Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 383
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Clin Infect Dis ; 78(6): 1757-1768, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38537255

RESUMO

INTRODUCTION: A surge of human influenza A(H7N9) cases began in 2016 in China from an antigenically distinct lineage. Data are needed about the safety and immunogenicity of 2013 and 2017 A(H7N9) inactivated influenza vaccines (IIVs) and the effects of AS03 adjuvant, prime-boost interval, and priming effects of 2013 and 2017 A(H7N9) IIVs. METHODS: Healthy adults (n = 180), ages 19-50 years, were enrolled into this partially blinded, randomized, multicenter phase 2 clinical trial. Participants were randomly assigned to 1 of 6 vaccination groups evaluating homologous versus heterologous prime-boost strategies with 2 different boost intervals (21 vs 120 days) and 2 dosages (3.75 or 15 µg of hemagglutinin) administered with or without AS03 adjuvant. Reactogenicity, safety, and immunogenicity measured by hemagglutination inhibition and neutralizing antibody titers were assessed. RESULTS: Two doses of A(H7N9) IIV were well tolerated, and no safety issues were identified. Although most participants had injection site and systemic reactogenicity, these symptoms were mostly mild to moderate in severity; injection site reactogenicity was greater in vaccination groups receiving adjuvant. Immune responses were greater after an adjuvanted second dose, and with a longer interval between prime and boost. The highest hemagglutination inhibition geometric mean titer (95% confidence interval) observed against the 2017 A(H7N9) strain was 133.4 (83.6-212.6) among participants who received homologous, adjuvanted 3.75 µg + AS03/2017 doses with delayed boost interval. CONCLUSIONS: Administering AS03 adjuvant with the second H7N9 IIV dose and extending the boost interval to 4 months resulted in higher peak antibody responses. These observations can broadly inform strategic approaches for pandemic preparedness. Clinical Trials Registration. NCT03589807.


Assuntos
Anticorpos Antivirais , Imunização Secundária , Subtipo H7N9 do Vírus da Influenza A , Vacinas contra Influenza , Influenza Humana , Vacinas de Produtos Inativados , Humanos , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/efeitos adversos , Adulto , Masculino , Feminino , Pessoa de Meia-Idade , Subtipo H7N9 do Vírus da Influenza A/imunologia , Vacinas de Produtos Inativados/imunologia , Vacinas de Produtos Inativados/administração & dosagem , Vacinas de Produtos Inativados/efeitos adversos , Anticorpos Antivirais/sangue , Influenza Humana/prevenção & controle , Influenza Humana/imunologia , Adulto Jovem , Esquemas de Imunização , Testes de Inibição da Hemaglutinação , Estados Unidos , Imunogenicidade da Vacina , Anticorpos Neutralizantes/sangue , Polissorbatos/administração & dosagem , Polissorbatos/efeitos adversos , alfa-Tocoferol/administração & dosagem , alfa-Tocoferol/efeitos adversos , Esqualeno/administração & dosagem , Esqualeno/efeitos adversos , Esqualeno/imunologia , Voluntários Saudáveis , Combinação de Medicamentos , Adjuvantes de Vacinas/administração & dosagem , Vacinação/métodos , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/efeitos adversos
2.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33941704

RESUMO

Intranasal (i.n.) immunization is a promising vaccination route for infectious respiratory diseases such as influenza. Recombinant protein vaccines can overcome the safety concerns and long production phase of virus-based influenza vaccines. However, soluble protein vaccines are poorly immunogenic if administered by an i.n. route. Here, we report that polyethyleneimine-functionalized graphene oxide nanoparticles (GP nanoparticles) showed high antigen-loading capacities and superior immunoenhancing properties. Via a facile electrostatic adsorption approach, influenza hemagglutinin (HA) was incorporated into GP nanoparticles and maintained structural integrity and antigenicity. The resulting GP nanoparticles enhanced antigen internalization and promoted inflammatory cytokine production and JAWS II dendritic cell maturation. Compared with soluble HA, GP nanoparticle formulations induced significantly enhanced and cross-reactive immune responses at both systemic sites and mucosal surfaces in mice after i.n. immunization. In the absence of any additional adjuvant, the GP nanoparticle significantly boosted antigen-specific humoral and cellular immune responses, comparable to the acknowledged potent mucosal immunomodulator CpG. The robust immune responses conferred immune protection against challenges by homologous and heterologous viruses. Additionally, the solid self-adjuvant effect of GP nanoparticles may mask the role of CpG when coincorporated. In the absence of currently approved mucosal adjuvants, GP nanoparticles can be developed into potent i.n. influenza vaccines, providing broad protection. With versatility and flexibility, the GP nanoplatform can be easily adapted for constructing mucosal vaccines for different respiratory pathogens.


Assuntos
Reações Cruzadas/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Nanopartículas/química , Infecções por Orthomyxoviridae/imunologia , Administração Intranasal , Animais , Linhagem Celular , Citocinas/imunologia , Citocinas/metabolismo , Feminino , Grafite/química , Grafite/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Humanos , Imunidade Humoral/efeitos dos fármacos , Imunidade Humoral/imunologia , Imunidade nas Mucosas/efeitos dos fármacos , Imunidade nas Mucosas/imunologia , Vírus da Influenza A Subtipo H3N2/efeitos dos fármacos , Vírus da Influenza A Subtipo H3N2/fisiologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/química , Influenza Humana/prevenção & controle , Influenza Humana/virologia , Camundongos Endogâmicos BALB C , Nanopartículas/administração & dosagem , Oligodesoxirribonucleotídeos/química , Oligodesoxirribonucleotídeos/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Polietilenoimina/química , Vacinação/métodos
3.
J Virol ; 96(19): e0100622, 2022 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-36106872

RESUMO

Intranasal vaccination offers the potential advantage of needle-free prevention of respiratory pathogens such as influenza viruses with induction of mucosal immune responses. Optimal design of adjuvants and antigen delivery vehicles for intranasal delivery has not yet been well established. Here, we report that an adjuvant-containing nanoliposome antigen display system that converts soluble influenza hemagglutinin antigens into nanoparticles is effective for intranasal immunization. Intranasal delivery of nanoliposomes in mice delivers the particles to resident immune cells in the respiratory tract, inducing a mucosal response in the respiratory system as evidenced by nasal and lung localized IgA antibody production, while also producing systemic IgG antibodies. Intranasal vaccination with nanoliposome particles decorated with nanogram doses of hemagglutinin protected mice from homologous and heterologous H3N2 and H1N1 influenza virus challenge. IMPORTANCE A self-assembling influenza virus vaccine platform that seamlessly converts soluble antigens into nanoparticles is demonstrated with various H1N1 and H3N2 influenza antigens to protect mice against influenza virus challenge following intranasal vaccination. Mucosal immune responses following liposome delivery to lung antigen-presenting cells are demonstrated.


Assuntos
Glicoproteínas de Hemaglutininação de Vírus da Influenza , Imunidade nas Mucosas , Vacinas contra Influenza , Infecções por Orthomyxoviridae , Adjuvantes Imunológicos , Administração Intranasal , Animais , Anticorpos Antivirais/imunologia , Células Apresentadoras de Antígenos/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/administração & dosagem , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Vírus da Influenza A Subtipo H1N1 , Vírus da Influenza A Subtipo H3N2 , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Lipossomos , Camundongos , Nanopartículas , Infecções por Orthomyxoviridae/prevenção & controle , Vacinação
4.
Virol J ; 20(1): 187, 2023 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-37605141

RESUMO

BACKGROUND: Influenza A virus (IAV) causes respiratory disease in pigs and is a major concern for public health. Vaccination of pigs is the most successful measure to mitigate the impact of the disease in the herds. Influenza-based virosome is an effective immunomodulating carrier that replicates the natural antigen presentation pathway and has tolerability profile due to their purity and biocompatibility. METHODS: This study aimed to develop a polyvalent virosome influenza vaccine containing the hemagglutinin and neuraminidase proteins derived from the swine IAVs (swIAVs) H1N1, H1N2 and H3N2 subtypes, and to investigate its effectiveness in mice as a potential vaccine for swine. Mice were immunized with two vaccine doses (1 and 15 days), intramuscularly and intranasally. At 21 days and eight months later after the second vaccine dose, mice were euthanized. The humoral and cellular immune responses in mice vaccinated intranasally or intramuscularly with a polyvalent influenza virosomal vaccine were investigated. RESULTS: Only intramuscular vaccination induced high hemagglutination inhibition (HI) titers. Seroconversion and seroprotection (> 4-fold rise in HI antibody titers, reaching a titer of ≥ 1:40) were achieved in 80% of mice (intramuscularly vaccinated group) at 21 days after booster immunization. Virus-neutralizing antibody titers against IAV were detected at 8 months after vaccination, indicating long-lasting immunity. Overall, mice immunized with the virosome displayed greater ability for B, effector-T and memory-T cells from the spleen to respond to H1N1, H1N2 and H3N2 antigens. CONCLUSIONS: All findings showed an efficient immune response against IAVs in mice vaccinated with a polyvalent virosome-based influenza vaccine.


Assuntos
Vacinas contra Influenza , Influenza Humana , Vacinas Virossomais , Lavagem Broncoalveolar , Vírus da Influenza A Subtipo H1N1 , Vírus da Influenza A Subtipo H1N2 , Vírus da Influenza A Subtipo H3N2 , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Influenza Humana/imunologia , Baço/citologia , Baço/imunologia , Vacinas Combinadas/administração & dosagem , Vacinas Virossomais/administração & dosagem , Vacinas Virossomais/imunologia , Virossomos/ultraestrutura , Humanos , Animais , Camundongos
5.
Int J Mol Sci ; 23(3)2022 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-35163772

RESUMO

The SARS-CoV-2 pandemic caused a massive health and societal crisis, although the fast development of effective vaccines reduced some of the impact. To prepare for future respiratory virus pandemics, a pan-viral prophylaxis could be used to control the initial virus outbreak in the period prior to vaccine approval. The liposomal vaccine adjuvant CAF®09b contains the TLR3 agonist polyinosinic:polycytidylic acid, which induces a type I interferon (IFN-I) response and an antiviral state in the affected tissues. When testing CAF09b liposomes as a potential pan-viral prophylaxis, we observed that intranasal administration of CAF09b liposomes to mice resulted in an influx of innate immune cells into the nose and lungs and upregulation of IFN-I-related gene expression. When CAF09b liposomes were administered prior to challenge with mouse-adapted influenza A/Puerto Rico/8/1934 virus, it protected from severe disease, although the virus was still detectable in the lungs. However, when CAF09b liposomes were administered after influenza challenge, the mice had a similar disease course to controls. In conclusion, CAF09b may be a suitable candidate as a pan-viral prophylactic treatment for epidemic viruses, but must be administered prior to virus exposure to be effective.


Assuntos
Adjuvantes de Vacinas/uso terapêutico , Vacinas contra Influenza/uso terapêutico , Influenza Humana/prevenção & controle , Infecções por Orthomyxoviridae/prevenção & controle , Desenvolvimento de Vacinas/métodos , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/uso terapêutico , Adjuvantes de Vacinas/administração & dosagem , Adjuvantes de Vacinas/química , Adjuvantes de Vacinas/farmacologia , Administração Intranasal , Animais , COVID-19/prevenção & controle , Vacinas contra COVID-19/síntese química , Vacinas contra COVID-19/uso terapêutico , Células Cultivadas , Embrião de Galinha , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/química , Vacinas contra Influenza/farmacologia , Interferon Tipo I/genética , Lipossomos/química , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Prevenção Primária/métodos , SARS-CoV-2/imunologia
6.
FASEB J ; 34(9): 12406-12418, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-33411367

RESUMO

The clinically successful adjuvant MF59 is used in seasonal influenza vaccines, which is proposed to enhance immunity by creating an immune-competent microenvironment in the muscle that allows recruitment of immune cells that drive adaptive immune responses. Here, we examined whether the clinically successful adjuvants MF59/AddaVax could be used for subcutaneous use and how antigen delivery can be synergized with cellular dynamics at the vaccination site. Subcutaneous injection of AddaVax leads to thickening of the skin, characterized by a neutrophil-monocyte recruitment sequence. Skin-infiltrating CCR2+Ly6Chigh monocytes showed differentiation to CD11b+Ly6C+MHCII+CD11c+CD64+ monocyte-derived DCs over time in the hypodermal layers of the skin, expressing high levels of CD209a/mDC-SIGN. Surprisingly, skin thickening was accompanied with increased white adipose tissue highly enriched with monocytes. Analysis of the skin-draining lymph nodes revealed early increases in neutrophils and moDCs at 12 hours after injection and later increases in migratory cDC2s. Subcutaneous vaccination with AddaVax enhanced antigen-specific CD8+ and CD4+ T cell responses, while moDC targeting using antigen-coupled CD209a antibody additionally boosted humoral responses. Hence, oil-in-water emulsions provide an attractive immune modulatory adjuvants aimed at increasing cellular responses, as well as antibody responses when combined with moDC targeting.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas contra Influenza/imunologia , Polissorbatos/administração & dosagem , Pele/imunologia , Esqualeno/administração & dosagem , Animais , Células Dendríticas/imunologia , Vacinas contra Influenza/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/fisiologia , Neutrófilos/fisiologia , Linfócitos T/imunologia , Vacinação
7.
Mol Ther ; 28(7): 1569-1584, 2020 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-32359470

RESUMO

Influenza viruses are respiratory pathogens of public health concern worldwide with up to 650,000 deaths occurring each year. Seasonal influenza virus vaccines are employed to prevent disease, but with limited effectiveness. Development of a universal influenza virus vaccine with the potential to elicit long-lasting, broadly cross-reactive immune responses is necessary for reducing influenza virus prevalence. In this study, we have utilized lipid nanoparticle-encapsulated, nucleoside-modified mRNA vaccines to intradermally deliver a combination of conserved influenza virus antigens (hemagglutinin stalk, neuraminidase, matrix-2 ion channel, and nucleoprotein) and induce strong immune responses with substantial breadth and potency in a murine model. The immunity conferred by nucleoside-modified mRNA-lipid nanoparticle vaccines provided protection from challenge with pandemic H1N1 virus at 500 times the median lethal dose after administration of a single immunization, and the combination vaccine protected from morbidity at a dose of 50 ng per antigen. The broad protective potential of a single dose of combination vaccine was confirmed by challenge with a panel of group 1 influenza A viruses. These findings support the advancement of nucleoside-modified mRNA-lipid nanoparticle vaccines expressing multiple conserved antigens as universal influenza virus vaccine candidates.


Assuntos
Antígenos Virais/genética , Vírus da Influenza A Subtipo H1N1/imunologia , Nucleosídeos/química , Infecções por Orthomyxoviridae/prevenção & controle , Vacinas Sintéticas/administração & dosagem , Animais , Anticorpos Antivirais/metabolismo , Antígenos Virais/química , Modelos Animais de Doenças , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/química , Vacinas contra Influenza/imunologia , Injeções Intradérmicas , Lipossomos , Camundongos , Células NIH 3T3 , Nanopartículas , Neuraminidase/química , Neuraminidase/genética , Proteínas do Nucleocapsídeo/química , Proteínas do Nucleocapsídeo/genética , Infecções por Orthomyxoviridae/imunologia , Vacinas Sintéticas/química , Vacinas Sintéticas/imunologia , Vacinas de mRNA
8.
PLoS Med ; 17(3): e1003024, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32181756

RESUMO

BACKGROUND: The Vaxxas high-density microarray patch (HD-MAP) consists of a high density of microprojections coated with vaccine for delivery into the skin. Microarray patches (MAPs) offer the possibility of improved vaccine thermostability as well as the potential to be safer, more acceptable, easier to use, and more cost-effective for the administration of vaccines than injection by needle and syringe (N&S). Here, we report a phase I trial using the Vaxxas HD-MAP to deliver a monovalent influenza vaccine that was to the best of our knowledge the first clinical trial to evaluate the safety, tolerability, and immunogenicity of lower doses of influenza vaccine delivered by MAPs. METHODS AND FINDINGS: HD-MAPs were coated with a monovalent, split inactivated influenza virus vaccine containing A/Singapore/GP1908/2015 H1N1 haemagglutinin (HA). Between February 2018 and March 2018, 60 healthy adults (age 18-35 years) in Melbourne, Australia were enrolled into part A of the study and vaccinated with either: HD-MAPs delivering 15 µg of A/Singapore/GP1908/2015 H1N1 HA antigen (A-Sing) to the volar forearm (FA); uncoated HD-MAPs; intramuscular (IM) injection of commercially available quadrivalent influenza vaccine (QIV) containing A/Singapore/GP1908/2015 H1N1 HA (15 µg/dose); or IM injection of H1N1 HA antigen (15 µg/dose). After 22 days' follow-up and assessment of the safety data, a further 150 healthy adults were enrolled and randomly assigned to 1 of 9 treatment groups. Participants (20 per group) were vaccinated with HD-MAPs delivering doses of 15, 10, 5, 2.5, or 0 µg of HA to the FA or 15 µg HA to the upper arm (UA), or IM injection of QIV. The primary objectives of the study were safety and tolerability. Secondary objectives were to assess the immunogenicity of the influenza vaccine delivered by HD-MAP. Primary and secondary objectives were assessed for up to 60 days post-vaccination. Clinical staff and participants were blind as to which HD-MAP treatment was administered and to administration of IM-QIV-15 or IM-A/Sing-15. All laboratory investigators were blind to treatment and participant allocation. Two further groups in part B (5 participants per group), not included in the main safety and immunological analysis, received HD-MAPs delivering 15 µg HA or uncoated HD-MAPs applied to the forearm. Biopsies were taken on days 1 and 4 for analysis of the cellular composition from the HD-MAP application sites. The vaccine coated onto HD-MAPs was antigenically stable when stored at 40°C for at least 12 months. HD-MAP vaccination was safe and well tolerated; any systemic or local adverse events (AEs) were mild or moderate. Observed systemic AEs were mostly headache or myalgia, and local AEs were application-site reactions, usually erythema. HD-MAP administration of 2.5 µg HA induced haemagglutination inhibition (HAI) and microneutralisation (MN) titres that were not significantly different to those induced by 15 µg HA injected IM (IM-QIV-15). HD-MAP delivery resulted in enhanced humoral responses compared with IM injection with higher HAI geometric mean titres (GMTs) at day 8 in the MAP-UA-15 (GMT 242.5, 95% CI 133.2-441.5), MAP-FA-15 (GMT 218.6, 95% CI 111.9-427.0), and MAP-FA-10 (GMT 437.1, 95% CI 254.3-751.3) groups compared with IM-QIV-15 (GMT 82.8, 95% CI 42.4-161.8), p = 0.02, p = 0.04, p < 0.001 for MAP-UA-15, MAP-FA-15, and MAP-FA-10, respectively. Higher titres were also observed at day 22 in the MAP-FA-10 (GMT 485.0, 95% CI 301.5-780.2, p = 0.001) and MAP-UA-15 (367.6, 95% CI 197.9-682.7, p = 0.02) groups compared with the IM-QIV-15 group (GMT 139.3, 95% CI 79.3-244.5). Results from a panel of exploratory immunoassays (antibody-dependent cellular cytotoxicity, CD4+ T-cell cytokine production, memory B cell (MBC) activation, and recognition of non-vaccine strains) indicated that, overall, Vaxxas HD-MAP delivery induced immune responses that were similar to, or higher than, those induced by IM injection of QIV. The small group sizes and use of a monovalent influenza vaccine were limitations of the study. CONCLUSIONS: Influenza vaccine coated onto the HD-MAP was stable stored at temperatures up to 40°C. Vaccination using the HD-MAP was safe and well tolerated and resulted in immune responses that were similar to or significantly enhanced compared with IM injection. Using the HD-MAP, a 2.5 µg dose (1/6 of the standard dose) induced HAI and MN titres similar to those induced by 15 µg HA injected IM. TRIAL REGISTRATION: Australian New Zealand Clinical Trials Registry (ANZCTR.org.au), trial ID 108 ACTRN12618000112268/U1111-1207-3550.


Assuntos
Imunogenicidade da Vacina , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/administração & dosagem , Influenza Humana/prevenção & controle , Vacinação , Administração Cutânea , Adolescente , Adulto , Anticorpos Antivirais/sangue , Austrália , Células Cultivadas , Estabilidade de Medicamentos , Feminino , Humanos , Imunoglobulina A/metabolismo , Vacinas contra Influenza/efeitos adversos , Influenza Humana/imunologia , Influenza Humana/virologia , Injeções Intramusculares , Masculino , Saliva/imunologia , Saliva/virologia , Linfócitos T/imunologia , Linfócitos T/virologia , Fatores de Tempo , Adesivo Transdérmico , Resultado do Tratamento , Vacinação/efeitos adversos , Adulto Jovem
9.
BMC Vet Res ; 16(1): 319, 2020 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-32867774

RESUMO

BACKGROUND: The global prevalence of economic primary infection of poultry by H9N2 virus, including the Lineage A, panzootic group ME1, and associated with secondary infection by Mycoplasma gallisepticum (MG), is alarming to the sustainability of the poultry sector. This research evaluated in broilers the immunity and protection induced by aerosolization of liposomal nanoparticles vaccine, encapsulating antigens of H9N2 virus and MG, with or without the incorporation of Echinacea extract (EE) immuno-stimulant. Six different treatments (TRTs) of broilers were included in the experimental design, with three replicate pens/TRT and stocking of 20 day-old birds/replicate. RESULTS: The tracheobronchial washings of birds subjected to aerosolization of liposomal nanoparticles, encapsulating antigens of H9N2 and MG and EE had the highest significant mean levels of each of IgA and IgG specific to H9N2 and MG, associated with lowest tracheal MG colonization, tracheal H9N2 recovery, tracheal histopathologic lesions, mortality, and best performance in body weight and feed conversion compared to all other challenged birds allocated to different treatments (P < 0.05). However, the control broilers, free from challenge with MG and H9N2, had the lowest mortality and tracheal lesions, and the highest production performance. CONCLUSION: The aerosolization of liposomal nanoparticles, encapsulating antigens of H9N2 and MG and EE resulted in enough local immunity for protection of broilers against infection, and in attaining the highest production performance in challenged birds. The potential implication of vaccinating with safe killed nanoparticle vaccines is of utmost importance to the global poultry sector.


