Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Immunol Cell Biol ; 101(6): 514-524, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36401824

RESUMO

Influenza A viruses (IAVs) exist as distinct serological subtypes, with limited antibody cross reactivity compared with T-cell responses, leading to universal vaccines that elicit robust T-cell responses entering clinical trials to combat pandemic and zoonotic outbreaks. Previously we have extensively characterized the viral-vectored universal vaccine, Wyeth/IL-15/5flu, a group 1 hemagglutinin, H5N1-based vaccine using a vaccinia backbone with interleukin (IL)-15. The vaccine elicits robust T-cell responses to provide heterosubtypic protection from lethal infection; however, we have also observed short-term morbidity of vaccinated mice with a disparity between the effects of sublethal infection with group 1 and 2 IAV strains. At day 3 of H3N2 (group 2 IAV) infection, there was a heavily skewed T helper type 1 response in vaccinated infected mice with overproduction of cytokines and reduced chemokines, whereas H1N1 (group 1 IAV) infection had increased innate cellular responses. These findings suggest that increased and early immune activation by T-cell activating vaccines may induce mild immunopathology when there is a mismatch between non-neutralizing antibody and cross-reactive memory T-cell responses leading to exuberant cytokine production. Therefore, to avoid overstimulating proinflammatory immune responses upon infection, universal influenza vaccines that elicit strong T-cell immunity will need a robust cross-reactive antibody response.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Virus da Influenza A Subtipo H5N1 , Vacinas contra Influenza , Influenza Humana , Infecções por Orthomyxoviridae , Animais , Camundongos , Humanos , Citocinas , Vírus da Influenza A Subtipo H3N2 , Anticorpos Antivirais
2.
Nat Med ; 29(1): 147-157, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36228659

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine immunogenicity varies between individuals, and immune responses correlate with vaccine efficacy. Using data from 1,076 participants enrolled in ChAdOx1 nCov-19 vaccine efficacy trials in the United Kingdom, we found that inter-individual variation in normalized antibody responses against SARS-CoV-2 spike and its receptor-binding domain (RBD) at 28 days after first vaccination shows genome-wide significant association with major histocompatibility complex (MHC) class II alleles. The most statistically significant association with higher levels of anti-RBD antibody was HLA-DQB1*06 (P = 3.2 × 10-9), which we replicated in 1,677 additional vaccinees. Individuals carrying HLA-DQB1*06 alleles were less likely to experience PCR-confirmed breakthrough infection during the ancestral SARS-CoV-2 virus and subsequent Alpha variant waves compared to non-carriers (hazard ratio = 0.63, 0.42-0.93, P = 0.02). We identified a distinct spike-derived peptide that is predicted to bind differentially to HLA-DQB1*06 compared to other similar alleles, and we found evidence of increased spike-specific memory B cell responses in HLA-DQB1*06 carriers at 84 days after first vaccination. Our results demonstrate association of HLA type with Coronavirus Disease 2019 (COVID-19) vaccine antibody response and risk of breakthrough infection, with implications for future vaccine design and implementation.


Assuntos
Infecções Irruptivas , Vacinas contra COVID-19 , COVID-19 , Antígenos de Histocompatibilidade Classe II , Imunogenicidade da Vacina , Humanos , Alelos , Anticorpos Antivirais , ChAdOx1 nCoV-19 , COVID-19/genética , COVID-19/prevenção & controle , Vacinas contra COVID-19/imunologia , SARS-CoV-2 , Vacinação
3.
Sci Adv ; 8(14): eabl5209, 2022 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-35385318

RESUMO

To determine the potential for viral adaptation to T cell responses, we probed the full influenza virus genome by next-generation sequencing directly ex vivo from infected mice, in the context of an experimental T cell-based vaccine, an H5N1-based viral vectored vaccinia vaccine Wyeth/IL-15/5Flu, versus the current standard-of-care, seasonal inactivated influenza vaccine (IIV) and unvaccinated conditions. Wyeth/IL-15/5Flu vaccination was coincident with increased mutation incidence and frequency across the influenza genome; however, mutations were not enriched within T cell epitope regions, but high allele frequency mutations within conserved hemagglutinin stem regions and PB2 mammalian adaptive mutations arose. Depletion of CD4+ and CD8+ T cell subsets led to reduced frequency of mutants in vaccinated mice; therefore, vaccine-mediated T cell responses were important drivers of virus diversification. Our findings suggest that Wyeth/IL-15/5Flu does not generate T cell escape mutants but increases stochastic events for virus adaptation by stringent bottlenecks.


Assuntos
Virus da Influenza A Subtipo H5N1 , Vacinas contra Influenza , Influenza Humana , Animais , Genoma Viral , Humanos , Virus da Influenza A Subtipo H5N1/genética , Influenza Humana/prevenção & controle , Interleucina-15/genética , Camundongos , Mutação , Infecções por Orthomyxoviridae , Prevalência , Vacinação
4.
Viruses ; 13(9)2021 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-34578360

RESUMO

Next generation influenza vaccines that target conserved epitopes are becoming a clinical reality but still have challenges to overcome. Universal next generation vaccines are considered a vital tool to combat future pandemic viruses and have the potential to vastly improve long-term protection against seasonal influenza viruses. Key vaccine strategies include HA-stem and T cell activating vaccines; however, they could have unintended effects for virus adaptation as they recognise the virus after cell entry and do not directly block infection. This may lead to immune pressure on residual viruses. The potential for immune escape is already evident, for both the HA stem and T cell epitopes, and mosaic approaches for pre-emptive immune priming may be needed to circumvent key variants. Live attenuated influenza vaccines have not been immunogenic enough to boost T cells in adults with established prior immunity. Therefore, viral vectors or peptide approaches are key to harnessing T cell responses. A plethora of viral vector vaccines and routes of administration may be needed for next generation vaccine strategies that require repeated long-term administration to overcome vector immunity and increase our arsenal against diverse influenza viruses.


Assuntos
Vacinas contra Influenza/imunologia , Orthomyxoviridae/imunologia , Vacinas contra Adenovirus , Animais , Anticorpos Antivirais , Linfócitos T CD8-Positivos/imunologia , Epitopos , Humanos , Influenza Humana , Infecções por Orthomyxoviridae , Linfócitos T/imunologia , Vacinação , Vacinas Atenuadas/imunologia
5.
Front Med (Lausanne) ; 8: 793102, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-35004764

RESUMO

T cell responses are a key cornerstone to viral immunity to drive high-quality antibody responses, establishing memory for recall and for viral clearance. Inefficient recruitment of T cell responses plays a role in the development of severe COVID-19 and is also represented by reduced cellular responses in men, children, and diversity compared with other epitope-specific subsets and available T cell receptor diversity. SARS-CoV-2-specific T cell responses are elicited by multiple vaccine formats and augmented by prior infection for hybrid immunity. Epitope conservation is relatively well-maintained leading to T cell crossreactivity for variants of concern that have diminished serological responses.

6.
Front Immunol ; 9: 1479, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30013557

RESUMO

Influenza viruses circulate worldwide causing annual epidemics that have a substantial impact on public health. This is despite vaccines being in use for over 70 years and currently being administered to around 500 million people each year. Improvements in vaccine design are needed to increase the strength, breadth, and duration of immunity against diverse strains that circulate during regular epidemics, occasional pandemics, and from animal reservoirs. Universal vaccine strategies that target more conserved regions of the virus, such as the hemagglutinin (HA)-stalk, or recruit other cellular responses, such as T cells and NK cells, have the potential to provide broader immunity. Many pre-pandemic vaccines in clinical development do not utilize new vaccine platforms but use "tried and true" recombinant HA protein or inactivated virus strategies despite substantial leaps in fundamental research on universal vaccines. Significant hurdles exist for universal vaccine development from bench to bedside, so that promising preclinical data is not yet translating to human clinical trials. Few studies have assessed immune correlates derived from asymptomatic influenza virus infections, due to the scale of a study required to identity these cases. The realization and implementation of a universal influenza vaccine requires identification and standardization of set points of protective immune correlates, and consideration of dosage schedule to maximize vaccine uptake.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...