Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
ACS Infect Dis ; 10(5): 1552-1560, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38623820

ABSTRACT

Tyrosine cross-linking has recently been used to produce nanoclusters (NCs) from peptides to enhance their immunogenicity. In this study, NCs were generated using the ectodomain of the ion channel Matrix 2 (M2e) protein, a conserved influenza surface antigen. The NCs were administered via intranasal (IN) or intramuscular (IM) routes in a mouse model in a prime-boost regimen in the presence of the adjuvant CpG. After boost, a significant increase in anti-M2e IgG and its subtypes was observed in the serum and lungs of mice vaccinated through the IM and IN routes; however, significant enhancement in anti-M2e IgA in lungs was observed only in the IN group. Analysis of cytokine concentrations in stimulated splenocyte cultures indicated a Th1/Th17-biased response. Mice were challenged with a lethal dose of A/California/07/2009 (H1N1pdm), A/Puerto Rico/08/1934 (H1N1), or A/Hong Kong/08/1968 (H3N2) strains. Mice that received M2e NCs + CpG were significantly protected against these strains and showed decreased lung viral titers compared with the naive mice and M2e NC-alone groups. The IN-vaccinated group showed superior protection against the H3N2 strain as compared to the IM group. This research extends our earlier efforts involving the tyrosine-based cross-linking method and highlights the potential of this technology in enhancing the immunogenicity of short peptide immunogens.


Subject(s)
Antibodies, Viral , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Orthomyxoviridae Infections , Tyrosine , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Mice , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Tyrosine/chemistry , Tyrosine/pharmacology , Influenza A Virus, H1N1 Subtype/immunology , Female , Antibodies, Viral/blood , Antibodies, Viral/immunology , Viral Matrix Proteins/immunology , Viral Matrix Proteins/genetics , Mice, Inbred BALB C , Influenza A Virus, H3N2 Subtype/immunology , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/administration & dosage , Lung/virology , Lung/immunology , Administration, Intranasal , Injections, Intramuscular , Cytokines , Cross Protection , Viroporin Proteins
2.
Nature ; 618(7965): 590-597, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37258672

ABSTRACT

Rapidly evolving influenza A viruses (IAVs) and influenza B viruses (IBVs) are major causes of recurrent lower respiratory tract infections. Current influenza vaccines elicit antibodies predominantly to the highly variable head region of haemagglutinin and their effectiveness is limited by viral drift1 and suboptimal immune responses2. Here we describe a neuraminidase-targeting monoclonal antibody, FNI9, that potently inhibits the enzymatic activity of all group 1 and group 2 IAVs, as well as Victoria/2/87-like, Yamagata/16/88-like and ancestral IBVs. FNI9 broadly neutralizes seasonal IAVs and IBVs, including the immune-evading H3N2 strains bearing an N-glycan at position 245, and shows synergistic activity when combined with anti-haemagglutinin stem-directed antibodies. Structural analysis reveals that D107 in the FNI9 heavy chain complementarity-determinant region 3 mimics the interaction of the sialic acid carboxyl group with the three highly conserved arginine residues (R118, R292 and R371) of the neuraminidase catalytic site. FNI9 demonstrates potent prophylactic activity against lethal IAV and IBV infections in mice. The unprecedented breadth and potency of the FNI9 monoclonal antibody supports its development for the prevention of influenza illness by seasonal and pandemic viruses.


Subject(s)
Antibodies, Viral , Antibody Specificity , Influenza A virus , Influenza B virus , Influenza Vaccines , Influenza, Human , Molecular Mimicry , Neuraminidase , Animals , Humans , Mice , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , Antibody Specificity/immunology , Arginine/chemistry , Catalytic Domain , Hemagglutinins, Viral/immunology , Influenza A virus/classification , Influenza A virus/enzymology , Influenza A virus/immunology , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H3N2 Subtype/immunology , Influenza B virus/classification , Influenza B virus/enzymology , Influenza B virus/immunology , Influenza Vaccines/chemistry , Influenza Vaccines/immunology , Influenza Vaccines/therapeutic use , Influenza, Human/immunology , Influenza, Human/prevention & control , Neuraminidase/antagonists & inhibitors , Neuraminidase/chemistry , Neuraminidase/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Seasons , Sialic Acids/chemistry
3.
Phytomedicine ; 91: 153668, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34385093

ABSTRACT

BACKGROUND: Aloe vera is a functional food with various pharmacological functions, including an immune-modulating effect. Until now, A. vera has never been studied as an adjuvant in influenza vaccine, and its effects on upper respiratory tract infection (URI) are unknown. PURPOSE: The objective of our study was to investigate the effect of processed A. vera gel (PAG) on immunogenicity of quadrivalent inactivated influenza vaccine and URI in healthy adults. STUDY DESIGN: A randomized, double-blind, placebo-controlled clinical trial was performed. METHODS: This study was conducted in 100 healthy adults at a single center from September 2017 to May 2018. Subjects were randomly divided into a PAG group (n = 50) and a placebo group (n = 50). The enrolled subjects were instructed to ingest the study drug for 8 weeks. The participants received a single dose of quadrivalent inactivated influenza vaccine after taking the study drug for the first 4 weeks of the study. The primary endpoint was seroprotection rate against at least one viral strain at 4 weeks post-vaccination. Other outcomes were seroprotection rate at 24 weeks post-vaccination, seroconversion rate, geometric mean fold increase (GMFI) at 4 and 24 weeks post-vaccination, seroprotection rate ratio and geometric mean titer ratio (GMTR) at 4 weeks post-vaccination between PAG and placebo groups, and incidence, severity, and duration of URI. RESULTS: The European Committee for proprietary medicinal products (CPMP) evaluation criteria were met at least one in the PAG and placebo groups for all strains. However, there was no significant difference in the seroprotection rate at 4 weeks post-vaccination against all strains in both PAG and placebo groups. Among secondary endpoints, the GMFI at 4 weeks post-vaccination for the A/H3N2 was significantly higher in the PAG than in placebo group. The GMTR as adjuvant effect was 1.382 (95% CI, 1.014-1.1883). Kaplan-Meier curve analysis showed a reduction in incidence of URI (p = 0.035), and a generalized estimating equation model identified a decrease in repeated URI events (odds ratio 0.57; 95% CI, 0.39-0.83; p = 0.003) in the PAG group. CONCLUSIONS: Oral intake of PAG did not show a significant increase in seroprotection rate from an immunogenicity perspective. However, it reduced the number of URI episodes. A well-designed further study is needed on the effect of PAG's antibody response against A/H3N2 in the future.


Subject(s)
Adjuvants, Immunologic , Immunogenicity, Vaccine , Influenza Vaccines , Influenza, Human , Plant Preparations/chemistry , Adult , Double-Blind Method , Hemagglutination Inhibition Tests , Humans , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/prevention & control
4.
Viruses ; 13(3)2021 03 18.
Article in English | MEDLINE | ID: mdl-33803680

ABSTRACT

BACKGROUND: Data suggest that pediatric patients might react differently to influenza vaccination, both in terms of immunity and side effects. We have recently shown that using a whole virion vaccine with aluminum phosphate adjuvants, reduced dose vaccines containing 6 µg of viral hemagglutinin (HA) per strain are immunogenic, and well tolerated in adult and elderly patients. Here we show the results of a multicenter clinical trial of pediatric patients, using reduced doses of a new, whole virion, aluminum phosphate adjuvanted vaccine (FluArt, Budapest, Hungary). METHODS: A total of 120 healthy volunteers were included in two age groups (3-11 years, receiving 3 µg of HA per strain, and 12-18 years, receiving 6 µg of HA per strain). We used hemagglutination inhibition testing to assess immunogenicity, based on EMA and FDA licensing criteria, including post/pre-vaccination geometric mean titer ratios, seroconversion and seropositivity rates. Safety and tolerability were assessed using CHMP guidelines. RESULTS: All subjects entered the study and were vaccinated (ITT population). All 120 subjects attended the control visit on Day 21 (PP population). All immunogenicity licensing criteria were met in both age groups for all three vaccine virus strains. No serious adverse events were detected and the vaccine was well tolerated by both age groups. DISCUSSION: Using a whole virion vaccine and aluminum phosphate adjuvants, a reduction in the amount of the viral hemmaglutinin is possible while maintaining immunogenicity, safety and tolerability in pediatric and adolescent patients.


Subject(s)
Adjuvants, Immunologic , Aluminum Compounds , Influenza Vaccines , Influenza, Human/prevention & control , Phosphates , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/adverse effects , Adolescent , Aluminum Compounds/administration & dosage , Aluminum Compounds/adverse effects , Child , Child, Preschool , Female , Humans , Hungary/epidemiology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza B virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/adverse effects , Male , Phosphates/administration & dosage , Phosphates/adverse effects , Prospective Studies , Virion/immunology
5.
Phytomedicine ; 64: 152904, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31454654

ABSTRACT

BACKGROUND: Millions of people are infected by the influenza virus worldwide every year. Current selections of anti-influenza agents are limited and their effectiveness and drug resistance are still of concern. PURPOSE: Investigation on in vitro and in vivo effect of aloin from Aloe vera leaves against influenza virus infection. METHODS: In vitro antiviral property of aloin was measured by plaque reduction assay in which MDCK cells were infected with oseltamivir-sensitive A(H1N1)pdm09, oseltamivir-resistant A(H1N1)pdm09, H1N1 or H3N2 influenza A or with influenza B viruses in the presence of aloin. In vivo activity was tested in H1N1 influenza virus infected mice. Aloin-mediated inhibition of influenza neuraminidase activity was tested by MUNANA assay. Aloin treatment-mediated modulation of anti-influenza immunity was tested by the study of hemagglutinin-specific T cells in vivo. RESULTS: Aloin significantly reduced in vitro infection by all the tested strains of influenza viruses, including oseltamivir-resistant A(H1N1)pdm09 influenza viruses, with an average IC50 value 91.83 ± 18.97 µM. In H1N1 influenza virus infected mice, aloin treatment (intraperitoneal, once daily for 5 days) reduced virus load in the lungs and attenuated body weight loss and mortality. Adjuvant aloin treatment also improved the outcome with delayed oseltamivir treatment. Aloin inhibited viral neuraminidase and impeded neuraminidase-mediated TGF-ß activation. Viral neuraminidase mediated immune suppression with TGF-ß was constrained and influenza hemagglutinin-specific T cell immunity was increased. There was more infiltration of hemagglutinin-specific CD4+ and CD8+ T cells in the lungs and their production of effector cytokines IFN-γ and TNF-α was boosted. CONCLUSION: Aloin from Aloe vera leaves is a potent anti-influenza compound that inhibits viral neuraminidase activity, even of the oseltamivir-resistant influenza virus. With suppression of this virus machinery, aloin boosts host immunity with augmented hemagglutinin-specific T cell response to the infection. In addition, in the context of compromised benefit with delayed oseltamivir treatment, adjuvant aloin treatment ameliorates the disease and improves survival. Taken together, aloin has the potential to be further evaluated for clinical applications in human influenza.


Subject(s)
Aloe/chemistry , Antiviral Agents/pharmacology , Emodin/analogs & derivatives , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Influenza B virus/drug effects , Influenza, Human/drug therapy , Neuraminidase/antagonists & inhibitors , Animals , Cell Line , Drug Resistance, Viral , Emodin/pharmacology , Hemagglutinins/immunology , Humans , Influenza A Virus, H1N1 Subtype/enzymology , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/enzymology , Influenza A Virus, H3N2 Subtype/immunology , Influenza B virus/enzymology , Influenza B virus/immunology , Influenza, Human/immunology , Influenza, Human/virology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oseltamivir/pharmacology , Plant Leaves/chemistry , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Viral Proteins/antagonists & inhibitors
6.
Vaccine ; 37(28): 3638-3645, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31155418

ABSTRACT

Adjuvants are chemical/biological substances that are used in vaccines to increase the immunogenicity of antigens. A few adjuvants have been developed for use in human vaccines because of their limitations including lack of efficacy, unacceptable local or systemic toxicity, the difficulty of manufacturing, poor stability, and high cost. For that reasons, novel adjuvants/adjuvant systems are under search. Astragaloside VII (AST-VII), isolated from Astragalus trojanus, exhibited significant cellular and humoral immune responses. The polysaccharides (APS) obtained from the roots of Astragalus species have been used in traditional Chinese medicine and possess strong immunomodulatory properties. In the present study, the immunomodulatory effects of a newly developed nanocarrier system (APNS: APS containing carrier) and its AST-VII containing formulation (ANS: AST-VII + APNS), on seasonal influenza A (H3N2) vaccine were investigated. Inactivated H3N2 alone or its combinations with test compounds/formulations were intramuscularly injected into Swiss albino mice. Four weeks after immunization, the immune responses were evaluated in terms of antibody and cytokine responses as well as splenocyte proliferation. APNS demonstrated Th2 mediated response by increasing IgG1 antibody titers, whereas ANS showed response towards Th1/Th2 balance and Th17 by producing of IFN-γ, IL-17A and IgG2a. Based on these results, we propose that APNS and ANS are good candidates to be utilized in seasonal influenza A vaccines as adjuvants/carrier systems.


Subject(s)
Drug Carriers/chemistry , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Nanoparticles/chemistry , Orthomyxoviridae Infections/immunology , Saponins/chemistry , Tragacanth/chemistry , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/chemistry , Animals , Cytokines/immunology , Female , Immunity, Cellular/immunology , Immunity, Humoral/immunology , Immunoglobulin G/immunology , Influenza Vaccines/chemistry , Male , Mice , Saponins/immunology , Seasons , Th1 Cells/immunology , Th2 Cells/immunology , Tragacanth/immunology , Vaccination/methods
7.
Hum Vaccin Immunother ; 14(3): 518-522, 2018 03 04.
Article in English | MEDLINE | ID: mdl-28857677

ABSTRACT

Influenza A/H3N2 viruses are the most common and virulent subtypes for humans. Antigenic drift, changes in antigenicity through the accumulation of mutations in the hemagglutinin (HA) gene is chiefly responsible for the continuing circulation of A/H3N2 viruses, resulting in frequent updates of vaccine strains based on new variant analyses. In humans, these drift-related mutations are considered to be primarily caused by the immune pressure elicited by natural infection. Whether or not the immune pressure elicited by vaccination (vaccine pressure) can have a certain effect on drift-related mutations is unclear. Recently, our findings suggested the possible effect of vaccine pressure on HA mutations by directly comparing amino acid differences from the corresponding vaccine strains between isolates from vaccinated and unvaccinated patients. It is possible that influenza vaccine pressure selects variants genetically distant from the vaccine strains. Considering the effect of vaccine pressure on HA mutations would contribute to further understanding the mechanism of antigenic drift, which would be helpful for predicting future epidemic viruses.


Subject(s)
Antigenic Variation/immunology , Influenza Vaccines/immunology , Influenza, Human/epidemiology , Influenza, Human/immunology , Amino Acids/immunology , Antibodies, Viral/immunology , Antigens, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/prevention & control , Seasons , Vaccination/methods
8.
PLoS One ; 12(12): e0189511, 2017.
Article in English | MEDLINE | ID: mdl-29252990

ABSTRACT

Influenza virus evolves rapidly due to the accumulated genetic variations on the viral sequence. Unlike in North America and Europe, influenza season in the tropical Southeast Asia spans both the rainy and cool seasons. Thus, influenza epidemiology and viral evolution sometimes differ from other regions, which affect the ever-changing efficacy of the vaccine. To monitor the current circulating influenza viruses in this region, we determined the predominant influenza virus strains circulating in Thailand between January 2016 and June 2017 by screening 7,228 samples from patients with influenza-like illness. During this time, influenza A(H3N2) virus was the predominant influenza virus detected. We then phylogenetically compared the hemagglutinin (HA) gene from a subset of these A(H3N2) strains (n = 62) to the reference sequences and evaluated amino acid changes in the dominant antigenic epitopes on the HA protein structure. The divergence of the circulating A(H3N2) from the A/Hong Kong/4801/2014 vaccine strain formed five genetic groups (designated I to V) within the 3C.2a clade. Our results suggest a marked drift of the current circulating A(H3N2) strains in Thailand, which collectively contributed to the declining predicted vaccine effectiveness (VE) from 74% in 2016 down to 48% in 2017.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/epidemiology , Influenza, Human/immunology , Amino Acids/chemistry , Epitopes/immunology , Genetic Variation , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Phylogeny , RNA, Viral/genetics , Seasons , Thailand/epidemiology
9.
Sci Rep ; 7(1): 17360, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29234060

ABSTRACT

Lactic acid bacteria (LAB) are the common probiotics. Here, we investigated the antiviral protective effects of heat-killed LAB strain Lactobacillus casei DK128 (DK128) on influenza viruses. Intranasal treatment of mice with DK128 conferred protection against different subtypes of influenza viruses by lessening weight loss and lowering viral loads. Protection via heat-killed DK128 was correlated with an increase in alveolar macrophage cells in the lungs and airways, early induction of virus specific antibodies, reduced levels of pro-inflammatory cytokines and innate immune cells. Importantly, the mice that were protected against primary viral infection as a result of heat-killed DK128 pretreatment developed subsequent heterosubtypic immunity against secondary virus infection. For protection against influenza virus via heat-killed DK128 pretreatment, B cells and partially CD4 T cells but not CD8 T cells were required as inferred from studies using knockout mouse models. Our study provides insight into how hosts can be equipped with innate and adaptive immunity via heat-killed DK128 treatment to protect against influenza virus, supporting that heat-killed LAB may be developed as anti-virus probiotics.


Subject(s)
Coinfection/prevention & control , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/prevention & control , Lacticaseibacillus casei/immunology , Probiotics/administration & dosage , Administration, Intranasal , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Coinfection/immunology , Coinfection/virology , Cross Protection/drug effects , Cross Protection/immunology , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Hot Temperature , Humans , Influenza A Virus, H1N1 Subtype/isolation & purification , Influenza A Virus, H3N2 Subtype/isolation & purification , Influenza, Human/immunology , Influenza, Human/mortality , Influenza, Human/virology , Mice , Mice, Knockout , Treatment Outcome , Viral Load/drug effects
10.
Sci Rep ; 7(1): 1283, 2017 04 28.
Article in English | MEDLINE | ID: mdl-28455520

ABSTRACT

Influenza vaccines of H7N9 subtype are consistently less immunogenic in humans than vaccines developed for other subtypes. Although prior immunoinformatic analysis identified T-cell epitopes in H7 hemagglutinin (HA) which potentially enhance regulatory T cell response due to conservation with the human genome, the links between the T-cell epitopes and low immunogenicity of H7 HA remains unknown due to the lack of animal models reproducing the response observed in humans. Here, we utilized a humanized mouse model to recapitulate the low immunogenicity of H7 HA. Our analysis demonstrated that modification of a single H7 epitope by changing 3 amino acids so that it is homologous with a known H3 immunogenic epitope sequence significantly improved the immunogenicity of the H7 HA in the humanized mouse model, leading to a greater than 4-fold increase in HA-binding IgG responses. Thus, we provide experimental evidence for the important contribution of this H7-specific T cell epitope in determining the immunogenicity of an influenza vaccine. Furthermore, this study delineates strategies that can be used for screening and selecting vaccine strains using immunoinformatics tools and a humanized mouse model.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Immunogenicity, Vaccine , Influenza A Virus, H7N9 Subtype/immunology , Influenza Vaccines/immunology , Influenza, Human/immunology , Amino Acids/genetics , Amino Acids/immunology , Animals , Antibodies, Viral/immunology , Disease Models, Animal , Epitopes, T-Lymphocyte/genetics , Humans , Influenza A Virus, H3N2 Subtype/immunology , Mice, Inbred BALB C , Mutation
11.
Molecules ; 22(1)2017 Jan 11.
Article in English | MEDLINE | ID: mdl-28085062

ABSTRACT

Influenza remains one of the major epidemic diseases worldwide, and rapid virus replication and collateral lung tissue damage caused by excessive pro-inflammatory host immune cell responses lead to high mortality rates. Thus, novel therapeutic agents that control influenza A virus (IAV) propagation and attenuate excessive pro-inflammatory responses are needed. Polysaccharide extract from Radix isatidis, a traditional Chinese herbal medicine, exerted potent anti-IAV activity against human seasonal influenza viruses (H1N1 and H3N2) and avian influenza viruses (H6N2 and H9N2) in vitro. The polysaccharides also significantly reduced the expression of pro-inflammatory cytokines (IL-6) and chemokines (IP-10, MIG, and CCL-5) stimulated by A/PR/8/34 (H1N1) at a range of doses (7.5 mg/mL, 15 mg/mL, and 30 mg/mL); however, they were only effective against progeny virus at a high dose. Similar activity was detected against inflammation induced by avian influenza virus H9N2. The polysaccharides strongly inhibited the protein expression of TLR-3 induced by PR8, suggesting that they impair the upregulation of pro-inflammatory factors induced by IAV by inhibiting activation of the TLR-3 signaling pathway. The polysaccharide extract from Radix isatidis root therefore has the potential to be used as an adjunct to antiviral therapy for the treatment of IAV infection.


Subject(s)
Antiviral Agents/pharmacology , Drugs, Chinese Herbal/chemistry , Influenza A Virus, H1N1 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H9N2 Subtype/drug effects , Polysaccharides/pharmacology , Toll-Like Receptor 3/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/isolation & purification , Anti-Inflammatory Agents/pharmacology , Antiviral Agents/isolation & purification , Bronchi/cytology , Bronchi/drug effects , Bronchi/immunology , Cell Line , Cell Survival/drug effects , Chemokine CCL5/genetics , Chemokine CCL5/immunology , Chemokine CXCL9/genetics , Chemokine CXCL9/immunology , Chickens , Dogs , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Gene Expression Regulation , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Madin Darby Canine Kidney Cells , Polysaccharides/isolation & purification , Signal Transduction/drug effects , Signal Transduction/immunology , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/immunology , Zygote/virology
12.
Vaccine ; 35(5): 713-715, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28065476

ABSTRACT

We previously reported a 2011/12 study in elderly showing that immunization with the universal influenza vaccine candidate, M-001, three weeks before administering trivalent influenza vaccine (TIV) enhanced seroconversion of Hemagglutination Inhibition (HAI) antibodies against known influenza vaccine strains circulating at that time. We now report that those subjects primed with M-001 prior to TIV in 2011 also showed, in their 2011 sera, significantly more HAI antibodies with improved seroprotection and seroconversion against strain A/Switzerland/9715293/2013(H3N2-like) that caused the 2014/15 influenza epidemic and that wasn't known to circulate in 2011/12. These data indicate that M-001 can provide broadened enhanced immunity extending even to influenza strains destined to circulate in future years. The fact that M-001 stimulates T cell activation and is devoid of HA hypervariable epitopes indicates that such broadened HAI responses effected by M-001 priming is due to extensive T cell priming.


Subject(s)
Antibodies, Neutralizing/biosynthesis , Antibodies, Viral/biosynthesis , Immunization/methods , Influenza A Virus, H3N2 Subtype/drug effects , Influenza Vaccines/administration & dosage , Influenza, Human/prevention & control , Adjuvants, Immunologic/administration & dosage , Aged , Aged, 80 and over , Aluminum Compounds/administration & dosage , Cross Protection , Female , Hemagglutination Inhibition Tests , Humans , Immunization Schedule , Immunogenicity, Vaccine , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/virology , Male , Phosphates/administration & dosage , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/virology , Vaccines, Subunit
13.
Hum Vaccin Immunother ; 12(12): 3043-3055, 2016 12.
Article in English | MEDLINE | ID: mdl-27690762

ABSTRACT

In this study we describe the immunogenicity results from a subset of older people (N = 5187) who participated in a Phase 3 randomized, observer-blinded trial of AS03-TIV versus TIV (Fluarix™) (ClinicalTrials.gov, NCT00753272). Participants received one dose of AS03-TIV or TIV in each study year and antibody titers against the vaccine strains were assessed using hemagglutination-inhibition (HI) assay at 21 d and 180 d post-vaccination in each vaccine group in the 2008/09 (Year 1) and 2009/10 (Year 2) influenza seasons. Manufacturing consistency of 3 lots of AS03-TIV for HI antibody responses in Year 1 was a co-primary objective. In a post-hoc analysis, a statistical regression model included 4830 subjects in whom immunogenicity and laboratory-confirmed attack rate data were available; the analysis was performed to assess HI antibody titers against A/H3N2 as a correlate of protection for laboratory-confirmed A/H3N2 influenza. AS03-TIV and TIV elicited strong HI antibody responses against each vaccine strain 21 d post-vaccination in both years. The manufacturing consistency of 3 lots of AS03-TIV was demonstrated. In both years and each vaccine group, HI antibody responses were lower for A/H1N1 than the other vaccine strains. Day 180 seroconversion rates (proportion with ≥4-fold increase in titer compared with pre-vaccination titer) in Year 1 in the AS03-TIV and TIV groups, respectively, were 87.7% and 74.1% for A/H3N2, 69.7% and 59.6% for influenza B, and 58.3% and 47.4% for A/H1N1. The post-hoc statistical model based on A/H3N2 attack rates and HI antibody titers estimated that a 4-fold increase in post-vaccination titers against A/H3N2 was associated with a 2-fold decrease in the odds of A/H3N2 infection.


Subject(s)
Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Polysorbates/administration & dosage , Squalene/administration & dosage , alpha-Tocopherol/administration & dosage , Aged , Aged, 80 and over , Antibodies, Viral/blood , Drug Combinations , Female , Hemagglutination Inhibition Tests , Humans , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/virology , Male , Single-Blind Method , Treatment Outcome , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/immunology
14.
J Control Release ; 232: 75-82, 2016 06 28.
Article in English | MEDLINE | ID: mdl-27094605

ABSTRACT

The delivery of vaccines to the sublingual mucosa is an attractive prospect due to the ease and acceptability of such an approach. However, novel adjuvant and delivery approaches are required to optimally vaccinate at this site. We have previously shown that conjugation of protein antigen to the iron transport molecule, transferrin, can significantly enhance mucosal immune responses. We tested whether conjugating influenza haemagglutinin to transferrin could improve the immune response to sublingually delivered antigen. Transferrin conjugated haemagglutinin induced a significant antibody and T cell response in both naïve animals and previously immunized animals. The immune response generated was able to protect mice against influenza virus challenge. Sublingually administered antigen dispersed more widely through the gastro-intestinal tract than intranasally delivered antigen and transferrin conjugation had a more marked effect on sublingually delivered antigen than intranasal immunisation. From these studies we conclude that transferrin conjugation of antigen is effective at boosting immune responses to sublingually delivered antigen and may be an attractive approach for influenza vaccines, particularly when mass campaigns are required.


Subject(s)
Antigens, CD/administration & dosage , Hemagglutinins, Viral/administration & dosage , Influenza A Virus, H3N2 Subtype , Influenza Vaccines/administration & dosage , Orthomyxoviridae Infections/prevention & control , Receptors, Transferrin/administration & dosage , Administration, Intranasal , Administration, Sublingual , Animals , Antibodies, Viral/immunology , Antigens, CD/chemistry , Female , Hemagglutinins, Viral/chemistry , Immunoglobulin G/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/chemistry , Lung/virology , Mice, Inbred BALB C , Orthomyxoviridae Infections/immunology , Receptors, Transferrin/chemistry , T-Lymphocytes/immunology
15.
Kidney Blood Press Res ; 41(2): 139-47, 2016.
Article in English | MEDLINE | ID: mdl-26914585

ABSTRACT

BACKGROUND/AIMS: The immune response to influenza vaccine may be influenced by many factors, e.g. age, comorbidities or inflammation, and iron status. METHODS: We studied the vaccine-induced production of hemagglutination-inhibition antibodies (HI) in 133 hemodialysis patients (HD) and 40 controls. To identify variables associated with the immune response, uni- and multivariate regression analyses were performed with seroconversion in HI titers as a dependent variable, with demographics, comorbidities, previous vaccination, inflammation, and iron status as independent variables. RESULTS: Seroconversion rates were lower in HD than in controls [43% versus 73% (H1N1 strain; p < 0.05); 43% versus 53% (H3N2; P=NS); 36% versus 62% (B; p < 0.05)]. In both HD and control groups, the predictors of the inferior HI production were pre-vaccination seroprotection, vaccination in the previous season, and old age. We did not find associations between seroconversion rates and inflammation and iron status in the studied populations. This was also true for a subanalysis of patients without pre-vaccination seroprotection. CONCLUSION: The influenza vaccine-induced antibody production was lower in HD than in controls and was independent of inflammation and iron status in both groups. Besides dependence on dialysis, the variables associated with inferior seroconversion rates included pre-vaccination seroprotection, previous vaccination, and old age.


Subject(s)
Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Iron/blood , Renal Dialysis/trends , Vaccination/trends , Age Factors , Aged , Female , Humans , Immunity, Cellular/drug effects , Immunity, Cellular/immunology , Inflammation/blood , Inflammation/diagnosis , Inflammation/immunology , Influenza A Virus, H1N1 Subtype/metabolism , Influenza A Virus, H3N2 Subtype/metabolism , Male , Middle Aged , Predictive Value of Tests
16.
Hum Vaccin Immunother ; 12(4): 1003-8, 2016 04 02.
Article in English | MEDLINE | ID: mdl-26751915

ABSTRACT

INTRODUCTION: An effective immune response to vaccination may be related to nutritional status. This study examined the association of plasma mineral levels with hemagglutination inhibition (HI) titers produced in response to influenza vaccine in older adults. METHODS: Prior to (Day 0) and 21 (range = 19-28) days after receiving the 2013-14 influenza vaccine, 109 adults ages 51-81 years, provided blood samples. Serum samples were tested for HI activity against the A/H1N1 and A/H3N2 2013-2014 vaccine virus strains. Plasma minerals were collected in zinc-free tubes and assayed by inductively coupled plasma mass spectrometry. HI titers were reported as seroprotection (≥1:40) and seroconversion (≥ 4-fold rise from Day 0 (minimum HI = 1:10) to Day 21). Both HI titers and mineral values were skewed and thus log2 transformed. Magnesium (Mg), phosphorus (P), zinc (Zn), copper (Cu), iron (Fe), potassium (K) and the Cu to Zn ratio were tested. Logistic regression analyses were used to determine the associations between mineral levels and seroconversion and seroprotection of HI titers for each influenza A strain. RESULTS: Participants were 61% white, 28% male, 39% diabetic, and 81% overweight/obese with a mean age of 62.6 y. In logistic regression, Day 21 A/H1N1 seroprotection was associated with P and Zn at Day 21(P < 0.05). Seroconversion of A/H1N1 was associated with Day 21 Cu, P, and Mg (P < 0.03). Day 21 A/H3N2 seroprotection and seroconversion were associated with Day 21 P (P < 0.05). CONCLUSIONS: Phosphorus was associated with seroprotection and seroconversion to influenza A after vaccination; these associations warrant additional studies with larger, more diverse population groups.


Subject(s)
Antibodies, Viral/blood , Influenza Vaccines/immunology , Minerals/blood , Seroconversion , Age Factors , Aged , Aged, 80 and over , Antibodies, Viral/immunology , Female , Hemagglutination Inhibition Tests , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza, Human/prevention & control , Logistic Models , Male , Mass Spectrometry , Middle Aged , Phosphorus/blood
17.
J Control Release ; 225: 192-204, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26774221

ABSTRACT

Dissolvable microneedle (DMN) patches for immunization have multiple benefits, including vaccine stability and ease-of-use. However, conventional DMN fabrication methods have several drawbacks. Here we describe a novel, microfluidic, drop dispensing-based dissolvable microneedle production method that overcomes these issues. Uniquely, heterogeneous arrays, consisting of microneedles of diverse composition, can be easily produced on the same patch. Robustness of the process was demonstrated by incorporating and stabilizing adenovirus and MVA vaccines. Clinically-available trivalent inactivated influenza vaccine (TIV) in DMN patches is fully stable for greater than 6months at 40°C. Immunization using low dose TIV-loaded DMN patches induced significantly higher antibody responses compared to intramuscular-based immunization in mice. TIV-loaded patches also induced a broader, heterosubtypic neutralizing antibody response. By addressing issues that will be faced in large-scale fill-finish DMN fabrication processes and demonstrating superior thermostable characteristics and immunogenicity, this study progresses the translation of this microneedle platform to eventual clinical deployment.


Subject(s)
Drug Delivery Systems , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/administration & dosage , Needles , Adenoviridae , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Dimethylpolysiloxanes , Drug Stability , Female , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunoglobulin G/blood , Mice, Inbred BALB C , Microinjections , Silicon , Solubility , Vaccination/instrumentation , Vaccination/methods , Vaccines, Inactivated , Vaccinia virus
18.
Vet Microbiol ; 179(3-4): 250-63, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26210951

ABSTRACT

The development of a universal influenza vaccine that provides broad cross protection against existing and unforeseen influenza viruses is a critical challenge. In this study, we constructed and expressed conserved sM2 and HA2 influenza antigens with cholera toxin subunit A1 (CTA1) on the surface of Lactobacillus casei (pgsA-CTA1sM2HA2/L. casei). Oral and nasal administrations of recombinant L. casei into mice resulted in high levels of serum immunoglobulin G (IgG) and their isotypes (IgG1 & IgG2a) as well as mucosal IgA. The mucosal administration of pgsA-CTA1sM2HA2/L. casei may also significantly increase the levels of sM2- or HA2-specific cell-mediated immunity because increased release of both IFN-γ and IL-4 was observed. The recombinant pgsA-CTA1sM2HA2/L. casei provided better protection of BALB/c mice against 10 times the 50% mouse lethal doses (MLD50) of homologous A/EM/Korea/W149/06(H5N1) or A/Aquatic bird/Korea/W81/2005 (H5N2) and heterologous A/Puerto Rico/8/34(H1N1), or A/Chicken/Korea/116/2004(H9N2) or A/Philippines/2/08(H3N2) viruses, compared with L. casei harboring sM2HA2 and also the protection was maintained up to seven months after administration. These results indicate that recombinant L. casei expressing the highly conserved sM2, HA2 of influenza and CTA1 as a mucosal adjuvant could be a potential mucosal vaccine candidate or tool to protect against divergent influenza viruses for human and animal.


Subject(s)
Cross Protection/immunology , Immunity, Cellular/immunology , Influenza A virus/genetics , Influenza A virus/immunology , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Adjuvants, Immunologic , Administration, Intranasal , Animals , Antigens, Surface/immunology , Cholera Toxin/immunology , Drug Evaluation, Preclinical , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N2 Subtype/immunology , Influenza A Virus, H9N2 Subtype/immunology , Interleukin-4/immunology , Lactobacillus/immunology , Lactobacillus/metabolism , Mice , Mice, Inbred BALB C , Republic of Korea
19.
PLoS One ; 9(6): e98671, 2014.
Article in English | MEDLINE | ID: mdl-24910991

ABSTRACT

BACKGROUND: Smokers have increased susceptibility and altered innate host defense responses to influenza virus infection. Broccoli sprouts are a source of the Nrf2 activating agentsulforaphane, and short term ingestion of broccoli sprout homogenates (BSH) has been shown to reduce nasal inflammatory responses to oxidant pollutants. OBJECTIVES: Assess the effects of BSH on nasal cytokines, virus replication, and Nrf2-dependent enzyme expression in smokers and nonsmokers. METHODS: We conducted a randomized, double-blind, placebo-controlled trial comparing the effects of BSH on serially sampled nasal lavage fluid (NLF) cytokines, viral sequence quantity, and Nrf2-dependent enzyme expression in NLF cells and biopsied epithelium. Healthy young adult smokers and nonsmokers ingested BSH or placebo (alfalfa sprout homogenate) for 4 days, designated Days -1, 0, 1, 2. On Day 0 they received standard vaccine dose of live attenuated influenza virus (LAIV) intranasally. Nasal lavage fluids and nasal biopsies were collected serially to assess response to LAIV. RESULTS: In area under curve analyses, post-LAIV IL-6 responses (P = 0.03) and influenza sequences (P = 0.01) were significantly reduced in NLF from BSH-treated smokers, while NAD(P)H: quinoneoxidoreductasein NLF cells was significantly increased. In nonsmokers, a similar trend for reduction in virus quantity with BSH did not reach statistical significance. CONCLUSIONS: In smokers, short term ingestion of broccoli sprout homogenates appears to significantly reduce some virus-induced markers of inflammation, as well as reducing virus quantity. Nutritional antioxidant interventions have promise as a safe, low-cost strategy for reducing influenza risk among smokers and other at risk populations. TRIAL REGISTRATION: ClinicalTrials.gov NCT01269723.


Subject(s)
Brassica/chemistry , Influenza Vaccines/immunology , Nose/drug effects , Orthomyxoviridae/immunology , Plant Extracts/pharmacology , Plant Shoots/chemistry , Smoking , Adult , Double-Blind Method , Female , Gene Expression Regulation/drug effects , Heme Oxygenase-1/genetics , Humans , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H3N2 Subtype/immunology , Male , NAD(P)H Dehydrogenase (Quinone)/genetics , Nasal Lavage Fluid/immunology , Nose/immunology , Nose/pathology , Vaccines, Attenuated/immunology
20.
J Pharm Sci ; 103(3): 821-7, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24425059

ABSTRACT

The recombinant hemagglutinin (rHA)-based influenza vaccine Flublok® has recently been approved in the United States as an alternative to the traditional egg-derived flu vaccines. Flublok is a purified vaccine with a hemagglutinin content that is threefold higher than standard inactivated influenza vaccines. When rHA derived from an H3N2 influenza virus was expressed, purified, and stored for 1 month, a rapid loss of in vitro potency (∼50%) was observed as measured by the single radial immunodiffusion (SRID) assay. A comprehensive characterization of the rHA protein antigen was pursued to identify the potential causes and mechanisms of this potency loss. In addition, the biophysical and chemical stability of the rHA in different formulations and storage conditions was evaluated over time. Results demonstrate that the potency loss over time did not correlate with trends in changes to the higher order structure or hydrodynamic size of the rHA. The most likely mechanism for the early loss of potency was disulfide-mediated cross-linking of rHA, as the formation of non-native disulfide-linked multimers over time correlated well with the observed potency loss. Furthermore, a loss of free thiol content, particularly in specific cysteine residues in the antigen's C-terminus, was correlated with potency loss measured by SRID.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza A Virus, H3N2 Subtype/metabolism , Influenza Vaccines/chemistry , Chemical Phenomena , Cysteine/analysis , Cysteine/chemistry , Cystine/analysis , Cystine/chemistry , Drug Stability , Drug Storage , Excipients/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/pharmacology , Hydrodynamics , Immunodiffusion , Influenza A Virus, H3N2 Subtype/drug effects , Influenza A Virus, H3N2 Subtype/growth & development , Influenza A Virus, H3N2 Subtype/immunology , Influenza Vaccines/genetics , Influenza Vaccines/metabolism , Influenza Vaccines/pharmacology , Octoxynol/chemistry , Oxidation-Reduction , Peptide Mapping , Protein Stability , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Spectroscopy, Fourier Transform Infrared , Temperature , Thioglycolates/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL