Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 14(4): e0215321, 2019.
Article in English | MEDLINE | ID: mdl-30986224

ABSTRACT

Current approaches to influenza control rely on vaccines matched to viruses in circulation. Universal influenza vaccines would offer the advantage of providing broad protection against diverse strains of influenza virus. Candidate universal vaccines are developed using model systems, often testing in naïve animals. Yet the human population is not naïve, having varied immune histories that include exposure to viruses. We studied a candidate universal influenza vaccine (replication deficient adenoviruses expressing the conserved influenza A antigens NP and M2 [A/NP+M2-rAd]) given intranasally, the route previously shown to be most effective. To model recipients exposed to viruses, we used mice given rhinovirus (RV1B), respiratory syncytial virus (RSV-A2), influenza B virus, or influenza A virus before or after universal influenza vaccine. Vaccine performance was assessed by measuring immune responses to NP and M2, and monitoring weight loss and survival following influenza A challenge. Prior influenza A virus infection enhanced the response to the vaccine by priming to conserved influenza A antigens. RSV-A2 or RV1B had no effect on antibody responses to NP and M2 in serum. None of the viruses inhibited the ability of the vaccine to protect against influenza A virus challenge. The study demonstrates that the usefulness of this universal vaccine is not confined to the immunologically naïve and supports possible use in a human population with a varied history of respiratory infections.


Subject(s)
Common Variable Immunodeficiency/immunology , Coxsackievirus Infections/immunology , Enterovirus/immunology , Immunogenicity, Vaccine , Influenza A virus/immunology , Influenza B virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Animals , Common Variable Immunodeficiency/virology , Coxsackievirus Infections/pathology , Female , HeLa Cells , Humans , Influenza Vaccines/pharmacology , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/pathology
3.
J Virol ; 88(9): 5100-8, 2014 May.
Article in English | MEDLINE | ID: mdl-24574396

ABSTRACT

UNLABELLED: The lack of a vaccine against respiratory syncytial virus (RSV) is a challenging and serious gap in preventive medicine. Herein, we characterize the immunogenicity of an adenovirus serotype 5-based RSV vaccine encoding the fusion (F) protein (Ad5.RSV-F) and the protection provided following immunization with Ad5.RSV-F and assess its potential for producing enhanced disease in a cotton rat (CR) model. Animals were immunized intranasally (i.n.) and/or intramuscularly (i.m.) and subsequently challenged with RSV/A/Tracy (i.n.) to assess protection. Robust immune responses were seen in CRs vaccinated with Ad5.RSV-F given i.m. or i.n., and these responses correlated with reduced replication of the virus in noses and lungs after challenge. Neutralizing antibody responses following immunization with a single dose of Ad5.RSV-F at 1 × 10(11) viral particles (v.p.) elicited antibody titers 64- to 256-fold greater than those seen after natural infection. CRs boosted with Ad5.RSV-F i.n. 28 days after an i.m. dose also had significant increases in neutralizing antibody titers. Antibody affinity for different F-protein antigenic sites revealed substantial differences between antibodies elicited by Ad5.RSV-F and those seen after RSV infection; differences in antibody profiles were also seen between CRs given Ad5.RSV-F i.m. and CRs given Ad5.RSV-F i.n. Ad5.RSV-F priming did not result in enhanced disease following live-virus challenge, in contrast to the histopathology seen in CRs given the formalin-inactivated RSV/A/Burnett vaccine. IMPORTANCE: Respiratory syncytial virus (RSV) is the most common cause of acute lower respiratory infection in infants and young children and a serious health threat in the immunocompromised and the elderly. Infection severity increased in children in an immunization trial, hampering the over 4-decade-long quest for a successful RSV vaccine. In this study, we show that a genetically engineered RSV-F-encoding adenoviral vector provides protective immunity against RSV challenge without enhanced lung disease in cotton rats (CRs). CRs were vaccinated under a number of different regimens, and the immunity induced by the recombinant adenoviral RSV vaccine administered by use of an intramuscular prime-intranasal boost regimen may provide the best protection for young infants and children at risk of RSV infection, since this population is naive to adenoviral preformed immunity. Overall, this report describes a potential RSV vaccine candidate that merits further evaluation in a phase I clinical study in humans.


Subject(s)
Adenoviridae/genetics , Drug Carriers , Genetic Vectors , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/immunology , Administration, Intranasal , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Models, Animal , Injections, Intramuscular , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/genetics , Sigmodontinae , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology
4.
Vaccine ; 31(37): 3987-94, 2013 Aug 20.
Article in English | MEDLINE | ID: mdl-23742994

ABSTRACT

We have established a new reporter gene-based RSV neutralization test using Renilla luciferase. The RSV-Luciferase Neutralization Test (RSV-Luc-NeuT) is a simple, rapid, high throughput, and less labor intensive functional serological assay than the traditional RSV-PRNT, capable of measuring a broad range of anti-RSV neutralizing antibodies targeting both RSV-F and RSV-G proteins. Specificity and sensitivity of the RSV-Luc-NeuT are comparable to the RSV-PRNT. Panels of pre-vaccination and post-vaccination animal sera, monoclonal antibodies and animal polyclonal anti-RSV sera confirmed assay specificity. A panel of 60 human sera demonstrated high assay sensitivity for measurement of RSV neutralizing antibodies that strongly correlated with the RSV-PRNT titers (R(2)=0.864). Neutralization in the presence of guinea pig complement (GPC) increased PRNT titers more than the RSV-Luc-NeuT neutralizing antibody titers for these human sera. This newly developed simple, high throughput, RSV-Luc-NeuT could be easily automated and applied in measurement of RSV neutralization titers in large vaccine trials.


Subject(s)
Luciferases/genetics , Neutralization Tests/methods , Respiratory Syncytial Viruses/immunology , Animals , Antibodies, Neutralizing/analysis , Cell Line/virology , Complement System Proteins/immunology , Epitopes/immunology , Genes, Reporter , High-Throughput Screening Assays , Humans , Immune Sera , Mice , Rabbits , Sensitivity and Specificity , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Fusion Proteins/immunology
5.
J Virol ; 81(1): 261-71, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17050595

ABSTRACT

It has been shown previously that the fusion glycoprotein of human respiratory syncytial virus (RSV-F) interacts with cellular heparan sulfate. Synthetic overlapping peptides derived from the F-protein sequence of RSV subtype A (strain A2) were tested for their ability to bind heparin using heparin-agarose affinity chromatography (HAAC). This evaluation identified 15 peptides representing eight linear heparin-binding domains (HBDs) located within F1 and F2 and spanning the protease cleavage activation site. All peptides bound to Vero and A549 cells, and binding was inhibited by soluble heparins and diminished by either enzymatic treatment to remove cell surface glycosaminoglycans or by treatment with sodium chlorate to decrease cellular sulfation. RSV-F HBD peptides were less likely to bind to glycosaminoglycan-deficient CHO-745 cells than parental CHO-K1 cells that express these molecules. Three RSV-F HBD peptides (F16, F26, and F55) inhibited virus infectivity; two of these peptides (F16 and F55) inhibited binding of virus to Vero cells, while the third (F26) did not. These studies provided evidence that two of the linear HBDs mapped by peptides F16 and F55 may mediate one of the first steps in the attachment of virus to cells while the third, F26, inhibited infectivity at a postattachment step, suggesting that interactions with cell surface glycosaminoglycans may play a role in infectivity of some RSV strains.


Subject(s)
Heparin/metabolism , Peptides/pharmacology , Respiratory Syncytial Virus, Human/pathogenicity , Viral Fusion Proteins/chemistry , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Chlorates/pharmacology , Chlorocebus aethiops , Cricetinae , Cricetulus , Gene Products, gag/genetics , Gene Products, gag/metabolism , Glycoproteins/chemistry , Glycoproteins/metabolism , Humans , Lyases/pharmacology , Molecular Sequence Data , Peptide Mapping , Peptides/chemistry , Peptides/metabolism , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/metabolism , Vero Cells , Viral Fusion Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...