Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Virol ; 97(1): e0172322, 2023 01 31.
Article in English | MEDLINE | ID: mdl-36533954

ABSTRACT

Most human influenza vaccine antigens are produced in fertilized chicken eggs. Recent H3N2 egg-based vaccine antigens have limited effectiveness, partially due to egg-adaptive substitutions that alter the antigenicity of the hemagglutinin (HA) protein. The nucleoside-modified mRNA encapsulated in lipid nanoparticles (mRNA-LNP) vaccine platform is a promising alternative for egg-based influenza vaccines because mRNA-LNP-derived antigens are not subject to adaptive pressures that arise during the production of antigens in chicken eggs. Here, we compared H3N2-specific antibody responses in mice vaccinated with either 3c.2A H3-encoding mRNA-LNP or a conventional egg-based Fluzone vaccine (which included an egg-adapted 3c.2A antigen) supplemented with an MF59-like adjuvant. We tested mRNA-LNP encoding wild-type and egg-adapted H3 antigens. We found that mRNA-LNP encoding wild-type H3 elicited antibodies that neutralized the wild-type 3c.2A H3N2 virus more effectively than antibodies elicited by mRNA-LNP encoding egg-adapted H3 or the egg-based Fluzone vaccine. mRNA-LNP expressing either wild-type or egg-adapted H3 protected mice against infection with the wild-type 3c2.A H3N2, whereas the egg-based Fluzone vaccine did not. We found that both mRNA-LNP vaccines elicited high levels of group 2 HA stalk-reactive antibodies, which likely contributed to protection in vivo. Our studies indicate that nucleoside-modified mRNA-LNP-based vaccines can circumvent problems associated with egg adaptations with recent 3c2.A H3N2 viruses. IMPORTANCE This study shows that the nucleoside-modified mRNA-LNP vaccine platform is a promising alternative for egg-based influenza vaccines. We show that mRNA-LNP vaccines expressing H3 antigens elicit high levels of antibodies in mice and protect against H3N2 influenza virus infection.


Subject(s)
Influenza A Virus, H3N2 Subtype , Influenza Vaccines , Nucleosides , mRNA Vaccines , Animals , Humans , Mice , Antibodies, Viral , Chickens , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza Vaccines/genetics , Influenza Vaccines/immunology , Influenza, Human/prevention & control , RNA, Messenger/genetics , mRNA Vaccines/genetics , mRNA Vaccines/immunology
2.
Science ; 378(6622): 899-904, 2022 11 25.
Article in English | MEDLINE | ID: mdl-36423275

ABSTRACT

Seasonal influenza vaccines offer little protection against pandemic influenza virus strains. It is difficult to create effective prepandemic vaccines because it is uncertain which influenza virus subtype will cause the next pandemic. In this work, we developed a nucleoside-modified messenger RNA (mRNA)-lipid nanoparticle vaccine encoding hemagglutinin antigens from all 20 known influenza A virus subtypes and influenza B virus lineages. This multivalent vaccine elicited high levels of cross-reactive and subtype-specific antibodies in mice and ferrets that reacted to all 20 encoded antigens. Vaccination protected mice and ferrets challenged with matched and mismatched viral strains, and this protection was at least partially dependent on antibodies. Our studies indicate that mRNA vaccines can provide protection against antigenically variable viruses by simultaneously inducing antibodies against multiple antigens.


Subject(s)
Influenza A virus , Influenza B virus , Orthomyxoviridae Infections , Vaccines, Combined , Vaccines, Synthetic , mRNA Vaccines , Animals , Mice , Ferrets , Nucleosides/chemistry , Nucleosides/genetics , Orthomyxoviridae Infections/prevention & control , Vaccines, Combined/genetics , Vaccines, Combined/immunology , mRNA Vaccines/genetics , mRNA Vaccines/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Influenza A virus/immunology , Influenza B virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Cross Reactions
3.
Sci Transl Med ; 12(525)2020 01 08.
Article in English | MEDLINE | ID: mdl-31915303

ABSTRACT

Maternal antibodies provide short-term protection to infants against many infections. However, they can inhibit de novo antibody responses in infants elicited by infections or vaccination, leading to increased long-term susceptibility to infectious diseases. Thus, there is a need to develop vaccines that are able to elicit protective immune responses in the presence of antigen-specific maternal antibodies. Here, we used a mouse model to demonstrate that influenza virus-specific maternal antibodies inhibited de novo antibody responses in mouse pups elicited by influenza virus infection or administration of conventional influenza vaccines. We found that a recently developed influenza vaccine, nucleoside-modified mRNA encapsulated in lipid nanoparticles (mRNA-LNP), partially overcame this inhibition by maternal antibodies. The mRNA-LNP influenza vaccine established long-lived germinal centers in the mouse pups and elicited stronger antibody responses than did a conventional influenza vaccine approved for use in humans. Vaccination with mRNA-LNP vaccines may offer a promising strategy for generating robust immune responses in infants in the presence of maternal antibodies.


Subject(s)
Antibody Formation/immunology , Nucleosides/metabolism , RNA, Messenger/immunology , Vaccination , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Viral/immunology , Disease Models, Animal , Female , Germinal Center , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza Vaccines/immunology , Lipids/chemistry , Mice, Inbred BALB C , Mice, Inbred C57BL , Nanoparticles/chemistry , Orthomyxoviridae/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/virology
4.
Clin Infect Dis ; 71(6): 1447-1453, 2020 09 12.
Article in English | MEDLINE | ID: mdl-31598646

ABSTRACT

BACKGROUND: The H3N2 component of egg-based 2017-2018 influenza vaccines possessed an adaptive substitution that alters antigenicity. Several influenza vaccines include antigens that are produced through alternative systems, but a systematic comparison of different vaccines used during the 2017-2018 season has not been completed. METHODS: We compared antibody responses in humans vaccinated with Fluzone (egg-based, n = 23), Fluzone High-Dose (egg-based, n = 16), Flublok (recombinant protein-based, n = 23), or Flucelvax (cell-based, n = 23) during the 2017-2018 season. We completed neutralization assays using an egg-adapted H3N2 virus, a cell-based H3N2 virus, wild-type 3c2.A and 3c2.A2 H3N2 viruses, and the H1N1 vaccine strain. We also performed enzyme-linked immunosorbent assays using a recombinant wild-type 3c2.A hemagglutinin. Antibody responses were compared in adjusted analysis. RESULTS: Postvaccination neutralizing antibody titers to 3c2.A and 3c2.A2 were higher in Flublok recipients compared with Flucelvax or Fluzone recipients (P < .01). Postvaccination titers to 3c2.A and 3c2.A2 were similar in Flublok and Fluzone High-Dose recipients, though seroconversion rates trended higher in Flublok recipients. Postvaccination titers in Flucelvax recipients were low to all H3N2 viruses tested, including the cell-based H3N2 strain. Postvaccination neutralizing antibody titers to H1N1 were similar among the different vaccine groups. CONCLUSIONS: These data suggest that influenza vaccine antigen match and dose are both important for eliciting optimal H3N2 antibody responses in humans. Future studies should be designed to determine if our findings directly impact vaccine effectiveness. CLINICAL TRIALS REGISTRATION: NCT03068949.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Influenza, Human , Antibodies, Viral , Antibody Formation , Antigens, Viral , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus , Humans , Influenza A Virus, H3N2 Subtype , Influenza, Human/prevention & control , Recombinant Proteins , Seasons
5.
Cell Rep ; 29(13): 4460-4470.e8, 2019 12 24.
Article in English | MEDLINE | ID: mdl-31875553

ABSTRACT

Antibodies targeting the receptor binding site (RBS) of the influenza virus hemagglutinin (HA) protein are usually not broadly reactive because their footprints are typically large and extend to nearby variable HA residues. Here, we identify several human H3N2 HA RBS-targeting monoclonal antibodies (mAbs) that are sensitive to substitutions in conventional antigenic sites and are therefore not broadly reactive. However, we also identify an H3N2 HA RBS-targeting mAb that is exceptionally broadly reactive despite being sensitive to substitutions in residues outside of the RBS. We show that similar antibodies are present at measurable levels in the sera of some individuals but that they are inefficiently elicited by conventional vaccines. Our data indicate that HA RBS-targeting antibodies can be effective against variable viral strains even when they are somewhat sensitive to substitutions in HA residues adjacent to the RBS.


Subject(s)
Antibodies, Viral/chemistry , Epitopes/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/immunology , Vaccination , Amino Acid Substitution , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/isolation & purification , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/isolation & purification , Antibodies, Neutralizing/metabolism , Antibodies, Viral/isolation & purification , Antibodies, Viral/metabolism , Antibody Specificity , Binding Sites , Epitopes/immunology , Epitopes/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Humans , Immune Sera/chemistry , Influenza A Virus, H3N2 Subtype/genetics , Influenza, Human/prevention & control , Influenza, Human/virology , Models, Molecular , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs
6.
Nat Commun ; 9(1): 3361, 2018 08 22.
Article in English | MEDLINE | ID: mdl-30135514

ABSTRACT

Currently available influenza virus vaccines have inadequate effectiveness and are reformulated annually due to viral antigenic drift. Thus, development of a vaccine that confers long-term protective immunity against antigenically distant influenza virus strains is urgently needed. The highly conserved influenza virus hemagglutinin (HA) stalk represents one of the potential targets of broadly protective/universal influenza virus vaccines. Here, we evaluate a potent broadly protective influenza virus vaccine candidate that uses nucleoside-modified and purified mRNA encoding full-length influenza virus HA formulated in lipid nanoparticles (LNPs). We demonstrate that immunization with HA mRNA-LNPs induces antibody responses against the HA stalk domain of influenza virus in mice, rabbits, and ferrets. The HA stalk-specific antibody response is associated with protection from homologous, heterologous, and heterosubtypic influenza virus infection in mice.


Subject(s)
Antibodies, Viral/immunology , Hemagglutinins/immunology , Influenza A Virus, H5N1 Subtype/immunology , Orthomyxoviridae/immunology , RNA, Messenger/chemistry , RNA, Messenger/immunology , Animals , Cells, Cultured , Dogs , Enzyme-Linked Immunosorbent Assay , Female , Ferrets , Flow Cytometry , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Phylogeny , Rabbits
7.
Vaccine ; 36(25): 3650-3665, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29764680

ABSTRACT

Vaccination remains the most cost-effective biomedical approach for controlling influenza disease. In times of pandemics, however, these vaccines cannot be produced in sufficient quantities for worldwide use by the current manufacturing capacities and practices. What is needed is the development of adjuvanted vaccines capable of inducing an adequate or better immune response at a decreased antigen dose. Previously we showed that the protein adjuvant rOv-ASP-1 augments influenza-specific antibody titers and survival after virus challenge in both young adult and old-age mice when administered with the trivalent inactivated influenza vaccine (IIV3). In this study we show that a reduced amount of rOv-ASP-1, with 40-times less IIV3 can also induce protection. Apparently the potency of the rOv-ASP-1 adjuvanted IIV3 vaccine is independent of the IIV3-specific Th1/Th2 associated antibody responses, and independent of the presence of HAI antibodies. However, CD4+ T helper cells were indispensable for the protection. Further, rOv-ASP-1 with or without IIV3 elicited the increased level of various chemokines, which are known chemoattractant for immune cells, into the muscle 4 h after immunization, and significantly induced the recruitment of monocytes, macrophages and neutrophils into the muscles. The recruited monocytes had higher expression of the activation marker MHCII on their surface as well as CXCR3 and CCR2; receptors for IP-10 and MCP-1, respectively. These results show that the rOv-ASP-1 adjuvant allows substantial antigen sparing of IIV3 by stimulating at the site of injection the accumulation of chemokines and the recruitment of immune cells that can augment the activation of CD4+ T cell immune responses, essential for the production of antibody responses. Protection elicited by the rOv-ASP-1 adjuvanted IIV3 vaccine also appears to function in the absence of MyD88-signaling. Future studies will attempt to delineate the precise mechanisms by which the rOv-ASP-1 adjuvanted IIV3 vaccine works.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Aging/immunology , Antibodies, Viral/biosynthesis , Antigens, Helminth/administration & dosage , Helminth Proteins/administration & dosage , Immunization/methods , Influenza Vaccines/administration & dosage , Orthomyxoviridae Infections/prevention & control , Aging/genetics , Animals , Female , Gene Expression Regulation , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/virology , Mice , Mice, Knockout , Monocytes/drug effects , Monocytes/immunology , Monocytes/virology , Muscle, Skeletal/drug effects , Muscle, Skeletal/immunology , Muscle, Skeletal/virology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/virology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/virology , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Survival Analysis , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology , Viral Load/drug effects , Viral Load/immunology
8.
J Exp Med ; 215(6): 1571-1588, 2018 06 04.
Article in English | MEDLINE | ID: mdl-29739835

ABSTRACT

T follicular helper (Tfh) cells are required to develop germinal center (GC) responses and drive immunoglobulin class switch, affinity maturation, and long-term B cell memory. In this study, we characterize a recently developed vaccine platform, nucleoside-modified, purified mRNA encapsulated in lipid nanoparticles (mRNA-LNPs), that induces high levels of Tfh and GC B cells. Intradermal vaccination with nucleoside-modified mRNA-LNPs encoding various viral surface antigens elicited polyfunctional, antigen-specific, CD4+ T cell responses and potent neutralizing antibody responses in mice and nonhuman primates. Importantly, the strong antigen-specific Tfh cell response and high numbers of GC B cells and plasma cells were associated with long-lived and high-affinity neutralizing antibodies and durable protection. Comparative studies demonstrated that nucleoside-modified mRNA-LNP vaccines outperformed adjuvanted protein and inactivated virus vaccines and pathogen infection. The incorporation of noninflammatory, modified nucleosides in the mRNA is required for the production of large amounts of antigen and for robust immune responses.


Subject(s)
B-Lymphocytes/immunology , Germinal Center/cytology , Nucleosides/metabolism , RNA, Messenger/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Vaccines, Subunit/immunology , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Neutralizing/immunology , Antibody Formation/immunology , Antigens/metabolism , Lipids/chemistry , Macaca mulatta , Nanoparticles/chemistry , Protein Subunits/metabolism , Time Factors , Vaccination
9.
Clin Infect Dis ; 67(3): 327-333, 2018 07 18.
Article in English | MEDLINE | ID: mdl-29471464

ABSTRACT

Background: Influenza vaccination aims to prevent infection by influenza virus and reduce associated morbidity and mortality; however, vaccine effectiveness (VE) can be modest, especially for subtype A(H3N2). Low VE has been attributed to mismatches between the vaccine and circulating influenza strains and to the vaccine's elicitation of protective immunity in only a subset of the population. The low H3N2 VE in the 2012-2013 season was attributed to egg-adaptive mutations that created antigenic mismatch between the actual vaccine strain (IVR-165) and both the intended vaccine strain (A/Victoria/361/2011) and the predominant circulating strains (clades 3C.2 and 3C.3). Methods: We investigated the basis of low VE in 2012-2013 by determining whether vaccinated and unvaccinated individuals were infected by different viral strains and by assessing the serologic responses to IVR-165, A/Victoria/361/2011, and 3C.2 and 3C.3 strains in an adult cohort before and after vaccination. Results: We found no significant genetic differences between the strains that infected vaccinated and unvaccinated individuals. Vaccination increased titers to A/Victoria/361/2011 and 3C.2 and 3C.3 representative strains as much as to IVR-165. These results are consistent with the hypothesis that vaccination boosted cross-reactive immune responses instead of specific responses against unique vaccine epitopes. Only approximately one-third of the cohort achieved a ≥4-fold increase in titer. Conclusions: In contrast to analyses based on ferret studies, low H3N2 VE in 2012-2013 in adults does not appear to be due to egg adaptation of the vaccine strain. Instead, low VE might have been caused by low vaccine immunogenicity in a subset of the population.


Subject(s)
Immunogenicity, Vaccine , Influenza A Virus, H3N2 Subtype/genetics , Influenza Vaccines/immunology , Influenza, Human/epidemiology , Adaptation, Physiological , Adult , Aged , Aged, 80 and over , Animals , Antigens, Viral/immunology , Cohort Studies , Cross Reactions , Eggs/virology , Ferrets , Genome, Viral , High-Throughput Nucleotide Sequencing , Humans , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza Vaccines/therapeutic use , Influenza, Human/prevention & control , Mutation , Phylogeny , Seasons
10.
Cell Rep ; 22(2): 411-426, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29320737

ABSTRACT

Acute respiratory tract viral infections (ARTIs) cause significant morbidity and mortality. CD8 T cells are fundamental to host responses, but transcriptional alterations underlying anti-viral mechanisms and links to clinical characteristics remain unclear. CD8 T cell transcriptional circuitry in acutely ill pediatric patients with influenza-like illness was distinct for different viral pathogens. Although changes included expected upregulation of interferon-stimulated genes (ISGs), transcriptional downregulation was prominent upon exposure to innate immune signals in early IFV infection. Network analysis linked changes to severity of infection, asthma, sex, and age. An influenza pediatric signature (IPS) distinguished acute influenza from other ARTIs and outperformed other influenza prediction gene lists. The IPS allowed a deeper investigation of the connection between transcriptional alterations and clinical characteristics of acute illness, including age-based differences in circuits connecting the STAT1/2 pathway to ISGs. A CD8 T cell-focused systems immunology approach in pediatrics identified age-based alterations in ARTI host response pathways.