Assuntos
Vacinas Bacterianas/imunologia , Galinhas , Vírus da Influenza A Subtipo H9N2/imunologia , Vacinas contra Influenza/imunologia , Mycoplasma gallisepticum/imunologia , Nanopartículas/administração & dosagem , Aerossóis , Animais , Antígenos Virais , Vacinas Bacterianas/administração & dosagem , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Vacinas contra Influenza/administração & dosagem , Influenza Aviária/prevenção & controle , Lipossomos , Infecções por Mycoplasma/prevenção & controle , Infecções por Mycoplasma/veterinária
10.
Proc Natl Acad Sci U S A ; 113(7): 1853-8, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26755593

RESUMO

The dynamics and molecular mechanisms underlying vaccine immunity in early childhood remain poorly understood. Here we applied systems approaches to investigate the innate and adaptive responses to trivalent inactivated influenza vaccine (TIV) and MF59-adjuvanted TIV (ATIV) in 90 14- to 24-mo-old healthy children. MF59 enhanced the magnitude and kinetics of serum antibody titers following vaccination, and induced a greater frequency of vaccine specific, multicytokine-producing CD4(+) T cells. Compared with transcriptional responses to TIV vaccination previously reported in adults, responses to TIV in infants were markedly attenuated, limited to genes regulating antiviral and antigen presentation pathways, and observed only in a subset of vaccinees. In contrast, transcriptional responses to ATIV boost were more homogenous and robust. Interestingly, a day 1 gene signature characteristic of the innate response (antiviral IFN genes, dendritic cell, and monocyte responses) correlated with hemagglutination at day 28. These findings demonstrate that MF59 enhances the magnitude, kinetics, and consistency of the innate and adaptive response to vaccination with the seasonal influenza vaccine during early childhood, and identify potential molecular correlates of antibody responses.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Vacinas contra Influenza/administração & dosagem , Polissorbatos/administração & dosagem , Esqualeno/administração & dosagem , Biologia de Sistemas , Anticorpos Antivirais/biossíntese , Linfócitos B/imunologia , Humanos , Memória Imunológica , Lactente , Vacinas contra Influenza/imunologia , Transcriptoma
11.
J Infect Dis ; 218(6): 856-867, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-29701840

RESUMO

Background: Influenza A virus (IAV) vaccines offer little protection from mismatched viruses with antigenically distant hemagglutinin (HA) glycoproteins. We sought to determine if a cationic lipid/DNA complex (CLDC) adjuvant could induce heterosubtypic protection if added to a whole inactivated IAV vaccine (WIV). Methods: Adult rhesus macaques (RMs) were vaccinated and at 2 weeks boosted with either an H1N1-WIV or an H3N2-WIV, with and without CLDC adjuvant. Four weeks postboost, animals were challenged with an H1N1 IAV matched to the H1N1-WIV vaccine. Results: After challenge, viral RNA (vRNA) levels in the trachea of control RMs and RMs vaccinated with the unadjuvanted H1 or H3 WIV vaccines were similar. However, vRNA levels in the trachea of both the H1-WIV/CLDC- and the H3-WIV/CLDC-vaccinated RMs (P < 0.01 and P < 0.05, respectively) were significantly lower than in unvaccinated control RMs. Heterosubtypic protection in H3-WIV/CLDC RMs was associated with significantly higher levels of nucleoprotein (NP) and matrix-1-specific immunoglobulin G antibodies (P < 0.05) and NP-specific nonneutralizing antibody-dependent natural killer cell activation (P < 0.01) compared with unprotected H3-WIV RMs. Conclusions: Addition of the CLDC adjuvant to a simple WIV elicited immunity to conserved virus structural proteins in RMs that correlate with protection from uncontrolled virus replication after heterosubtypic influenza virus challenge.


Assuntos
DNA/administração & dosagem , Vírus da Influenza A Subtipo H1N1/fisiologia , Vírus da Influenza A Subtipo H3N2/fisiologia , Vacinas contra Influenza/administração & dosagem , Lipídeos/administração & dosagem , Infecções por Orthomyxoviridae/prevenção & controle , Vacinas Atenuadas/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Animais , Modelos Animais de Doenças , Feminino , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H3N2/imunologia , Vacinas contra Influenza/farmacologia , Lipossomos/administração & dosagem , Macaca mulatta/imunologia , Macaca mulatta/virologia , Masculino , Proteínas do Nucleocapsídeo , Infecções por Orthomyxoviridae/imunologia , Plasmídeos/genética , Proteínas de Ligação a RNA/imunologia , Traqueia/virologia , Vacinas Atenuadas/farmacologia , Proteínas do Core Viral/imunologia , Replicação Viral/efeitos dos fármacos
12.
PLoS Pathog ; 12(12): e1006064, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27997610

RESUMO

CD8+ cytotoxic T lymphocytes (CTLs) are critical for clearing many viral infections, and protective CTL memory can be induced by vaccination with attenuated viruses and vectors. Non-replicating vaccines are typically potentiated by the addition of adjuvants that enhance humoral responses, however few are capable of generating CTL responses. Adjuplex is a carbomer-lecithin-based adjuvant demonstrated to elicit robust humoral immunity to non-replicating antigens. We report that mice immunized with non-replicating Adjuplex-adjuvanted vaccines generated robust antigen-specific CTL responses. Vaccination by the subcutaneous or the intranasal route stimulated systemic and mucosal CTL memory respectively. However, only CTL memory induced by intranasal vaccination was protective against influenza viral challenge, and correlated with an enhancement of memory CTLs in the airways and CD103+ CD69+ CXCR3+ resident memory-like CTLs in the lungs. Mechanistically, Myd88-deficient mice mounted primary CTL responses to Adjuplex vaccines that were similar in magnitude to wild-type mice, but exhibited altered differentiation of effector cell subsets. Immune potentiating effects of Adjuplex entailed alterations in the frequency of antigen-presenting-cell subsets in vaccine draining lymph nodes, and in the lungs and airways following intranasal vaccination. Further, Adjuplex enhanced the ability of dendritic cells to promote antigen-induced proliferation of naïve CD8 T cells by modulating antigen uptake, its intracellular localization, and rate of processing. Taken together, we have identified an adjuvant that elicits both systemic and mucosal CTL memory to non-replicating antigens, and engenders protective CTL-based heterosubtypic immunity to influenza A virus in the respiratory tract. Further, findings presented in this manuscript have provided key insights into the mechanisms and factors that govern the induction and programming of systemic and protective memory CTLs in the respiratory tract.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Linfócitos T CD8-Positivos/imunologia , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/imunologia , Linfócitos T Citotóxicos/imunologia , Resinas Acrílicas/administração & dosagem , Administração Intranasal , Transferência Adotiva , Animais , Modelos Animais de Doenças , Citometria de Fluxo , Vírus da Influenza A/imunologia , Vacinas contra Influenza/administração & dosagem , Lecitinas/administração & dosagem , Lecitinas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Confocal , Infecções Respiratórias/imunologia , Infecções Respiratórias/virologia
13.
Cell Immunol ; 329: 27-30, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29665972

RESUMO

Intranasal vaccination of pigs with poly lactic-co-glycolic acid and polyanhydride nanoparticles delivered inactivated influenza virus provides cross-reactive T-cell response, but not antibody response, resulting in incomplete protection and no reduction in nasal virus shedding. Expression of BAFF and Th2 transcription factor GATA-3 were downregulated in lungs of pigs vaccinated with influenza nanovaccine, but in mice it upregulated the expression of BAFF and cytokine TGFß in cervical lymph nodes. However, the intranasal iNKT cell adjuvant, α-Galctosylceramide upregulates the expression of BAFF in pig lungs. In conclusion, expression of BAFF is differentially regulated by intranasal nanovaccine and α-Galctosylceramide in pig respiratory tract.


Assuntos
Fator Ativador de Células B/efeitos dos fármacos , Vacinas contra Influenza/farmacologia , Infecções por Orthomyxoviridae/prevenção & controle , Administração Intranasal/métodos , Animais , Fator Ativador de Células B/genética , Quimioterapia Adjuvante/métodos , Regulação da Expressão Gênica , Vacinas contra Influenza/administração & dosagem , Células Matadoras Naturais/fisiologia , Camundongos , Nanopartículas/uso terapêutico , Polianidridos/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/farmacologia , Suínos/imunologia , Suínos/virologia
14.
J Virol ; 90(15): 6976-6988, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27226368

RESUMO

UNLABELLED: CD4(+) T cells play a central role in orchestrating adaptive immunity. To better understand the roles of CD4(+) T cells in the effects of adjuvants, we investigated the efficacy of a T-dependent influenza virus split vaccine with MF59 or alum in CD4 knockout (CD4KO) and wild-type (WT) mice. CD4(+) T cells were required for the induction of IgG antibody responses to the split vaccine and the effects of alum adjuvant. In contrast, MF59 was found to be highly effective in raising isotype-switched IgG antibodies to a T-dependent influenza virus split vaccine in CD4KO mice or CD4-depleted WT mice equivalent to those in intact WT mice, thus overcoming the deficiency of CD4(+) T cells in helping B cells and inducing immunity against influenza virus. Vaccination with the MF59-adjuvanted influenza virus vaccine was able to induce protective CD8(+) T cells and long-lived antibody-secreting cells in CD4KO mice. The effects of MF59 adjuvant in CD4KO mice might be associated with uric acid, inflammatory cytokines, and the recruitment of multiple immune cells at the injection site, but their cellularity and phenotypes were different from those in WT mice. These findings suggest a new paradigm of CD4-independent adjuvant mechanisms, providing the rationales to improve vaccine efficacy in infants, the elderly, immunocompromised patients, as well as healthy adults. IMPORTANCE: MF59-adjuvanted influenza vaccines were licensed for human vaccination, but the detailed mechanisms are not fully elucidated. CD4(+) T cells are required to induce antibody isotype switching and long-term memory responses. In contrast, we discovered that MF59 was highly effective in inducing isotype-switched IgG antibodies and long-term protective immune responses to a T-dependent influenza vaccine independent of CD4(+) T cells. These findings are highly significant for the following reasons: (i) MF59 can overcome a defect of CD4(+) T cells in inducing protective immunity to vaccination with a T-dependent influenza virus vaccine; (ii) a CD4-independent pathway can be an alternative mechanism for certain adjuvants such as MF59; and (iii) this study has significant implications for improving vaccine efficacies in young children, the elderly, and immunocompromised populations.


Assuntos
Anticorpos/metabolismo , Linfócitos T CD4-Positivos , Imunoglobulina G/imunologia , Isotipos de Imunoglobulinas/imunologia , Vacinas contra Influenza/administração & dosagem , Pneumonia/prevenção & controle , Polissorbatos/administração & dosagem , Esqualeno/administração & dosagem , Linfócitos T/imunologia , Adjuvantes Imunológicos , Animais , Células Produtoras de Anticorpos , Antígenos CD4/fisiologia , Células Cultivadas , Feminino , Imunização/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/complicações , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/virologia , Pneumonia/etiologia
15.
J Immunol ; 195(5): 2472-82, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26216889

RESUMO

The development of an anti-influenza vaccine with the potential for cross-protection against seasonal drift variants as well as occasionally emerging reassortant viruses is essential. In this study, we successfully generated a novel anti-influenza vaccine system combining conserved matrix protein 2 (sM2) and stalk domain of hemagglutinin (HA2) fusion protein (sM2HA2) and poly-γ-glutamic acid (γ-PGA)-based vaccine adjuvant systems that can act as a mucoadhesive delivery vehicle of sM2HA2 as well as a robust strategy for the incorporation of hydrophobic immunostimulatory 3-O-desacyl-4'-monophosphoryl lipid A (MPL) and QS21. Intranasal coadministration of sM2HA2 and the combination adjuvant γ-PGA/MPL/QS21 (CA-PMQ) was able to induce a high degree of protective mucosal, systemic, and cell-mediated immune responses. The sM2HA2/CA-PMQ immunization was able to prevent disease symptoms, confering complete protection against lethal infection with divergent influenza subtypes (H5N1, H1N1, H5N2, H7N3, and H9N2) that lasted for at least 6 mo. Therefore, our data suggest that mucosal administration of sM2HA2 in combination with CA-PMQ could be a potent strategy for a broad cross-protective influenza vaccine, and CA-PMQ as a mucosal adjuvant could be used for effective mucosal vaccines.


Assuntos
Adjuvantes Imunológicos/química , Vacinas contra Influenza/química , Vacinas contra Influenza/imunologia , Polímeros/química , Adjuvantes Imunológicos/administração & dosagem , Animais , Proteção Cruzada/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Interações Hospedeiro-Patógeno/imunologia , Sistema Imunitário/imunologia , Imunidade Celular/imunologia , Imunidade nas Mucosas/imunologia , Imunização , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Virus da Influenza A Subtipo H5N1/imunologia , Virus da Influenza A Subtipo H5N1/fisiologia , Vírus da Influenza A Subtipo H5N2/imunologia , Vírus da Influenza A Subtipo H5N2/fisiologia , Vírus da Influenza A Subtipo H7N3/imunologia , Vírus da Influenza A Subtipo H7N3/fisiologia , Vírus da Influenza A Subtipo H9N2/imunologia , Vírus da Influenza A Subtipo H9N2/fisiologia , Vacinas contra Influenza/administração & dosagem , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Infecções por Orthomyxoviridae/virologia , Ácido Poliglutâmico/análogos & derivados , Ácido Poliglutâmico/química , Ácido Poliglutâmico/imunologia , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/imunologia
16.
Nature ; 470(7335): 543-7, 2011 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-21350488

RESUMO

Many successful vaccines induce persistent antibody responses that can last a lifetime. The mechanisms by which they do so remain unclear, but emerging evidence indicates that they activate dendritic cells via Toll-like receptors (TLRs). For example, the yellow fever vaccine YF-17D, one of the most successful empiric vaccines ever developed, activates dendritic cells via multiple TLRs to stimulate proinflammatory cytokines. Triggering specific combinations of TLRs in dendritic cells can induce synergistic production of cytokines, which results in enhanced T-cell responses, but its impact on antibody responses remain unknown. Learning the critical parameters of innate immunity that program such antibody responses remains a major challenge in vaccinology. Here we demonstrate that immunization of mice with synthetic nanoparticles containing antigens plus ligands that signal through TLR4 and TLR7 induces synergistic increases in antigen-specific, neutralizing antibodies compared to immunization with nanoparticles containing antigens plus a single TLR ligand. Consistent with this there was enhanced persistence of germinal centres and of plasma-cell responses, which persisted in the lymph nodes for >1.5 years. Surprisingly, there was no enhancement of the early short-lived plasma-cell response relative to that observed with single TLR ligands. Molecular profiling of activated B cells, isolated 7 days after immunization, indicated that there was early programming towards B-cell memory. Antibody responses were dependent on direct triggering of both TLRs on B cells and dendritic cells, as well as on T-cell help. Immunization protected completely against lethal avian and swine influenza virus strains in mice, and induced robust immunity against pandemic H1N1 influenza in rhesus macaques.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Formação de Anticorpos/imunologia , Imunidade Inata/imunologia , Memória Imunológica/imunologia , Vacinas contra Influenza/imunologia , Animais , Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Células Dendríticas/citologia , Células Dendríticas/imunologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Vacinas contra Influenza/administração & dosagem , Ácido Láctico , Ligantes , Linfonodos/citologia , Linfonodos/imunologia , Ativação Linfocitária , Macaca mulatta/imunologia , Macaca mulatta/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Nanopartículas/administração & dosagem , Plasmócitos/citologia , Plasmócitos/imunologia , Plasmócitos/metabolismo , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Linfócitos T/imunologia , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo
17.
Arch Phys Med Rehabil ; 98(12): 2442-2448, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28645769

RESUMO

OBJECTIVE: To examine differences in access to preventive services for working-age adults with physical limitations, nonphysical limitations, and no limitations. DESIGN: Observational data were pooled across calendar years 2003 through 2012 from the Medical Expenditure Panel Survey (MEPS), a nationally representative survey of noninstitutionalized U.S. adults. SETTING: Community. PARTICIPANTS: Working-age adults (N=75,145; age range, 18-64y) who responded to all 5 rounds of the MEPS during 2003 through 2012. INTERVENTIONS: Not applicable. MAIN OUTCOME MEASURES: Blood pressure checks, flu shots, and dental visits, in the last 12 months. RESULTS: Multivariate analyses showed that adults with physical limitations were more likely to receive a blood pressure check (92.2% vs 69.6%, respectively; P<.001) or flu shot (39.7% vs 23.4%, respectively; P<.001) than adults with no limitations. However, those with physical limitations were less likely to have a dental checkup (44.7% vs 59.4%, respectively; P<.001) than those with no limitation. Having a usual source of care increased the odds of receiving preventive services. CONCLUSIONS: The study has implications for providers and policymakers. Policies that support the adequacy of provider networks and continuity with a usual source of care may increase the use of preventive services in adults with physical limitations.


Assuntos
Acidentes por Quedas/estatística & dados numéricos , Pessoas com Deficiência/estatística & dados numéricos , Acessibilidade aos Serviços de Saúde/estatística & dados numéricos , Atenção Primária à Saúde/estatística & dados numéricos , Adolescente , Adulto , Pressão Sanguínea , Doença Crônica , Feminino , Humanos , Vacinas contra Influenza/administração & dosagem , Masculino , Saúde Mental , Pessoa de Meia-Idade , Saúde Bucal , Fatores Socioeconômicos , Adulto Jovem
18.
Nanomedicine ; 13(7): 2169-2178, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28579436

RESUMO

The most promising strategy to sustainably prevent infectious diseases is vaccination. However, emerging as well as re-emerging diseases still constitute a considerable threat. Furthermore, lack of compliance and logistic constrains often result in the failure of vaccination campaigns. To overcome these hurdles, novel vaccination strategies need to be developed, which fulfill maximal safety requirements, show maximal efficiency and are easy to administer. Mucosal vaccines constitute promising non-invasive approaches able to match these demands. Here we demonstrate that nanoparticle (polyphosphazenes)-based vaccine formulations including c-di-AMP as adjuvant, cationic innate defense regulator peptides (IDR) and ovalbumin (OVA) as model antigen were able to stimulate strong humoral and cellular immune responses, which conferred protection against the OVA expressing influenza strain A/WSN/OVAI (H1N1). The presented results confirm the potency of nanoparticle-based vaccine formulations to deliver antigens across the mucosal barrier, but also demonstrate the necessity to include adjuvants to stimulate efficient antigen-specific immune responses.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Fosfatos de Dinucleosídeos/administração & dosagem , Vacinas contra Influenza/administração & dosagem , Nanopartículas/química , Compostos Organofosforados/química , Infecções por Orthomyxoviridae/prevenção & controle , Ovalbumina/administração & dosagem , Polímeros/química , Adjuvantes Imunológicos/uso terapêutico , Administração Intranasal , Animais , Fosfatos de Dinucleosídeos/uso terapêutico , Feminino , Humanos , Imunidade Celular , Imunidade Humoral , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/uso terapêutico , Influenza Humana/imunologia , Influenza Humana/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Infecções por Orthomyxoviridae/imunologia , Ovalbumina/uso terapêutico , Vacinação/métodos
19.
J Liposome Res ; 27(3): 210-220, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28922045

RESUMO

Given the interest in the ectodomain of the matrix 2 (M2e) channel protein as a target for development of a universal influenza vaccine, we examined the role of the antigen configuration of M2e in generating a protective immune response. A series of M2e mutations and a truncated M2e segment were prepared as a means of controlling the formation of monomer, dimer, and higher order multimeric forms of M2e. Each of these M2e peptides was incorporated into a liposome-based vaccine technology platform previously shown to stimulate a protective response to influenza A infection using M2e as a mixture of monomers, dimers and multimers (L-M2e1-HD/MPL). Our results using these modified forms of M2e produced 90-100% survival following lethal challenge with H1N1 (A/PR/8/34) in both inbred BALB/c and outbred Swiss Webster mice vaccinated with a truncated monomeric form of the M2 protein, M2e1-15 in liposomes. These observations show that a tetrameric configuration is not required to elicit significant protection when the M2e antigen is formulated in immunogenic liposomes and further, that the first 15 amino acids of M2e likely play a primary role in providing the protective immune response.


Assuntos
Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vacinas contra Influenza/imunologia , Infecções por Orthomyxoviridae/prevenção & controle , Proteínas da Matriz Viral/imunologia , Animais , Feminino , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/química , Lipossomos , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Infecções por Orthomyxoviridae/imunologia , Multimerização Proteica , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/genética
20.
J Infect Dis ; 214(5): 722-31, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27247344

RESUMO

BACKGROUND: Tonsils play a key role in eliciting immune responses against respiratory pathogens. Little is known about how tonsils contribute to the local immune response after intranasal vaccination. Here, we uniquely report the mucosal humoral responses in tonsils and saliva after intranasal live attenuated influenza vaccine (LAIV) vaccination in children. METHODS: Blood, saliva, and tonsils samples were collected from 39 children before and after LAIV vaccination and from 16 age-matched, nonvaccinated controls. Serum antibody responses were determined by a hemagglutination inhibition (HI) assay. The salivary immunoglobulin A (IgA) level was measured by an enzyme-linked immunosorbent assay. Antibody-secreting cell (ASC) and memory B-cell (MBC) responses were enumerated in tonsils and blood. RESULTS: Significant increases were observed in levels of serum antibodies and salivary IgA to influenza A(H3N2) and influenza B virus strains as early as 14 days after vaccination but not to influenza A(H1N1). Influenza virus-specific salivary IgA levels correlated with serum HI responses, making this a new possible indicator of vaccine immunogenicity in children. LAIV augmented influenza virus-specific B-cell responses in tonsils and blood. Tonsillar MBC responses correlated with systemic MBC and serological responses. Naive children showed significant increases in MBC counts after LAIV vaccination. CONCLUSIONS: This is the first study to demonstrate that LAIV elicits humoral B-cell responses in tonsils of young children. Furthermore, salivary IgA analysis represents an easy method for measuring immunogenicity after vaccination.


Assuntos
Anticorpos Antivirais/análise , Linfócitos B/imunologia , Vacinas contra Influenza/imunologia , Tonsila Palatina/imunologia , Administração Intranasal , Adolescente , Células Apresentadoras de Antígenos/imunologia , Sangue/imunologia , Criança , Pré-Escolar , Ensaio de Imunoadsorção Enzimática , Feminino , Testes de Inibição da Hemaglutinação , Humanos , Imunoglobulina A/análise , Vacinas contra Influenza/administração & dosagem , Masculino , Saliva/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA