Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
NPJ Vaccines ; 7(1): 85, 2022 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-35906244

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the COVID-19 global pandemic. SARS-CoV-2 is an enveloped RNA virus that relies on its trimeric surface glycoprotein spike for entry into host cells. Here we describe the COVID-19 vaccine candidate MV-014-212, a live, attenuated, recombinant human respiratory syncytial virus expressing a chimeric SARS-CoV-2 spike as the only viral envelope protein. MV-014-212 was attenuated and immunogenic in African green monkeys (AGMs). One mucosal administration of MV-014-212 in AGMs protected against SARS-CoV-2 challenge, reducing by more than 200-fold the peak shedding of SARS-CoV-2 in the nose. MV-014-212 elicited mucosal immunoglobulin A in the nose and neutralizing antibodies in serum that exhibited cross-neutralization against virus variants of concern Alpha, Beta, and Delta. Intranasally delivered, live attenuated vaccines such as MV-014-212 entail low-cost manufacturing suitable for global deployment. MV-014-212 is currently in Phase 1 clinical trials as an intranasal COVID-19 vaccine.

2.
PLoS One ; 10(3): e0119509, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25793508

RESUMEN

BACKGROUND: Illness associated with Respiratory Syncytial Virus (RSV) remains an unmet medical need in both full-term infants and older adults. The fusion glycoprotein (F) of RSV, which plays a key role in RSV infection and is a target of neutralizing antibodies, is an attractive vaccine target for inducing RSV-specific immunity. METHODOLOGY AND PRINCIPAL FINDINGS: BALB/c mice and cotton rats, two well-characterized rodent models of RSV infection, were used to evaluate the immunogenicity of intramuscularly administered RSV vaccine candidates consisting of purified soluble F (sF) protein formulated with TLR4 agonist glucopyranosyl lipid A (GLA), stable emulsion (SE), GLA-SE, or alum adjuvants. Protection from RSV challenge, serum RSV neutralizing responses, and anti-F IgG responses were induced by all of the tested adjuvanted RSV sF vaccine formulations. However, only RSV sF + GLA-SE induced robust F-specific TH1-biased humoral and cellular responses. In mice, these F-specific cellular responses include both CD4 and CD8 T cells, with F-specific polyfunctional CD8 T cells that traffic to the mouse lung following RSV challenge. This RSV sF + GLA-SE vaccine formulation can also induce robust RSV neutralizing titers and prime IFNγ-producing T cell responses in Sprague Dawley rats. CONCLUSIONS/SIGNIFICANCE: These studies indicate that a protein subunit vaccine consisting of RSV sF + GLA-SE can induce robust neutralizing antibody and T cell responses to RSV, enhancing viral clearance via a TH1 immune-mediated mechanism. This vaccine may benefit older populations at risk for RSV disease.


Asunto(s)
Adyuvantes Inmunológicos , Inmunidad Celular , Inmunidad Humoral , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Células TH1/inmunología , Vacunas de Subunidad/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células CHO , Movimiento Celular/inmunología , Cricetulus , Modelos Animales de Enfermedad , Femenino , Inmunización , Interferón gamma/metabolismo , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Ratones , Ratas , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/virología , Células TH1/metabolismo , Células Th2/inmunología
3.
J Clin Invest ; 125(4): 1637-47, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25751145

RESUMEN

Respiratory syncytial virus (RSV) is the most common cause of serious viral bronchiolitis in infants, young children, and the elderly. Currently, there is not an FDA-approved vaccine available for RSV, though the mAb palivizumab is licensed to reduce the incidence of RSV disease in premature or at-risk infants. The palivizumab epitope is a well-characterized, approximately 24-aa helix-loop-helix structure on the RSV fusion (F) protein (F254-277). Here, we genetically inserted this epitope and multiple site variants of this epitope within a versatile woodchuck hepadnavirus core-based virus-like particle (WHcAg-VLP) to generate hybrid VLPs that each bears 240 copies of the RSV epitope in a highly immunogenic arrayed format. A challenge of such an epitope-focused approach is that to be effective, the conformational F254-277 epitope must elicit antibodies that recognize the intact virus. A number of hybrid VLPs containing RSV F254-277 were recognized by palivizumab in vitro and elicited high-titer and protective neutralizing antibody in rodents. Together, the results from this proof-of-principle study suggest that the WHcAg-VLP technology may be an applicable approach to eliciting a response to other structural epitopes.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , Antígenos Virales/inmunología , Epítopos Inmunodominantes/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitiales Respiratorios/inmunología , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/inmunología , Especificidad de Anticuerpos , Técnicas Químicas Combinatorias , Microscopía por Crioelectrón , Ensayo de Inmunoadsorción Enzimática , Secuencias Hélice-Asa-Hélice/inmunología , Virus de la Hepatitis B de la Marmota/genética , Humanos , Inmunoglobulina G/biosíntesis , Inmunoglobulina G/inmunología , Ratones , Ratones Endogámicos BALB C , Palivizumab , Conformación Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/inmunología , Sigmodontinae , Vacunación , Vacunas de Partículas Similares a Virus , Proteínas Virales de Fusión/química
4.
PLoS One ; 8(10): e77104, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24204744

RESUMEN

UNLABELLED: Despite substantial morbidity associated with respiratory syncytial virus (RSV) infection, there is no licensed vaccine. MEDI-559 is a live attenuated intranasal vaccine candidate being developed for prevention of lower respiratory illness due to RSV in young children. This randomized, placebo-controlled study evaluated safety of MEDI-559 in healthy, RSV-seronegative children. MEDI-559 or placebo was administered on 3 occasions, 2 months apart. Primary safety was based on solicited symptoms (SSs) and adverse events (AEs) collected for 28 days after each dose. Nasal wash samples were collected 3 times after each dose (days 7-10, 12-18, 28-34) and at sick visits. Serum was collected for measuring antibody immune responses to RSV prior to first vaccination and 28 days post final dose. Long-term safety was monitored for 365 days from first dose. SSs were mild and frequent (MEDI-559 84%; placebo 91%); most common SSs were runny/stuffy nose, cough, and irritability/fussiness. AEs occurred in 67% MEDI-559 and 57% placebo recipients: most common AE was upper respiratory tract infection (MEDI-559 35%; placebo 23%). Higher incidence of medically attended lower respiratory illness within 28 days after dosing occurred in the MEDI-559 arm compared to placebo (none associated with vaccine virus shedding). There was no evidence of enhanced RSV disease. Vaccine virus was detected only in MEDI-559 recipients; shedding occurred in 56%subjects, primarily post dose 1. A functional immune response was observed in 59% and 9% MEDI-559 and placebo recipients, respectively, by an RSV microneutralization assay. Vaccine take, assessed by proportion that shed vaccine-type virus or had a seroresponse against RSV, was seen in 95% MEDI-559 subjects. MEDI-559 is therefore biologically active and immunogenic in this seronegative pediatric population. Although the frequency of SSs and AEs was not considered clinically significant, the increase in medically attended lower respiratory illnesses in the vaccine group warrants expanded safety studies. TRIAL REGISTRATION: ClinicalTrials.gov NCT00767416.


Asunto(s)
Anticuerpos Antivirales/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Anticuerpos Antivirales/sangre , Preescolar , Estudios de Cohortes , Tos/inducido químicamente , Femenino , Humanos , Lactante , Masculino , Obstrucción Nasal/inducido químicamente , Infecciones por Virus Sincitial Respiratorio/sangre , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Factores de Tiempo , Resultado del Tratamiento , Vacunación/métodos , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/inmunología
5.
J Gen Virol ; 94(Pt 12): 2627-2635, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24092758

RESUMEN

Respiratory syncytial virus (RSV) causes severe lower respiratory tract infection in children, especially in infants less than 1 year of age. There are currently no licensed vaccines against RSV. rA2ΔM2-2 is a promising live-attenuated vaccine candidate that is currently being evaluated in the clinic. Attenuation of rA2ΔM2-2 is achieved by a single deletion of the M2-2 gene, which disrupts the balance between viral transcription and replication. Whilst performing a manufacturing feasibility study in a serum-free adapted Vero cell line, differences in growth kinetics and cytopathic effect (CPE) were identified between two rA2ΔM2-2 vaccine candidates. Comparative sequence analysis identified four amino acid differences between the two vaccine viruses. Recombinant rA2ΔM2-2 viruses carrying each of the four amino acid differences identified a K66E mutation in the F2 fragment of the fusion (F) protein as the cause of the growth and CPE differences. Syncytium-formation experiments with RSV F protein carrying mutations at aa 66 suggested that a change in charge at this residue within the F2 fragment can have a significant impact on fusion.


Asunto(s)
Mutación , Virus Sincitial Respiratorio Humano/crecimiento & desarrollo , Virus Sincitial Respiratorio Humano/patogenicidad , Proteínas Virales de Fusión/genética , Animales , Chlorocebus aethiops , Efecto Citopatogénico Viral , Células Gigantes/fisiología , Humanos , Modelos Moleculares , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/genética , Células Vero , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/metabolismo
6.
Expert Rev Vaccines ; 12(9): 1047-60, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24053398

RESUMEN

Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections in infants and the elderly. Despite its relatively low degree of antigenic variation, it causes frequent reinfection throughout life. Clinical manifestations of RSV disease and the immune response to infection differ in infants and the elderly, suggesting that vaccines designed to protect these two populations may require different attributes. Here, the authors describe the translational approach of utilizing data from epidemiology studies performed in these populations, the use of RSV diagnostics in clinical practice, lessons learned from previous vaccine clinical trials and the success of palivizumab in prevention of RSV disease in premature and high-risk infants to aid the development of safe and effective RSV vaccines.


Asunto(s)
Anticuerpos Monoclonales Humanizados/administración & dosificación , Antivirales/administración & dosificación , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Infecciones por Virus Sincitial Respiratorio/epidemiología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Vacunas contra Virus Sincitial Respiratorio/aislamiento & purificación , Virus Sincitiales Respiratorios/inmunología , Descubrimiento de Drogas/tendencias , Humanos , Palivizumab , Infecciones por Virus Sincitial Respiratorio/prevención & control
7.
Vaccine ; 31(26): 2822-7, 2013 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-23602668

RESUMEN

MEDI-534 is the first live vectored RSV vaccine candidate to be evaluated in seronegative children. It consists of the bovine parainfluenza virus type 3 (PIV3) genome with substituted human PIV3 F and HN glycoproteins engineered to express RSV F protein. A Phase 1 study of 49 healthy RSV and PIV3 seronegative children 6 to <24 months of age demonstrated an acceptable safety profile at the following doses: 10(4), 10(5) and 10(6)TCID50. After 3 doses of MEDI-534 at 10(6)TCID50, administered at 0, 2 and 4 month intervals, 100% of subjects seroresponded to PIV3, whereas only 50% seroresponded to RSV. To investigate the discordance in seroresponse rates, the RSV F transgene and its flanking non-coding nucleotides were sequenced from shed virus recovered from the nasal washes of 24 MEDI-534-vaccinated children. Eleven out of 24 samples contained no nucleotide changes in the analyzed region. The other 13 samples contained mixtures of variant subpopulations. Fifty-five percent exhibited changes in the transcription termination poly A gene sequences of the upstream bPIV3N gene while 21% had variant subpopulations in the RSV F open reading frame that resulted in pre-mature stop codons. Both types of changes are expected to reduce RSV F expression. Evaluation of the administered vaccine by dual immunofluorescence staining showed ~2.5% variants with low or no RSV F expression while single nucleotide primer extension detected ~1% variation at nucleotide 2045 that resulted in a pre-mature translational termination at codon 85. An association between shedding of variants and lower RSV F serological response was observed but it was not possible to establish a definitive clinical significance due to the small number of subjects in this study.


Asunto(s)
Virus de la Parainfluenza 3 Humana/genética , Infecciones por Virus Sincitial Respiratorio/prevención & control , Virus Sincitiales Respiratorios/genética , Vacunas Virales/genética , Animales , Anticuerpos Antivirales/sangre , Bovinos , Estudios de Cohortes , Humanos , Lactante , Virus de la Parainfluenza 3 Bovina/genética , Virus de la Parainfluenza 3 Bovina/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Infecciones por Virus Sincitial Respiratorio/inmunología , Virus Sincitiales Respiratorios/inmunología , Análisis de Secuencia de ADN , Transgenes , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Virales/inmunología , Esparcimiento de Virus
8.
Vaccine ; 31(36): 3756-62, 2013 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-23623857

RESUMEN

MEDI-534 is the first live, attenuated and vectored respiratory syncytial virus (RSV) vaccine to be evaluated in seronegative children. It consists of a bovine/human parainfluenza virus type 3 (PIV3) backbone with the RSV fusion glycoprotein (RSV-F) expressed from the second position. The PIV3 fusion and hemaglutinin-neuraminidase proteins are human-derived. No small animal appropriately replicates the restrictive growth of bovine PIV3 (bPIV3) based viruses relative to human PIV3 (hPIV3) observed in the respiratory tract of rhesus monkeys and humans, making analysis of the genetic stability of the attenuation phenotype and maintenance of RSV-F expression difficult. Screening of multiple cell-lines identified MRC-5 cells as supporting permissive growth of hPIV3 while restricting bPIV3 and MEDI-534 growth. In MRC-5 cells, the peak titers of MEDI-534 were more than 20-fold lower compared to hPIV3 peak titers. After more than 10 multicycle passages in MRC-5 cells, genetic alterations were detected in MEDI-534 that contributed to a partial loss in restricted growth in MRC-5 cells and a decrease in RSV-F expression. These adaptive mutations did not occur in the RSV-F gene but were found in the polyA sequence upstream of the transgene. MRC-5 adapted MEDI-534 viruses (1) lost some attenuation but did not replicate to the level of hPIV3 in this cell line, (2) did not completely lose RSV-F expression and (3) were able to elicit a protective anti-RSV immune response in hamsters despite lower levels of RSV-F expression. Interestingly analysis of shed MEDI-534 from a recent clinical trial indicates that in some recipients similar mutations arise by day 7 or day 12 post immunization (in press) suggesting that these mutations can arise rapidly in the human host. The utility and limits of MRC-5 cells for characterizing the attenuation and RSV-F expression of MEDI-534 is discussed.


Asunto(s)
Virus Sincitiales Respiratorios/genética , Proteínas Virales de Fusión/inmunología , Vacunas Virales/inmunología , Cultivo de Virus , Animales , Línea Celular , Chlorocebus aethiops , Ensayos Clínicos Fase I como Asunto , Cricetinae , Regulación Viral de la Expresión Génica , Humanos , Mesocricetus , Mutación , Virus de la Parainfluenza 3 Bovina/genética , Virus de la Parainfluenza 3 Humana/genética , ARN Viral/genética , Virus Sincitiales Respiratorios/clasificación , Virus Sincitiales Respiratorios/inmunología , Análisis de Secuencia de ARN , Vacunas Atenuadas/inmunología , Células Vero , Replicación Viral
9.
Clin Vaccine Immunol ; 20(2): 239-47, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23239796

RESUMEN

Respiratory syncytial virus (RSV) infects elderly (≥65 years) adults, causing medically attended illness and hospitalizations. While RSV neutralizing antibody levels correlate inversely with RSV-associated hospitalization in the elderly, the role of RSV-specific T cells in preventing disease in the elderly remains unclear. We examined RSV-specific humoral, mucosal, and cellular immune profiles in healthy elderly (65 to 85 years) and young (20 to 30 years) adults. RSV neutralization antibody titers in the elderly (10.5 ± 2.2 log(2)) and young (10.5 ± 2.1 log(2)) were similar. In contrast, levels of RSV F protein-specific gamma interferon (IFN-γ)-producing T cells were lower in elderly (180 ± 80 spot-forming cells [SFC]/10(6) peripheral blood mononuclear cells [PBMC]) than in young adults (1,250 ± 420 SFC/10(6) PBMC). Higher levels of interleukin-13 (IL-13; 3,000 ± 1,000 pg/ml) in cultured PBMC supernatants and lower frequency of RSV F-specific CD107a(+) CD8(+) T cells (3.0% ± 1.6% versus 5.0% ± 1.6%) were measured in PBMC from elderly than young adults. These results suggest that deficient RSV F-specific T cell responses contribute to susceptibility to severe RSV disease in elderly adults.


Asunto(s)
Envejecimiento/inmunología , Anticuerpos Antivirales/sangre , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Memoria Inmunológica , Virus Sincitiales Respiratorios/inmunología , Proteínas Virales de Fusión/inmunología , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Neutralizantes/sangre , Humanos , Memoria Inmunológica/inmunología , Interferón gamma/biosíntesis , Interleucina-13/análisis , Leucocitos Mononucleares/inmunología , Recuento de Linfocitos , Proteína 1 de la Membrana Asociada a los Lisosomas/biosíntesis , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Infecciones por Virus Sincitial Respiratorio/virología , Adulto Joven
10.
Virus Res ; 169(1): 38-47, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22771939

RESUMEN

MEDI-559 is a recombinant live attenuated intranasal RSV vaccine candidate currently being evaluated in 5 to <24 month old RSV seronegative infants for safety and immunogenicity. MEDI-559 and the previously tested rA2cp248/404/1030ΔSH both have 5 cold-passaged mutations, 3 temperature sensitive (ts) markers designated 248, 404, and 1030, and deletion of the SH gene that collectively contribute to their attenuation and temperature sensitive growth phenotypes. However, MEDI-559 differs from rA2cp248/404/1030ΔSH by 39 silent nucleotide substitutions. Nevertheless, these viruses have comparable in vitro and in vivo phenotypes. Temperature sensitivity is monitored by the efficiency of plaque formation at elevated temperatures. The efficiency of plaque formation of MEDI-559 is reduced by ≥ 100-fold at 35 ° C and by ≥ 1000 fold at 37 °C compared to 32 °C. Passaging of MEDI-559 at temperatures up to 37 °C resulted in generation of temperature sensitive intermediate (tsi) viruses. The most frequent change was a reversion to wildtype tyrosine at the 1030 ts site followed by a less frequently observed leucine to non-wildtype serine substitution at the 248 ts site. One tsi virus had changes at both the 248 and 1030 ts sites and another tsi virus that had maintained all of the 248, 404 and 1030 ts sites had two novel changes (Asp158Gly and Ser1313Cys) in the polymerase (L) gene. Asp158Gly and Ser1313Cys singly or in combination in the MEDI-559 genetic background were confirmed to result in a tsi growth phenotype. All the tsi viruses have small plaque phenotypes and are highly attenuated in the lungs of cotton rats.


Asunto(s)
Vacunas contra Virus Sincitial Respiratorio/genética , Virus Sincitiales Respiratorios/genética , Virus Sincitiales Respiratorios/efectos de la radiación , Vacunas Atenuadas/genética , Replicación Viral/efectos de la radiación , Animales , Humanos , Mutación , ARN Viral/genética , Ratas , Virus Sincitiales Respiratorios/fisiología , Temperatura , Ensayo de Placa Viral , Proteínas Virales
11.
Virus Genes ; 40(2): 212-21, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20111897

RESUMEN

Studies of the fusion activity of respiratory syncytial virus (RSV) F protein are significantly hindered by low recombinant expression levels. While infection produces F protein levels detectable by western blot, recombinant expression produces undetectable to low levels of F protein. Identifying the obstacles that hinder recombinant F protein expression may lead to improved expression and facilitate the study of F protein function. We hypothesized that nuclear localization and/or inefficient RNA polymerase II-mediated transcription contribute to poor recombinant F protein expression. This study shows a combination of stalled nuclear export, premature polyadenylation, and low mRNA abundance all contribute to low recombinant F protein expression levels. In addition, this study provides an expression optimization strategy that results in greater F protein expression levels than observed by codon-optimization of the F protein gene, which will be useful for future studies of F protein function.


Asunto(s)
Transporte Activo de Núcleo Celular , Expresión Génica , Procesamiento Postranscripcional del ARN , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Proteínas Virales de Fusión/biosíntesis , Animales , Línea Celular , Humanos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteínas Virales de Fusión/genética
12.
Hum Vaccin ; 5(9): 582-91, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19556888

RESUMEN

RSV bronchiolitis is the leading cause of infant hospitalization in industrialized countries. There is an unmet need to prevent RSV lower respiratory tract infection in young infants. Although many vaccinology approaches, including live attenuated, viral and bacterial vectored and adjuvanted subunit vaccines have been evaluated in rodent and primate models there is currently no approved RSV vaccine. A vaccine candidate for RSV-naive infants must provide immunogenicity in the presence of maternally acquired antibodies, avoid enhanced disease and have minimal reactogenicity. Because live RSV infection does not potentiate for enhanced disease and elicits systemic and mucosal immune responses, live RSV vaccine candidates are currently preferred. Two live attenuated RSV vaccine candidates, rA2cpts248/404/1030/DeltaSH, a temperature sensitive RSV with a deletion of the SH gene, and rb/h PIV3/RSV F2 which has RSV F vectored into a bovine/human chimeric parainfluenza type 3 genome, have recently advanced into clinical studies.


Asunto(s)
Investigación Biomédica/tendencias , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Infecciones del Sistema Respiratorio/prevención & control , Preescolar , Ensayos Clínicos como Asunto , Humanos , Lactante , Recién Nacido , Infecciones por Virus Sincitial Respiratorio/epidemiología , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Infecciones del Sistema Respiratorio/epidemiología , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/inmunología
13.
Vaccine ; 26(50): 6373-82, 2008 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-18822334

RESUMEN

MEDI-534 is a bivalent live attenuated vaccine candidate against human respiratory syncytial virus (hRSV) and human parainfluenza virus type 3 (hPIV3) that was previously shown to be immunogenic and to protect rodents and African green monkeys from wild-type (wt) hRSV challenge. We performed further preclinical evaluations to address the safety of MEDI-534 prior to human testing. MEDI-534 did not predispose rodents to enhanced RSV disease following wt-RSV challenge, and the tissue tropism of the chimeric virus was confined to the respiratory tract. Representative clinical trial material did not produce toxicity in rats. In adults, MEDI-534 was highly restricted in replication, did not boost RSV and PIV3 antibody titers, and produced no medically significant vaccine-related adverse events thereby warranting further evaluation in pediatric populations.


Asunto(s)
Vacunas contra la Parainfluenza , Virus de la Parainfluenza 3 Humana/inmunología , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio , Infecciones por Respirovirus/prevención & control , Vacunas Atenuadas , Adolescente , Adulto , Animales , Chlorocebus aethiops , Cricetinae , Modelos Animales de Enfermedad , Método Doble Ciego , Femenino , Vectores Genéticos , Humanos , Masculino , Vacunas contra la Parainfluenza/administración & dosificación , Vacunas contra la Parainfluenza/genética , Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 3 Humana/genética , Ratas , Ratas Sprague-Dawley , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/virología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/efectos adversos , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/inmunología , Infecciones por Respirovirus/inmunología , Infecciones por Respirovirus/virología , Sigmodontinae , Resultado del Tratamiento , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/efectos adversos , Vacunas Atenuadas/inmunología , Células Vero , Adulto Joven
14.
Virol J ; 5: 69, 2008 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-18519001

RESUMEN

BACKGROUND: Human metapneumovirus (hMPV) infection can cause acute lower respiratory tract illness in infants, the immunocompromised, and the elderly. Currently there are no licensed preventative measures for hMPV infections. Using a variant of hMPV/NL/1/00 that does not require trypsin supplementation for growth in tissue culture, we deleted the M2-2 gene and evaluated the replication of rhMPV/DeltaM2-2 virus in vitro and in vivo. RESULTS: In vitro studies showed that the ablation of M2-2 increased the propensity for insertion of U nucleotides in poly-U tracts of the genomic RNA. In addition, viral transcription was up-regulated although the level of genomic RNA remained comparable to rhMPV. Thus, deletion of M2-2 alters the ratio between hMPV genome copies and transcripts. In vivo, rhMPV/DeltaM2-2 was attenuated compared to rhMPV in the lungs and nasal turbinates of hamsters. Hamsters immunized with one dose of rhMPV/DeltaM2-2 were protected from challenge with 106 PFU of wild type (wt) hMPV/NL/1/00. CONCLUSION: Our results suggest that hMPV M2-2 alters regulation of transcription and influences the fidelity of the polymerase complex during viral genome replication. In the hamster model, rhMPVDeltaM2-2 is attenuated and protective suggesting that deletion of M2-2 may result in a potential live vaccine candidate. A more thorough knowledge of the hMPV polymerase complex and the role of M2-2 during hMPV replication are being studied as we develop a potential live hMPV vaccine candidate that lacks M2-2 expression.


Asunto(s)
Metapneumovirus/crecimiento & desarrollo , Mutación , Infecciones por Paramyxoviridae/virología , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral , Animales , Secuencia de Bases , Línea Celular , Chlorocebus aethiops , Cricetinae , Regulación Viral de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Mesocricetus , Metapneumovirus/genética , Metapneumovirus/fisiología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Células Vero , Vacunas Virales/administración & dosificación
15.
J Virol ; 79(16): 10678-89, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16051860

RESUMEN

Human metapneumovirus (hMPV), a recently described paramyxovirus, is a major etiological agent for lower respiratory tract disease in young children that can manifest with severe cough, bronchiolitis, and pneumonia. The hMPV fusion glycoprotein (F) shares conserved functional domains with other paramyxovirus F proteins that are important for virus entry and spread. For other paramyxovirus F proteins, cleavage of a precursor protein (F0) into F1 and F2 exposes a fusion peptide at the N terminus of the F1 fragment, a likely prerequisite for fusion activity. Many hMPV strains have been reported to require trypsin for growth in tissue culture. The majority of these strains contain RQSR at the putative cleavage site. However, strains hMPV/NL/1/00 and hMPV/NL/1/99 expanded in our laboratory contain the sequence RQPR and do not require trypsin for growth in Vero cells. The contribution of this single amino acid change was verified directly by generating recombinant virus (rhMPV/NL/1/00) with either proline or serine at position 101 in F. These results suggested that cleavage of F protein in Vero cells could be achieved by trypsin or S101P amino acid substitution in the putative cleavage site motif. Moreover, trypsin-independent cleavage of hMPV F containing 101P was enhanced by the amino acid substitution E93K. In hamsters, rhMPV/93K/101S and rhMPV/93K/101P grew to equivalent titers in the respiratory tract and replication was restricted to respiratory tissues. The ability of these hMPV strains to replicate efficiently in the absence of trypsin should greatly facilitate the generation, preclinical testing, and manufacturing of attenuated hMPV vaccine candidates.


Asunto(s)
Metapneumovirus/crecimiento & desarrollo , Tripsina/farmacología , Proteínas Virales de Fusión/fisiología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Chlorocebus aethiops , Cricetinae , Mesocricetus , Datos de Secuencia Molecular , Tropismo , Células Vero , Proteínas Virales de Fusión/química , Replicación Viral
16.
Vaccine ; 23(14): 1657-67, 2005 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-15705469

RESUMEN

Human metapneumovirus (hMPV) infection causes respiratory tract disease similar to that observed during human respiratory syncytial virus infection (hRSV). hMPV infections have been reported across the entire age spectrum although the most severe disease occurs in young children. No vaccines, chemotherapeutics or antibodies are presently available for preventing or treating hMPV infections. In this study, a bovine/human chimeric parainfluenza virus type 3 (b/h PIV3) expressing the human parainfluenza type 3 (hPIV3) fusion (F) and hemagglutinin-neuraminidase (HN) proteins was engineered to express hMPV fusion (F) protein from the second genome position (b/h PIV3/hMPV F2) with the goal of generating a novel hMPV vaccine. b/h PIV3/hMPV F2 was previously shown to protect hamsters from challenge with wt hMPV (Tang RS, Schickli JH, Macphail M, Fernandes F, Bicha L, Spaete J, et al. Effects of human metapneumovirus and respiratory syncytial virus antigen insertion in two 3' proximal genome positions of bovine/human parainfluenza virus type 3 on virus replication and immunogenicity. J Virol 2003;77:10819-28) and is here further evaluated for efficacy and immunogenicity in African green monkeys (AGMs). AGMs immunized intranasally and intratracheally with b/h PIV3/hMPV F2 generated hMPV- and hPIV3-specific humoral and cellular immune responses and were protected from wt hMPV infection. In a separate study, the host-range restriction of b/h PIV3/hMPV F2 replication relative to wt hPIV3 was performed in rhesus monkeys to demonstrate attenuation. These studies showed that b/h PIV3/hMPV F2 was immunogenic, protective and attenuated in non-human primates and warrants further evaluation in humans as a vaccine candidate for prevention of hMPV-associated respiratory tract diseases.


Asunto(s)
Chlorocebus aethiops , Regulación Viral de la Expresión Génica/inmunología , Metapneumovirus/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Infecciones por Paramyxoviridae/prevención & control , Proteínas Virales de Fusión/biosíntesis , Proteínas Virales de Fusión/inmunología , Animales , Humanos , Macaca mulatta , Metapneumovirus/metabolismo , Virus de la Parainfluenza 3 Humana/metabolismo , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/virología , Células Vero , Proteínas Virales de Fusión/genética
17.
J Virol ; 78(20): 11198-207, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15452239

RESUMEN

Respiratory syncytial virus (RSV) causes respiratory disease in young children, the elderly, and immunocompromised individuals, often resulting in hospitalization and/or death. After more than 40 years of research, a Food and Drug Administration-approved vaccine for RSV is still not available. In this study, a chimeric bovine/human (b/h) parainfluenza virus type 3 (PIV3) expressing the human PIV3 (hPIV3) fusion (F) and hemagglutinin-neuraminidase (HN) proteins from an otherwise bovine PIV3 (bPIV3) genome was employed as a vector for RSV antigen expression with the aim of generating novel RSV vaccines. b/h PIV3 vaccine candidates expressing native or soluble RSV F proteins were evaluated for efficacy and immunogenicity in a nonhuman primate model. b/h PIV3 is suited for development of pediatric vaccines since bPIV3 had already been evaluated in clinical studies in 1- and 2-month-old infants and was found to be safe, immunogenic, and nontransmissible in a day care setting (Karron et al., Pediatr. Infect. Dis. J. 15:650-654, 1996; Lee et al., J. Infect. Dis. 184:909-913, 2001). African green monkeys immunized with b/h PIV3 expressing either the native or soluble RSV F protein were protected from challenge with wild-type RSV and produced RSV neutralizing and RSV F-protein specific immunoglobulin G serum antibodies. The PIV3-vectored RSV vaccines evaluated here further underscore the utility of this vector system for developing safe and immunogenic pediatric respiratory virus vaccines.


Asunto(s)
Anticuerpos Antivirales/sangre , Vectores Genéticos , Virus de la Parainfluenza 3 Humana/metabolismo , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio/inmunología , Proteínas Virales/inmunología , Animales , Chlorocebus aethiops , Humanos , Virus de la Parainfluenza 3 Humana/genética , Virus de la Parainfluenza 3 Humana/fisiología , Vacunas contra Virus Sincitial Respiratorio/administración & dosificación , Vacunas contra Virus Sincitial Respiratorio/genética , Virus Sincitial Respiratorio Humano/inmunología , Virus Sincitial Respiratorio Humano/patogenicidad , Sistema Respiratorio/virología , Solubilidad , Vacunación , Células Vero , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
18.
J Virol ; 78(15): 8264-70, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15254198

RESUMEN

Human metapneumovirus (hMPV) is a newly discovered pathogen associated with respiratory tract illness, primarily in young children, immunocompromised individuals, and the elderly. The genomic sequence of the prototype hMPV isolate NL/1/00 without the terminal leader and trailer sequences has been reported previously. Here we describe the leader and trailer sequences of two hMPV isolates, NL/1/00 and NL/1/99, representing the two main genetic lineages of hMPV. Minigenome constructs in which the green fluorescent protein or chloramphenicol acetyltransferase genes are flanked by the viral genomic ends derived from both hMPV lineages and transcribed using a T7 RNA polymerase promoter-terminator cassette were generated. Cotransfection of minigenome constructs with plasmids expressing the polymerase complex components L, P, N, and M2.1 in 293T or baby hamster kidney cells resulted in expression of the reporter genes. When the minigenome was replaced by a sense or antisense full-length cDNA copy of the NL/1/00 or NL/1/99 viral genomes, recombinant virus was recovered from transfected cells. Viral titers up to 10(7.2) and 10(5.7) 50% tissue culture infective dose/ml were achieved with the sense and antisense plasmids, respectively. The recombinant viruses replicated with kinetics similar to those of the parental viruses in Vero cells. This reverse genetics system provides an important new tool for applied and fundamental research.


Asunto(s)
Metapneumovirus/genética , Regiones no Traducidas 5' , Secuencia de Bases , ADN Complementario/genética , Genoma Viral , Humanos , Metapneumovirus/clasificación , Datos de Secuencia Molecular , Recombinación Genética , Serotipificación , Ensamble de Virus , Replicación Viral
19.
J Gen Virol ; 85(Pt 6): 1655-1663, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15166450

RESUMEN

Human metapneumovirus (hMPV), a recently identified paramyxovirus, is the causative agent of respiratory tract disease in young children. Epidemiological studies have established the presence of hMPV in retrospective as well as current clinical samples in Europe, USA, Canada, Hong Kong and Australia. The hMPV disease incidence rate varied from 7 to 12 %. This rate of disease attack places hMPV in severity between respiratory syncytial virus and human parainfluenza virus type 3, two common respiratory pathogens of young children, the elderly and immunosuppressed individuals. To evaluate the effectiveness and safety of future hMPV antiviral drugs, therapeutic and prophylactic monoclonal antibodies (mAbs), and vaccine candidates, it was necessary to identify small-animal and primate models that efficiently supported hMPV replication in the respiratory tract and produced neutralizing serum antibodies, commonly a clinical correlate of protection in humans. In this study, various rodents (mice, cotton rats, hamsters and ferrets) and two primate species, rhesus macaques and African green monkeys (AGMs), were evaluated for hMPV replication in the respiratory tract. The results showed that hamsters, ferrets and AGMs supported hMPV replication efficiently and produced high levels of hMPV-neutralizing antibody titres. Hamsters vaccinated with subgroup A hMPV were protected from challenge with subgroup A or subgroup B hMPV, which has implications for hMPV vaccine design. Although these animal models do not mimic human hMPV disease signs, they will nevertheless be invaluable for the future evaluation of hMPV antivirals, mAbs and vaccines.


Asunto(s)
Metapneumovirus/inmunología , Vacunas Virales/inmunología , Animales , Chlorocebus aethiops , Cricetinae , Hurones , Macaca mulatta , Mesocricetus , Metapneumovirus/fisiología , Ratones , Ratones Endogámicos BALB C , Modelos Animales , Sistema Respiratorio/virología , Células Vero , Replicación Viral
20.
J Gen Virol ; 84(Pt 12): 3253-3261, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14645907

RESUMEN

Restricted replication in the respiratory tract of rhesus monkeys is an intrinsic property of bovine parainfluenza virus type 3 (bPIV-3) strains. This host range phenotype of bPIV-3 has been utilized as a marker to evaluate the attenuation of bPIV-3 vaccines for human use. Two safety, immunogenicity and efficacy studies in primates evaluated and compared three human parainfluenza virus type 3 (hPIV-3) vaccine candidates: biologically derived bPIV-3, a plasmid-derived bPIV-3 (r-bPIV-3) and a chimeric bovine/human PIV-3 (b/hPIV-3). These studies also examined the feasibility of substituting Vero cells, cultured in the presence or absence of foetal bovine serum, for foetal rhesus lung-2 (FRhL-2) cells as the tissue culture substrate for the production of bPIV-3 vaccine. The results demonstrated that (i) Vero cell-produced bPIV-3 was as attenuated, immunogenic and efficacious as bPIV-3 vaccine grown in FRhL-2 cells, (ii) plasmid-derived bPIV-3 was as attenuated, immunogenic and efficacious as the biologically derived bPIV-3 and (iii) the b/hPIV-3 chimera displayed an intermediate attenuation phenotype and protected animals completely from hPIV-3 challenge. These results support the use of bPIV-3 vaccines propagated in Vero cells in human clinical trials and the use of b/hPIV-3 as a virus vaccine vector to express foreign viral antigens.


Asunto(s)
Vacunas contra la Parainfluenza/inmunología , Virus de la Parainfluenza 3 Bovina/inmunología , Virus de la Parainfluenza 3 Humana/inmunología , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/prevención & control , Animales , Anticuerpos Antivirales/sangre , Células Cultivadas , Chlorocebus aethiops , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Vectores Genéticos , Inmunización Secundaria , Inmunoglobulina A/sangre , Macaca mulatta , Pruebas de Neutralización , Vacunas contra la Parainfluenza/administración & dosificación , Virus de la Parainfluenza 3 Bovina/genética , Virus de la Parainfluenza 3 Humana/genética , Infecciones por Paramyxoviridae/sangre , Plásmidos , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...