoxic T cell assays. However, systemic and mucosal humoral immune responses did not vary significantly with the route of vaccine administration. Given the importance of cell-mediated immune responses in slowing AIDS progression, intranasal delivery of a VSV-based AIDS vaccine may be an optimal as well as practical route for vaccination and should be considered in design of clinical trials.


Asunto(s)
Vacunas contra el SIDA/inmunología , Productos del Gen env/inmunología , Productos del Gen gag/inmunología , Vectores Genéticos/inmunología , Vacunas Atenuadas/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Vacunas contra el SIDA/administración & dosificación , Administración Intranasal , Animales , Anticuerpos Antivirales/sangre , Femenino , Productos del Gen env/genética , Productos del Gen env/metabolismo , Productos del Gen gag/genética , Productos del Gen gag/metabolismo , Vectores Genéticos/administración & dosificación , Anticuerpos Anti-VIH/sangre , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , VIH-1/inmunología , Humanos , Inyecciones Intramusculares , Interferón gamma/biosíntesis , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/inmunología , Linfocitos T/inmunología , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Virus de la Estomatitis Vesicular Indiana/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA