Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.185
Filter
1.
Vaccine ; 42(24): 126269, 2024 Oct 24.
Article in English | MEDLINE | ID: mdl-39241354

ABSTRACT

Recombinant influenza virus neuraminidase (NA) is a promising broadly protective influenza vaccine candidate. However, the recombinant protein alone is not sufficient to induce durable and protective immune responses and requires the coadministration of immunostimulatory molecules. Here, we evaluated the immunogenicity and cross-protective potential of a recombinant influenza virus N2 neuraminidase vaccine construct, adjuvanted with a toll-like receptor 9 (TLR9) agonist (CpG 1018® adjuvant), and alum. The combination of CpG 1018 adjuvant and alum induced a balanced and robust humoral and T-cellular immune response against the NA, which provided protection and reduced morbidity against homologous and heterologous viral challenges in mouse and hamster models. This study supports Syrian hamsters as a useful complementary animal model to mice for pre-clinical evaluation of influenza virus vaccines.


Subject(s)
Adjuvants, Immunologic , Antibodies, Viral , Influenza Vaccines , Neuraminidase , Orthomyxoviridae Infections , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Neuraminidase/immunology , Neuraminidase/genetics , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Mice , Adjuvants, Immunologic/administration & dosage , Female , Cricetinae , Antibodies, Viral/immunology , Antibodies, Viral/blood , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Adjuvants, Vaccine , Mice, Inbred BALB C , Cross Protection/immunology , Mesocricetus , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/immunology , Alum Compounds/administration & dosage , Disease Models, Animal , Immunity, Cellular
2.
Hum Vaccin Immunother ; 20(1): 2403175, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39291424

ABSTRACT

Influenza neuraminidase (NA) is a promising target for a broadly protective vaccine. In this study, the Computationally Optimized Broadly Reactive Antigen (COBRA) methodology was used to develop N2 NA vaccine candidates. The unique wild type (WT) N2 sequences of human and swine influenza strains isolated between 1957 and 2019 were used to design the COBRA N2-A NA vaccine, while the unique WT N2 sequences of human influenza strains isolated between 2000 and 2019 were used to design the COBRA N2-B NA vaccine. Sera collected from COBRA N2 NA vaccinated mice showed more broadly reactive antibody responses against a broad panel of H×N2 influenza virus strains than sera collected from mice vaccinated with WT N2 NA vaccines. Antibodies elicited by COBRA or WT N2 NA antigens cross react with recent human H3N2 influenza viruses from different clades, while the antibodies elicited by A/Switzerland/9715293/2013 hemagglutinin (HA) reacted with viruses from the same clade. Furthermore, mice vaccinated with COBRA N2-B NA vaccine had lower viral lung titers compared to mock vaccinated mice when challenged with human H3N2 influenza viruses. Thus, the COBRA N2 NA vaccines elicit broadly protective murine anti-NA antibodies against multiple strains across subtypes and the viral loads were significantly decreased in the lungs of the mice in the COBRA N2 NA vaccine groups, compared to the mice in the mock vaccinated group, indicating that the COBRA-based N2 subtype NA vaccines have a potential to be a component in a universal influenza vaccine.


Subject(s)
Antibodies, Viral , Influenza A Virus, H3N2 Subtype , Influenza Vaccines , Neuraminidase , Orthomyxoviridae Infections , Animals , Female , Humans , Mice , Antibodies, Viral/blood , Antibodies, Viral/immunology , Cross Reactions/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Influenza, Human/immunology , Lung/virology , Lung/immunology , Mice, Inbred BALB C , Neuraminidase/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Viral Load , Viral Proteins/immunology
3.
Proc Natl Acad Sci U S A ; 121(41): e2408469121, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39348541

ABSTRACT

Despite much research, considerable data suggest that influenza virus remains a serious health problem because i) the effectiveness of current vaccines ranges only from 19% to 60%, ii) available therapies remain ineffective in advanced stages of disease, iii) death rates vary between 25,000 and 72,000/year in the United States, and iv) avian influenza strains are now being transmitted to dairy cattle that in turn are infecting humans. To address these concerns, we have developed zanDR, a bispecific small molecule that binds and inhibits viral neuraminidase expressed on both free virus and virus-infected cells and recruits naturally occurring anti-rhamnose and anti-dinitrophenyl (DNP) antibodies with rhamnose and DNP haptens. Because the neuraminidase inhibition replicates the chemotherapeutic mechanism of zanamivir and oseltamivir, while rhamnose and DNP recruit endogenous antibodies much like an anti-influenza vaccine, zanDR reproduces most of the functions of current methods of protection against influenza virus infections. Importantly, studies on cells in culture demonstrate that both of the above protective mechanisms remain highly functional in the zanDR conjugate, while studies in lethally infected mice with advanced-stage disease establish that a single intranasal dose of zanDR not only yields 100% protection but also reduces lung viral loads faster and ~1,000× more thoroughly than current antiviral therapies. Since zanDR also lowers secretion of proinflammatory cytokines and protects against virus-induced damage to the lungs better than current therapies, we suggest that combining an immunotherapy with a chemotherapy in single pharmacological agent constitutes a promising approach for treating the more challenging forms of influenza.


Subject(s)
Orthomyxoviridae Infections , Animals , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/drug therapy , Orthomyxoviridae Infections/virology , Humans , Antiviral Agents/pharmacology , Influenza, Human/immunology , Influenza, Human/drug therapy , Influenza, Human/prevention & control , Neuraminidase/immunology , Neuraminidase/antagonists & inhibitors , Dogs , Female , Mice, Inbred BALB C , Antibodies, Viral/immunology
4.
Elife ; 132024 Sep 25.
Article in English | MEDLINE | ID: mdl-39319780

ABSTRACT

Influenza viruses continually evolve new antigenic variants, through mutations in epitopes of their major surface proteins, hemagglutinin (HA) and neuraminidase (NA). Antigenic drift potentiates the reinfection of previously infected individuals, but the contribution of this process to variability in annual epidemics is not well understood. Here, we link influenza A(H3N2) virus evolution to regional epidemic dynamics in the United States during 1997-2019. We integrate phenotypic measures of HA antigenic drift and sequence-based measures of HA and NA fitness to infer antigenic and genetic distances between viruses circulating in successive seasons. We estimate the magnitude, severity, timing, transmission rate, age-specific patterns, and subtype dominance of each regional outbreak and find that genetic distance based on broad sets of epitope sites is the strongest evolutionary predictor of A(H3N2) virus epidemiology. Increased HA and NA epitope distance between seasons correlates with larger, more intense epidemics, higher transmission, greater A(H3N2) subtype dominance, and a greater proportion of cases in adults relative to children, consistent with increased population susceptibility. Based on random forest models, A(H1N1) incidence impacts A(H3N2) epidemics to a greater extent than viral evolution, suggesting that subtype interference is a major driver of influenza A virus infection ynamics, presumably via heterosubtypic cross-immunity.


Seasonal influenza (flu) viruses cause outbreaks every winter. People infected with influenza typically develop mild respiratory symptoms. But flu infections can cause serious illness in young children, older adults and people with chronic medical conditions. Infected or vaccinated individuals develop some immunity, but the viruses evolve quickly to evade these defenses in a process called antigenic drift. As the viruses change, they can re-infect previously immune people. Scientists update the flu vaccine yearly to keep up with this antigenic drift. The immune system fights flu infections by recognizing two proteins, known as antigens, on the virus's surface, called hemagglutinin (HA) and neuraminidase (NA). However, mutations in the genes encoding these proteins can make them unrecognizable, letting the virus slip past the immune system. Scientists would like to know how these changes affect the size, severity and timing of annual influenza outbreaks. Perofsky et al. show that tracking genetic changes in HA and NA may help improve flu season predictions. The experiments compared the severity of 22 flu seasons caused by the A(H3N2) subtype in the United States with how much HA and NA had evolved since the previous year. The A(H3N2) subtype experiences the fastest rates of antigenic drift and causes more cases and deaths than other seasonal flu viruses. Genetic changes in HA and NA were a better predictor of A(H3N2) outbreak severity than the blood tests for protective antibodies that epidemiologists traditionally use to track flu evolution. However, the prevalence of another subtype of influenza A circulating in the population, called A(H1N1), was an even better predictor of how severe A(H3N2) outbreaks would be. Perofsky et al. are the first to show that genetic changes in NA contribute to the severity of flu seasons. Previous studies suggested a link between genetic changes in HA and flu season severity, and flu vaccines include the HA protein to help the body recognize new influenza strains. The results suggest that adding the NA protein to flu vaccines may improve their effectiveness. In the future, flu forecasters may want to analyze genetic changes in both NA and HA to make their outbreak predictions. Tracking how much of the A(H1N1) subtype is circulating may also be useful for predicting the severity of A(H3N2) outbreaks.


Subject(s)
Antigenic Drift and Shift , Epidemics , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H3N2 Subtype , Influenza, Human , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , United States/epidemiology , Influenza, Human/epidemiology , Influenza, Human/virology , Influenza, Human/immunology , Humans , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Antigenic Drift and Shift/genetics , Child , Adult , Neuraminidase/genetics , Neuraminidase/immunology , Adolescent , Child, Preschool , Antigens, Viral/immunology , Antigens, Viral/genetics , Young Adult , Evolution, Molecular , Seasons , Middle Aged
5.
Antiviral Res ; 230: 105978, 2024 10.
Article in English | MEDLINE | ID: mdl-39117282

ABSTRACT

Seasonal influenza is an annually severe crisis for global public health, and an ideal influenza vaccine is expected to provide broad protection against constantly drifted strains. Compared to highly flexible hemagglutinin (HA), increasing data have demonstrated that neuraminidase (NA) might be a potential target against influenza variants. In the present study, a series of genetic algorithm-based mosaic NA were designed, and then cloned into recombinant DNA and replication-defective Vesicular Stomatitis Virus (VSV) vector as a novel influenza vaccine candidate. Our Results showed that DNA prime/VSV boost strategy elicited a robust NA-specific Th1-dominated immune response, but the traditional inactivated influenza vaccine elicited a Th2-dominated immune response. More importantly, the superior NA-specific immunity induced by our strategy could confer both a full protection against lethal homologous influenza challenge and a partial protection against heterologous influenza infection. These findings will provide insights on designing NA-based universal vaccine strategy against influenza variants.


Subject(s)
Influenza Vaccines , Neuraminidase , Orthomyxoviridae Infections , Neuraminidase/immunology , Neuraminidase/genetics , Influenza Vaccines/immunology , Influenza Vaccines/genetics , Animals , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Mice , T-Lymphocytes/immunology , Mice, Inbred BALB C , Female , Humans , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza, Human/virology , Vaccines, DNA/immunology , Vaccines, DNA/genetics , Th1 Cells/immunology , Viral Proteins/genetics , Viral Proteins/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood
6.
J Virol ; 98(9): e0035424, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39171925

ABSTRACT

Development of next-generation influenza virus vaccines is crucial to improve protection against circulating and emerging viruses. Current vaccine formulations have to be updated annually due to mutations in seasonal strains and do not offer protection against strains with pandemic potential. Computationally optimized broadly reactive antigen (COBRA) methodology has been utilized by our group to generate broadly reactive immunogens for individual influenza subtypes, which elicit protective immune responses against a broad range of strains over numerous seasons. Octavalent mixtures of COBRA hemagglutinin (HA) (H1, H2, H3, H5, H7, and influenza B virus) plus neuraminidase (NA) (N1 and N2) recombinant proteins mixed with c-di-AMP adjuvant were administered intranasally to naive or pre-immune ferrets in prime-boost fashion. Four weeks after final vaccination, collected sera were analyzed for breadth of antibody response, and the animals were challenged with seasonal or pre-pandemic strains. The octavalent COBRA vaccine elicited antibodies that recognized a broad panel of strains representing different subtypes, and these vaccinated animals were protected against influenza virus challenges. Overall, this study demonstrated that the mixture of eight COBRA HA/NA proteins mixed with an intranasal adjuvant is a promising candidate for a universal influenza vaccine. IMPORTANCE: Influenza is a respiratory virus which infects around a billion people globally every year, with millions experiencing severe illness. Commercial vaccine efficacy varies year to year and can be low due to mismatch of circulating virus strains. Thus, the formulation of current vaccines has to be adapted accordingly every year. The development of a broadly reactive influenza vaccine would lessen the global economic and public health burden caused by the different types of influenza viruses. The significance of our research is producing a promising universal vaccine candidate which provides protection against a wider range of virus strains over a wider range of time.


Subject(s)
Administration, Intranasal , Antibodies, Viral , Ferrets , Hemagglutinin Glycoproteins, Influenza Virus , Influenza Vaccines , Orthomyxoviridae Infections , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Antibodies, Viral/blood , Antibodies, Viral/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Neuraminidase/immunology , Neuraminidase/genetics , Seasons , Adjuvants, Immunologic/administration & dosage , Vaccination/methods , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza, Human/virology , Humans , Female , Cross Protection/immunology , Pandemics/prevention & control
7.
J Virol ; 98(9): e0076624, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39194245

ABSTRACT

Antibody responses to influenza vaccines tend to be focused on epitopes encountered during prior influenza exposures, with little production of de novo responses to novel epitopes. To examine the contribution of circulating antibodies to this phenomenon, we passively transferred a hemagglutinin (HA)-specific monoclonal antibody (mAb) into mice before immunizing with whole inactivated virions. The HA mAb inhibited de novo HA-specific antibodies, plasmablasts, germinal center B cells, and memory B cells, while responses to a second antigen in the vaccine, neuraminidase (NA), were uninhibited. The HA mAb potently inhibited de novo antibody responses against epitopes near the HA mAb binding site. The HA mAb also promoted IgG1 class switching, an effect that, unlike the inhibition of HA responses, relied on signaling through Fc-gamma receptors. These studies suggest that circulating antibodies inhibit de novo B cell responses in an antigen-specific manner, which likely contributes to differences in antibody specificities elicited during primary and secondary influenza virus exposures.IMPORTANCEMost humans are exposed to influenza viruses in childhood and then subsequently exposed to antigenically drifted influenza variants later in life. It is unclear if antibodies elicited by earlier influenza virus exposures impact immunity against new influenza virus strains. Here, we used a mouse model to investigate how an anti-hemagglutinin (HA) monoclonal antibody (mAb) affects de novo B cell and antibody responses to the protein targeted by the monoclonal antibody (HA) and a second protein not targeted by the monoclonal antibody [neuraminidase (NA)]. Collectively, our studies suggest that circulating anti-influenza virus antibodies can potently modulate the magnitude and specificity of antibody responses elicited by secondary influenza virus exposures.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , B-Lymphocytes , Hemagglutinin Glycoproteins, Influenza Virus , Influenza Vaccines , Animals , Mice , Antibodies, Viral/immunology , Antibodies, Monoclonal/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , B-Lymphocytes/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/immunology , Female , Neuraminidase/immunology , Mice, Inbred BALB C , Immunoglobulin G/immunology , Epitopes/immunology , Antibody Formation/immunology
8.
Vaccine ; 42(24): 126270, 2024 Oct 24.
Article in English | MEDLINE | ID: mdl-39197219

ABSTRACT

Current influenza virus vaccines poorly display key neuraminidase (NA) epitopes and do not robustly induce NA-reactive antibodies; instead, they focus on the induction of hemagglutinin (HA)-reactive antibodies. Next-generation influenza vaccines should be optimized in order to activate NA-reactive B cells and to induce a broadly cross-reactive and protective antibody response. We aimed at enhancing the immunogenicity of the NA on vaccines by two strategies: (i) modifying the HA:NA ratio of the vaccine preparation and (ii) exposing epitopes on the lateral surface or beneath the head of the NA by extending the NA stalk. The H1N1 glycoproteins from the influenza virus A/California/04/2009 strain were displayed on human immunodeficiency virus 1 (HIV-1) gag-based virus-like particles (VLP). Using the baculovirus insect cell expression system, we biased the quantity of surface glycoproteins employing two different promoters, the very late baculovirus p10 promoter and the early and late gp64 promoter. This led to a 1:1 to 2:1 HA:NA ratio, which was approximately double or triple the amount of NA as present on the wild-type influenza A virus (HA:NA ratio 3:1 to 5:1). Furthermore, by insertion of 15 amino acids from the A-New York/61/2012 strain (NY12) which prolongates the NA stalk (NA long stalk; NA-LS), we intended to improve the accessibility of the NA. Six different types of VLPs were produced and purified using a platform downstream process based on Capto-Core 700™ followed by Capto-Heparin™ affinity chromatography combined with ultracentrifugation. These VLPs were then tested in a mouse model. Robust titers of antibodies that inhibit the neuraminidase activity were elicited even after vaccination with two low doses (0.3 µg) of the H1N1 VLPs without compromising the anti-HA responses. In conclusion, our results demonstrate the feasibility of the two developed strategies to retain HA immunogenicity and improve NA immunogenicity as a future influenza vaccine candidate.


Subject(s)
Antibodies, Viral , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Neuraminidase , Vaccines, Virus-Like Particle , Animals , Neuraminidase/immunology , Neuraminidase/genetics , Influenza Vaccines/immunology , Vaccines, Virus-Like Particle/immunology , Influenza A Virus, H1N1 Subtype/immunology , Antibodies, Viral/immunology , Mice , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Immunogenicity, Vaccine , Mice, Inbred BALB C , Female , HIV-1/immunology , HIV-1/genetics , Epitopes/immunology , Humans
9.
Vet Microbiol ; 296: 110188, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39018942

ABSTRACT

H9N2 avian influenza virus (AIV), one of the predominant subtypes circulating in the poultry industry, inflicts substantial economic damage. Mutations in the hemagglutinin (HA) and neuraminidase (NA) proteins of H9N2 frequently alter viral antigenicity and replication. In this paper, we analyzed the HA genetic sequences and antigenic properties of 26 H9N2 isolates obtained from chickens in China between 2012 and 2019. The results showed that these H9N2 viruses all belonged to h9.4.2.5, and were divided into two clades. We assessed the impact of amino acid substitutions at HA sites 145, 149, 153, 164, 167, 168, and 200 on antigenicity, and found that a mutation at site 164 significantly modified antigenic characteristics. Amino acid variations at sites 145, 153, 164 and 200 affected virus's hemagglutination and the growth kinetics in mammalian cells. These results underscore the critical need for ongoing surveillance of the H9N2 virus and provide valuable insights for vaccine development.


Subject(s)
Chickens , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H9N2 Subtype , Influenza in Birds , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/immunology , Animals , Chickens/virology , Influenza in Birds/virology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , China , Amino Acid Substitution , Poultry Diseases/virology , Mutation , Antigens, Viral/immunology , Antigens, Viral/genetics , Virus Replication , Phylogeny , Neuraminidase/genetics , Neuraminidase/immunology , Amino Acids/genetics
10.
Nat Commun ; 15(1): 5593, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961067

ABSTRACT

Human cases of avian influenza virus (AIV) infections are associated with an age-specific disease burden. As the influenza virus N2 neuraminidase (NA) gene was introduced from avian sources during the 1957 pandemic, we investigate the reactivity of N2 antibodies against A(H9N2) AIVs. Serosurvey of healthy individuals reveal the highest rates of AIV N2 antibodies in individuals aged ≥65 years. Exposure to the 1968 pandemic N2, but not recent N2, protected against A(H9N2) AIV challenge in female mice. In some older adults, infection with contemporary A(H3N2) virus could recall cross-reactive AIV NA antibodies, showing discernable human- or avian-NA type reactivity. Individuals born before 1957 have higher anti-AIV N2 titers compared to those born between 1957 and 1968. The anti-AIV N2 antibodies titers correlate with antibody titers to the 1957 N2, suggesting that exposure to the A(H2N2) virus contribute to this reactivity. These findings underscore the critical role of neuraminidase immunity in zoonotic and pandemic influenza risk assessment.


Subject(s)
Antibodies, Viral , Cross Reactions , Influenza A Virus, H3N2 Subtype , Influenza, Human , Neuraminidase , Pandemics , Neuraminidase/immunology , Neuraminidase/genetics , Animals , Humans , Antibodies, Viral/immunology , Antibodies, Viral/blood , Influenza A Virus, H3N2 Subtype/immunology , Female , Cross Reactions/immunology , Mice , Influenza, Human/immunology , Influenza, Human/epidemiology , Influenza, Human/virology , Aged , Influenza A Virus, H2N2 Subtype/immunology , Influenza A Virus, H2N2 Subtype/genetics , Male , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/epidemiology , Orthomyxoviridae Infections/veterinary , Birds/virology , Middle Aged , Influenza in Birds/epidemiology , Influenza in Birds/immunology , Influenza in Birds/virology , Influenza A Virus, H9N2 Subtype/immunology , Adult , Viral Proteins/immunology , Viral Proteins/genetics
11.
Immunity ; 57(6): 1413-1427.e9, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38823390

ABSTRACT

Influenza B viruses (IBVs) comprise a substantial portion of the circulating seasonal human influenza viruses. Here, we describe the isolation of human monoclonal antibodies (mAbs) that recognized the IBV neuraminidase (NA) glycoprotein from an individual following seasonal vaccination. Competition-binding experiments suggested the antibodies recognized two major antigenic sites. One group, which included mAb FluB-393, broadly inhibited IBV NA sialidase activity, protected prophylactically in vivo, and bound to the lateral corner of NA. The second group contained an active site mAb, FluB-400, that broadly inhibited IBV NA sialidase activity and virus replication in vitro in primary human respiratory epithelial cell cultures and protected against IBV in vivo when administered systemically or intranasally. Overall, the findings described here shape our mechanistic understanding of the human immune response to the IBV NA glycoprotein through the demonstration of two mAb delivery routes for protection against IBV and the identification of potential IBV therapeutic candidates.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Influenza B virus , Influenza, Human , Neuraminidase , Neuraminidase/immunology , Humans , Influenza B virus/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza Vaccines/immunology , Mice , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Viral Proteins/immunology , Virus Replication/drug effects
12.
Virology ; 597: 110119, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38850895

ABSTRACT

Highly pathogenic avian influenza (HPAI) viruses remain a major threat to both the poultry industry and human public health, and these viruses continue to spread worldwide. In this study, mice were vaccinated with COBRA H2, H5, and H7 hemagglutinin (HA) and two neuraminidase (NA) proteins, N1 and N2. Vaccinated mice were fully protected against lethal challenge with H5N6 influenza virus. Sera collected after vaccination showed cross-reactive IgG antibodies against a panel of wild-type H2, H5, and H7 HA proteins, and N1 and N2 NA proteins. Mice with pre-existing immunity to H1N1 and H3N2 influenza viruses that were subsequently vaccinated with COBRA HA/NA vaccines had enhanced anti-HA stem antibodies compared to vaccinated mice without pre-existing immunity. In addition, sera collected after vaccination had hemagglutinin inhibitory activity against a panel of H2Nx, H5Nx, and H7Nx influenza viruses. These protective antibodies were maintained up for up to 4 months after vaccination.


Subject(s)
Antibodies, Viral , Hemagglutinin Glycoproteins, Influenza Virus , Influenza Vaccines , Neuraminidase , Orthomyxoviridae Infections , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Neuraminidase/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Mice , Mice, Inbred BALB C , Female , Vaccination , Influenza A virus/immunology , Immunoglobulin G/blood , Immunoglobulin G/immunology , Cross Reactions , Humans , Viral Proteins/immunology , Viral Proteins/genetics
13.
Front Immunol ; 15: 1425842, 2024.
Article in English | MEDLINE | ID: mdl-38915410

ABSTRACT

Vaccination against influenza virus can reduce the risk of influenza by 40% to 60%, they rely on the production of neutralizing antibodies specific to influenza hemagglutinin (HA) ignoring the neuraminidase (NA) as an important surface target. Vaccination with standardized NA concentration may offer broader and longer-lasting protection against influenza infection. In this regard, we aimed to compare the potency of a NA displayed on the surface of a VLP with a soluble NA. The baculovirus expression system (BEVS) and the novel virus-free Tnms42 insect cell line were used to express N2 NA on gag-based VLPs. To produce VLP immunogens with high levels of purity and concentration, a two-step chromatography purification process combined with ultracentrifugation was used. In a prime/boost vaccination scheme, mice vaccinated with 1 µg of the N2-VLPs were protected from mortality, while mice receiving the same dose of unadjuvanted NA in soluble form succumbed to the lethal infection. Moreover, NA inhibition assays and NA-ELISAs of pre-boost and pre-challenge sera confirm that the VLP preparation induced higher levels of NA-specific antibodies outperforming the soluble unadjuvanted NA.


Subject(s)
Antibodies, Viral , Influenza Vaccines , Neuraminidase , Orthomyxoviridae Infections , Vaccines, Virus-Like Particle , Animals , Neuraminidase/immunology , Neuraminidase/genetics , Influenza Vaccines/immunology , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/administration & dosage , Mice , Antibodies, Viral/immunology , Antibodies, Viral/blood , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Female , Mice, Inbred BALB C , Recombinant Proteins/immunology , Recombinant Proteins/genetics , Vaccine Efficacy , Humans , Vaccination/methods
14.
mBio ; 15(7): e0108524, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38899870

ABSTRACT

Monoclonal antibodies (mAbs) are an attractive therapeutic platform for the prevention and treatment of influenza virus infection. There are two major glycoproteins on the influenza virion surface: hemagglutinin (HA), which is responsible for viral attachment and entry, and neuraminidase (NA), which mediates viral egress by enzymatically cleaving sialic acid to release budding particles from the host cell surface. Broadly neutralizing antibodies (bNAbs) that target the conserved HA central stalk region, such as CR9114, can inhibit both viral entry and egress. More recently, broadly binding mAbs that engage and inhibit the NA active site, such as 1G01, have been described to prevent viral egress. Here, we engineered bispecific antibodies (bsAbs) that combine the variable domains of CR9114 and 1G01 into a single molecule and evaluated if simultaneous targeting of two different glycoproteins improved antiviral properties in vitro and in vivo. Several CR9114/1G01 bsAbs were generated with various configurations of the two sets of the variable domains ("bsAb formats"). We found that combinations employing the addition of a single-chain variable fragment in the hinge region of an IgG scaffold had the best properties in terms of expression, stability, and binding. Further characterization of selected bsAbs showed potent neutralizing and egress-inhibiting activity. One such bsAb ("hSC_CR9114_1G01") provided higher levels of prophylactic protection from mortality and morbidity upon challenge with H1N1 than either of the parental mAbs at low dosing (1 mg/kg). These results highlight the potential use of bsAbs that simultaneously target HA and NA as new influenza immunotherapeutics. IMPORTANCE: Infection by the influenza virus remains a global health burden. The approaches utilized here to augment the activity of broadly protective influenza virus antibodies may lead to a new class of immunotherapies with enhanced activity.


Subject(s)
Antibodies, Bispecific , Antibodies, Viral , Hemagglutinin Glycoproteins, Influenza Virus , Neuraminidase , Neuraminidase/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , Antibodies, Viral/immunology , Animals , Humans , Mice , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Antibodies, Neutralizing/immunology , Antibodies, Monoclonal/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Influenza, Human/virology , Mice, Inbred BALB C , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H1N1 Subtype/drug effects
15.
Clin Nephrol ; 102(2): 89-96, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38856027

ABSTRACT

INTRODUCTION: IgA nephropathy (IgAN) is a kidney disorder characterized by the deposition of circulating immune complexes of IgG bound to galactose-deficient IgA1 (Gd-IgA1) in the mesangial glomeruli. However, limited research has been conducted on the levels of IgA binding in relation to the various sialylation profiles of IgG in IgAN. MATERIALS AND METHODS: Sialylated IgG (SA-IgG) and desialylated IgG (DSA-IgG) were isolated from IgAN patients. The IgG-IgA immune complex (IgG-IgA-IC) was detected using two customized commercial ELISA kits. Additionally, IgG was enzymatically digested with neuraminidase to produce DSA-IgG. Subsequently, the binding capacities of both intact IgG and the neuraminidase-digested DSA-IgG with Gd-IgA1 were determined using ELISA kits. RESULTS: Our research revealed that SA-IgG levels were negatively correlated with Gd-IgA1 (R = -0.16, p = 0.03) in IgAN patients. The optical density (OD) levels of IgG-IgA complexes in SA-IgG samples were significantly lower (0.58 ± 0.09) compared to those in DSA-IgG samples (0.78 ± 0.12) when using the Gd-IgA1 assay kit. These results were confirmed using an IgG assay kit, which showed that the SA-IgG groups had significantly lower IgA indices (0.31 ± 0.12) compared to the DSA-IgG groups (0.57 ± 0.19). Furthermore, we investigated the binding capacity of IgG with different sialic acid levels to Gd-IgA1. The results revealed that neuraminidase digestion of IgG increased its propensity to bind to Gd-IgA1. Additionally, we examined the binding capacity of both intact IgG and DSA-IgG to Gd-IgA1 at different mix ratios (IgG 1.5 µg and Gd-IgA1 1.5 µg, IgG 1.5 µg and Gd-IgA1 3 µg, IgG 3 µg and Gd-IgA1 1.5 µg). Interestingly, DSA-IgG demonstrated significantly higher binding capacity to Gd-IgA1 compared to intact IgG at all mix ratios tested. CONCLUSION: The preliminary findings from our present study indicate that the binding level of IgA in purified sialylated IgG is lower than that in desialylated IgG.


Subject(s)
Glomerulonephritis, IGA , Immunoglobulin A , Immunoglobulin G , Humans , Glomerulonephritis, IGA/immunology , Glomerulonephritis, IGA/metabolism , Immunoglobulin A/metabolism , Immunoglobulin A/immunology , Immunoglobulin G/metabolism , Immunoglobulin G/immunology , Immunoglobulin G/blood , Male , Female , Adult , Middle Aged , Antigen-Antibody Complex/metabolism , Antigen-Antibody Complex/immunology , Young Adult , Enzyme-Linked Immunosorbent Assay , N-Acetylneuraminic Acid/metabolism , Neuraminidase/metabolism , Neuraminidase/immunology
16.
FEMS Microbiol Rev ; 48(3)2024 May 08.
Article in English | MEDLINE | ID: mdl-38734891

ABSTRACT

Avian influenza viruses evolve antigenically to evade host immunity. Two influenza A virus surface glycoproteins, the haemagglutinin and neuraminidase, are the major targets of host immunity and undergo antigenic drift in response to host pre-existing humoral and cellular immune responses. Specific sites have been identified as important epitopes in prominent subtypes such as H5 and H7, which are of animal and public health significance due to their panzootic and pandemic potential. The haemagglutinin is the immunodominant immunogen, it has been extensively studied, and the antigenic reactivity is closely monitored to ensure candidate vaccine viruses are protective. More recently, the neuraminidase has received increasing attention for its role as a protective immunogen. The neuraminidase is expressed at a lower abundance than the haemagglutinin on the virus surface but does elicit a robust antibody response. This review aims to compile the current information on haemagglutinin and neuraminidase epitopes and immune escape mutants of H5 and H7 highly pathogenic avian influenza viruses. Understanding the evolution of immune escape mutants and the location of epitopes is critical for identification of vaccine strains and development of broadly reactive vaccines that can be utilized in humans and animals.


Subject(s)
Birds , Epitopes , Hemagglutinin Glycoproteins, Influenza Virus , Influenza in Birds , Neuraminidase , Neuraminidase/immunology , Neuraminidase/genetics , Animals , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Epitopes/immunology , Epitopes/genetics , Birds/virology , Influenza in Birds/immunology , Influenza in Birds/virology , Antigenic Drift and Shift/immunology , Humans , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/genetics , Influenza, Human/immunology , Influenza, Human/virology , Influenza, Human/prevention & control , Viral Proteins/immunology , Viral Proteins/genetics , Viral Proteins/chemistry , Influenza A virus/immunology , Influenza A virus/genetics
17.
PLoS One ; 19(5): e0302865, 2024.
Article in English | MEDLINE | ID: mdl-38723016

ABSTRACT

Influenza A viruses (IAVs) continue to pose a huge threat to public health, and their prevention and treatment remain major international issues. Neuraminidase (NA) is the second most abundant surface glycoprotein on influenza viruses, and antibodies to NA have been shown to be effective against influenza infection. In this study, we generated a monoclonal antibody (mAb), named FNA1, directed toward N1 NAs. FNA1 reacted with H1N1 and H5N1 NA, but failed to react with the NA proteins of H3N2 and H7N9. In vitro, FNA1 displayed potent antiviral activity that mediated both NA inhibition (NI) and blocking of pseudovirus release. Moreover, residues 219, 254, 358, and 388 in the NA protein were critical for FNA1 binding to H1N1 NA. However, further validation is necessary to confirm whether FNA1 mAb is indeed a good inhibitor against NA for application against H1N1 and H5N1 viruses.


Subject(s)
Antibodies, Monoclonal , Influenza A Virus, H1N1 Subtype , Neuraminidase , Neuraminidase/immunology , Neuraminidase/metabolism , Neuraminidase/antagonists & inhibitors , Antibodies, Monoclonal/immunology , Influenza A Virus, H1N1 Subtype/immunology , Humans , Animals , Antibodies, Viral/immunology , Mice , Influenza A Virus, H5N1 Subtype/immunology , Mice, Inbred BALB C , Antiviral Agents/pharmacology , Viral Proteins/immunology , Viral Proteins/metabolism , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H7N9 Subtype/immunology
18.
Elife ; 122024 May 28.
Article in English | MEDLINE | ID: mdl-38805550

ABSTRACT

Human H3N2 influenza viruses are subject to rapid antigenic evolution which translates into frequent updates of the composition of seasonal influenza vaccines. Despite these updates, the effectiveness of influenza vaccines against H3N2-associated disease is suboptimal. Seasonal influenza vaccines primarily induce hemagglutinin-specific antibody responses. However, antibodies directed against influenza neuraminidase (NA) also contribute to protection. Here, we analysed the antigenic diversity of a panel of N2 NAs derived from human H3N2 viruses that circulated between 2009 and 2017. The antigenic breadth of these NAs was determined based on the NA inhibition (NAI) of a broad panel of ferret and mouse immune sera that were raised by infection and recombinant N2 NA immunisation. This assessment allowed us to distinguish at least four antigenic groups in the N2 NAs derived from human H3N2 viruses that circulated between 2009 and 2017. Computational analysis further revealed that the amino acid residues in N2 NA that have a major impact on susceptibility to NAI by immune sera are in proximity of the catalytic site. Finally, a machine learning method was developed that allowed to accurately predict the impact of mutations that are present in our N2 NA panel on NAI. These findings have important implications for the renewed interest to develop improved influenza vaccines based on the inclusion of a protective NA antigen formulation.


Two proteins, the hemagglutinin and the neuraminidase, protrude from the surface of the influenza virus. Their detection by the immune system allows the host organism to mount defences against the viral threat. The virus evolves in response to this pressure, which manifests as changes in the appearance of its hemagglutinin and neuraminidase. This process, known as antigenic drift, leads to the proteins evading detection. It is also why flu vaccines require frequent updates, as they rely on 'training' the immune system to recognise the most important strains in circulation ­ primarily by exposing it to appropriate versions of hemagglutinin. While the antigenic drift of hemagglutinin has been extensively studied, much less is known about how the neuraminidase accumulates mutations, and how these affect the immune response. To investigate this question, Catani et al. selected 43 genetically distant neuraminidases from human viral samples isolated between 2009 and 2017. Statistical analyses were applied to define their relatedness, revealing that a group of closely related neuraminidases predominated from 2009 to 2015, before they were being taken over by a second group. A third group, which was identified in viruses isolated in 2013, was remarkably close to the neuraminidase of strains that circulated in the late 1990s. The fourth and final group of neuraminidases was derived from influenza viruses that normally circulate in pigs but can also occasionally infect humans. Next, Catani et al. examined the immune response that these 43 neuraminidases could elicit in mice, as well as in ferrets ­ the animal most traditionally used in influenza research. This allowed them to pinpoint which changes in the neuraminidase sequences were important to escape recognition by the host. Data obtained from the two model species were comparable, suggesting that these experiments could be conducted on mice going forward, which are easier to work with than ferrets. Finally, Catani et al. used machine learning to build a computational model that could predict how strongly the immune system would respond to a specific neuraminidase variant. These findings could help guide the development of new vaccines that include neuraminidases tailored to best prime and train the immune system against a larger variety of strains. This may aid the development of 'supra-seasonal' vaccines that protect against a broad range of influenza viruses, reducing the need for yearly updates.


Subject(s)
Antigens, Viral , Ferrets , Influenza A Virus, H3N2 Subtype , Influenza, Human , Neuraminidase , Neuraminidase/immunology , Neuraminidase/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/enzymology , Humans , Animals , Antigens, Viral/immunology , Antigens, Viral/genetics , Mice , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza, Human/virology , Antibodies, Viral/immunology , Influenza Vaccines/immunology , Antigenic Variation , Viral Proteins/immunology , Viral Proteins/genetics , Viral Proteins/chemistry , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology
19.
Expert Rev Vaccines ; 23(1): 474-484, 2024.
Article in English | MEDLINE | ID: mdl-38632930

ABSTRACT

INTRODUCTION: Anti-neuraminidase (NA) immunity correlates with the protection against influenza virus infection in both human and animal models. The aim of this review is to better understand the mechanism of anti-NA immunity, and also to evaluate the approaches on developing NA-based influenza vaccines or enhancing immune responses against NA for current influenza vaccines. AREAS COVERED: In this review, the structure of influenza neuraminidase, the contribution of anti-NA immunity to protection, as well as the efforts and challenges of targeting the immune responses to NA were discussed. We also listed some of the newly discovered anti-NA monoclonal antibodies and discussed their contribution in therapeutic as well as the antigen design of a broadly protective NA vaccine. EXPERT OPINION: Targeting the immune response to both HA and NA may be critical for achieving the optimal protection since there are different mechanisms of HA and NA elicited protective immunity. Monoclonal antibodies (mAbs) that target the conserved protective lateral face or catalytic sites are effective therapeutics. The epitope discovery using monoclonal antibodies may benefit NA-based vaccine elicited broadly reactive antibody responses. Therefore, the potential for a vaccine that elicits cross-reactive antibodies against neuraminidase is a high priority for next-generation influenza vaccines.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Influenza Vaccines , Influenza, Human , Neuraminidase , Humans , Neuraminidase/immunology , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Antibodies, Monoclonal/immunology , Animals , Antibodies, Viral/immunology , Vaccine Development , Cross Reactions/immunology , Epitopes/immunology
20.
Virology ; 595: 110097, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38685171

ABSTRACT

Current influenza vaccine is not effective in providing cross-protection against variants. We evaluated the immunogenicity and efficacy of multi-subtype neuraminidase (NA) and M2 ectodomain virus-like particle (m-cNA-M2e VLP) and chimeric M2e-H3 stalk protein vaccines (M2e-H3 stalk) in ferrets. Our results showed that ferrets with recombinant m-cNA-M2e VLP or M2e-H3 stalk vaccination induced multi-vaccine antigen specific IgG antibodies (M2e, H3 stalk, NA), NA inhibition, antibody-secreting cells, and IFN-γ secreting cell responses. Ferrets immunized with either m-cNA-M2e VLP or M2e-H3 stalk vaccine were protected from H1N1 and H3N2 influenza viruses by lowering viral titers in nasal washes, trachea, and lungs after challenge. Vaccinated ferret antisera conferred broad humoral immunity in naïve mice. Our findings provide evidence that immunity to M2e and HA-stalk or M2e plus multi-subtype NA proteins induces cross-protection in ferrets.


Subject(s)
Antibodies, Viral , Cross Protection , Ferrets , Influenza A Virus, H1N1 Subtype , Influenza A Virus, H3N2 Subtype , Influenza Vaccines , Neuraminidase , Orthomyxoviridae Infections , Vaccines, Virus-Like Particle , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Cross Protection/immunology , Antibodies, Viral/immunology , Neuraminidase/immunology , Neuraminidase/genetics , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H1N1 Subtype/immunology , Vaccines, Virus-Like Particle/immunology , Vaccines, Virus-Like Particle/administration & dosage , Mice , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics , Female , Immunoglobulin G/blood , Immunoglobulin G/immunology , Viroporin Proteins , Viral Proteins
SELECTION OF CITATIONS
SEARCH DETAIL