Subject(s)
Genomics/methods , Respiratory Tract Infections/immunology , Female , Humans , Male , Respiratory Tract Infections/pathology
11.
Proc Natl Acad Sci U S A ; 114(47): 12578-12583, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29109276

ABSTRACT

H3N2 viruses continuously acquire mutations in the hemagglutinin (HA) glycoprotein that abrogate binding of human antibodies. During the 2014-2015 influenza season, clade 3C.2a H3N2 viruses possessing a new predicted glycosylation site in antigenic site B of HA emerged, and these viruses remain prevalent today. The 2016-2017 seasonal influenza vaccine was updated to include a clade 3C.2a H3N2 strain; however, the egg-adapted version of this viral strain lacks the new putative glycosylation site. Here, we biochemically demonstrate that the HA antigenic site B of circulating clade 3C.2a viruses is glycosylated. We show that antibodies elicited in ferrets and humans exposed to the egg-adapted 2016-2017 H3N2 vaccine strain poorly neutralize a glycosylated clade 3C.2a H3N2 virus. Importantly, antibodies elicited in ferrets infected with the current circulating H3N2 viral strain (that possesses the glycosylation site) and humans vaccinated with baculovirus-expressed H3 antigens (that possess the glycosylation site motif) were able to efficiently recognize a glycosylated clade 3C.2a H3N2 virus. We propose that differences in glycosylation between H3N2 egg-adapted vaccines and circulating strains likely contributed to reduced vaccine effectiveness during the 2016-2017 influenza season. Furthermore, our data suggest that influenza virus antigens prepared via systems not reliant on egg adaptations are more likely to elicit protective antibody responses that are not affected by glycosylation of antigenic site B of H3N2 HA.


Subject(s)
Antibodies, Viral/metabolism , Antigens, Viral/genetics , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Influenza A Virus, H3N2 Subtype/genetics , Influenza Vaccines/chemistry , Animals , Antigens, Viral/chemistry , Antigens, Viral/immunology , Chickens , Ferrets , Glycosylation , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Immunogenicity, Vaccine , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Mutation , Neutralization Tests , Ovum/virology
12.
Sci Immunol ; 2(8)2017 Feb.
Article in English | MEDLINE | ID: mdl-28620653

ABSTRACT

T follicular helper (Tfh) CD4 cells are crucial providers of B cell help during adaptive immune responses. A circulating population of CD4 T cells, termed cTfh, have similarity to lymphoid Tfh, can provide B cell help, and responded to influenza vaccination. However, it is unclear whether human vaccination-induced cTfh respond in an antigen-specific manner and whether they form long-lasting memory. Here, we identified a cTfh population that expressed multiple T cell activation markers and could be readily identified by coexpression of ICOS and CD38. This subset expressed more Bcl-6, c-Maf, and IL-21 than other blood CD4 subsets. Influenza vaccination induced a strong response in the ICOS+CD38+ cTfh at day 7, and this population included hemagglutinin-specific cells by tetramer staining and antigen-stimulated Activation Induced Marker (AIM) expression. Moreover, TCRB sequencing identified a clonal response in ICOS+CD38+ cTfh that correlated strongly with the increased circulating ICOS+CD38+ cTfh frequency and the circulating plasmablast response. In subjects who received successive annual vaccinations, a recurrent oligoclonal response was identified in the ICOS+CD38+ cTfh subset at 7 days after every vaccination. These oligoclonal responses in ICOS+CD38+ cTfh after vaccination persisted in the ICOS-CD38- cTfh repertoire in subsequent years, suggesting clonal maintenance in a memory reservoir in the more-stable ICOS-CD38- cTfh subset. These data highlight the antigen-specificity, lineage relationships and memory properties of human cTfh responses to vaccination, providing new avenues for tracking and monitoring cTfh responses during infection and vaccination in humans.

13.
J Infect Dis ; 214(12): 1947-1951, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27923954

ABSTRACT

During the 2013-2014 influenza season, nearly all circulating 2009 pandemic influenza A(H1N1) virus (A[H1N1]pdm09) strains possessed an antigenically important mutation in hemagglutinin (K166Q). Here, we performed hemagglutination-inhibition (HAI) assays, using sera collected from 382 individuals prior to the 2013-2014 season, and we determined whether HAI titers were associated with protection from A(H1N1)pdm09 infection. Protection was associated with HAI titers against an A(H1N1)pdm09 strain possessing the K166Q mutation but not with HAI titers against the current A(H1N1)pdm09 vaccine strain, which lacks this mutation. These data indicate that contemporary A(H1N1)pdm09 strains are antigenically distinct from the current A(H1N1)pdm09 vaccine strain.


Subject(s)
Antibodies, Viral/blood , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Adolescent , Adult , Aged , Cross Protection , Female , Hemagglutination Inhibition Tests , Humans , Influenza Vaccines/administration & dosage , Influenza, Human/immunology , Male , Middle Aged , Young Adult
14.
Vaccine ; 34(46): 5483-5487, 2016 11 04.
Article in English | MEDLINE | ID: mdl-27670073

ABSTRACT

Influenza A viruses infect many species and cross-species transmission occurs occasionally. An equine H3N8 influenza virus began circulating in dogs in 1999 and an avian H3N2 influenza virus began circulating in dogs in 2006. The canine H3N8 (cH3N8) viral strain has become endemic in parts of the United States and there is a commercially available vaccine against this strain. The canine H3N2 (cH3N2) strain did not circulate widely in the United States until 2015. Here, we used a mouse model to determine if the cH3N8 and cH3N2 strains are antigenically related and if a commercially available cH3N8 vaccine protects animals against the cH3N2 outbreak strain. We find that the cH3N8 vaccine elicits antibodies that react to internal viral proteins and the hemagglutinin stalk region of cH3N2 viruses. These antibodies do not provide sterilizing immunity against cH3N2 infection, but these antibodies limit cH3N2 replication in the lung.


Subject(s)
Dog Diseases/prevention & control , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N8 Subtype/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/veterinary , Animals , Antibodies, Viral/blood , Cross Protection , Disease Models, Animal , Dog Diseases/epidemiology , Dog Diseases/virology , Dogs , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N8 Subtype/genetics , Lung/virology , Mice , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , United States/epidemiology
15.
Cell Rep ; 12(1): 1-6, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26119736

ABSTRACT

Influenza vaccines must be updated regularly because influenza viruses continuously acquire mutations in antibody binding sites of hemagglutinin (HA). The majority of H3N2 strains circulating in the Northern Hemisphere during the 2014-2015 season are antigenically mismatched to the A/Texas/50/2012 H3N2 vaccine strain. Recent H3N2 strains possess several new HA mutations, and it is unknown which of these mutations contribute to the 2014-2015 vaccine mismatch. Here, we use reverse genetics to demonstrate that mutations in HA antigenic site B are primarily responsible for the current mismatch. Sera isolated from vaccinated humans and infected ferrets and sheep had reduced hemagglutination inhibition and in vitro neutralization titers against reverse-genetics-derived viruses possessing mutations in the HA antigenic site B. These data provide an antigenic explanation for the low influenza vaccine efficacy observed during the 2014-2015 influenza season. Furthermore, our data support the World Health Organization's decision to update the H3N2 component of future vaccine formulations.


Subject(s)
Antigens, Viral/genetics , Hemagglutinins/chemistry , Influenza A Virus, H3N2 Subtype/genetics , Mutation , Amino Acid Sequence , Antigens, Viral/immunology , Binding Sites, Antibody , Disease Outbreaks , Genetic Drift , Hemagglutinins/genetics , Hemagglutinins/immunology , Humans , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/isolation & purification , Molecular Sequence Data
16.
Proc Natl Acad Sci U S A ; 111(44): 15798-803, 2014 Nov 04.
Article in English | MEDLINE | ID: mdl-25331901

ABSTRACT

Influenza viruses typically cause the most severe disease in children and elderly individuals. However, H1N1 viruses disproportionately affected middle-aged adults during the 2013-2014 influenza season. Although H1N1 viruses recently acquired several mutations in the hemagglutinin (HA) glycoprotein, classic serological tests used by surveillance laboratories indicate that these mutations do not change antigenic properties of the virus. Here, we show that one of these mutations is located in a region of HA targeted by antibodies elicited in many middle-aged adults. We find that over 42% of individuals born between 1965 and 1979 possess antibodies that recognize this region of HA. Our findings offer a possible antigenic explanation of why middle-aged adults were highly susceptible to H1N1 viruses during the 2013-2014 influenza season. Our data further suggest that a drifted H1N1 strain should be included in future influenza vaccines to potentially reduce morbidity and mortality in this age group.


Subject(s)
Antigens, Viral/genetics , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/genetics , Mutation , Adult , Animals , Antigens, Viral/immunology , Female , Hemagglutinin Glycoproteins, Influenza Virus , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines , Influenza, Human/immunology , Influenza, Human/mortality , Male , Mice , Mice, Inbred BALB C , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...