Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Virol ; 95(4)2021 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-33268518

RESUMO

Swine influenza A virus (swIAV) infection causes substantial economic loss and disease burden in humans and animals. The 2009 pandemic H1N1 (pH1N1) influenza A virus is now endemic in both populations. In this study, we evaluated the efficacy of different vaccines in reducing nasal shedding in pigs following pH1N1 virus challenge. We also assessed transmission from immunized and challenged pigs to naive, directly in-contact pigs. Pigs were immunized with either adjuvanted, whole inactivated virus (WIV) vaccines or virus-vectored (ChAdOx1 and MVA) vaccines expressing either the homologous or heterologous influenza A virus hemagglutinin (HA) glycoprotein, as well as an influenza virus pseudotype (S-FLU) vaccine expressing heterologous HA. Only two vaccines containing homologous HA, which also induced high hemagglutination inhibitory antibody titers, significantly reduced virus shedding in challenged animals. Nevertheless, virus transmission from challenged to naive, in-contact animals occurred in all groups, although it was delayed in groups of vaccinated animals with reduced virus shedding.IMPORTANCE This study was designed to determine whether vaccination of pigs with conventional WIV or virus-vectored vaccines reduces pH1N1 swine influenza A virus shedding following challenge and can prevent transmission to naive in-contact animals. Even when viral shedding was significantly reduced following challenge, infection was transmissible to susceptible cohoused recipients. This knowledge is important to inform disease surveillance and control strategies and to determine the vaccine coverage required in a population, thereby defining disease moderation or herd protection. WIV or virus-vectored vaccines homologous to the challenge strain significantly reduced virus shedding from directly infected pigs, but vaccination did not completely prevent transmission to cohoused naive pigs.


Assuntos
Vírus da Influenza A Subtipo H1N1 , Vacinas contra Influenza/administração & dosagem , Infecções por Orthomyxoviridae/transmissão , Doenças dos Suínos/transmissão , Eliminação de Partículas Virais , Adjuvantes Imunológicos/administração & dosagem , Animais , Feminino , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/isolamento & purificação , Infecções por Orthomyxoviridae/prevenção & controle , Suínos , Doenças dos Suínos/prevenção & controle , Vacinação , Vacinas Atenuadas/administração & dosagem , Vacinas de Produtos Inativados/administração & dosagem
2.
Eur Respir J ; 57(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33060153

RESUMO

BACKGROUND: Respiratory syncytial virus (RSV) infection in older adults is recognised as an important health issue. We aimed to assess the community burden of RSV in Europe in older adults aged ≥60 years. METHODS: This international, prospective, observational cohort study is part of work by the REspiratory Syncytial virus Consortium in EUrope (RESCEU). Participants were recruited through general practitioners' (GPs) offices before two independent RSV seasons. Participants reported weekly about symptoms of acute respiratory tract infection (ARTI) during one RSV season. ARTI patients were tested for RSV during home visits and completed a daily symptom diary. RSV illness included PCR-confirmed ARTI and those showing seroconversion over the season. RSV ARTI was based on PCR alone (ClinicalTrials.gov, NCT03621930). RESULTS: We recruited 1040 participants (527 in season 2017-2018 and 513 in season 2018-2019) with a median age of 75 years (range 60-100 years). Of these, 1023 (99%) lived independently at home at baseline. RSV illness incidence was 22 out of 527 (4.2%) and 37 out of 513 (7.2%) in the respective seasons. RSV illness did not affect frailty or cardiopulmonary status during the course of the study. No patients were hospitalised or died from RSV illness. In the 36 patients with PCR confirmed RSV ARTI, symptom duration averaged 19 days, while a doctor's visit took place in 11 out of 36 cases (31%). RSV ARTI could not be differentiated clinically from all other ARTIs based on symptoms. CONCLUSION: This European study showed that RSV is prevalent in community-dwelling older adults and rarely causes severe disease. This suggests that watchful waiting, using a continuity of care approach to identify those who do need more intensive care, is often justified when RSV is suspected in family practice.


Assuntos
Infecções por Vírus Respiratório Sincicial , Vírus Sincicial Respiratório Humano , Idoso , Idoso de 80 Anos ou mais , Europa (Continente)/epidemiologia , Hospitalização , Humanos , Vida Independente , Lactente , Pessoa de Meia-Idade , Estudos Prospectivos , Infecções por Vírus Respiratório Sincicial/epidemiologia , Fatores de Risco
3.
J Allergy Clin Immunol ; 137(1): 214-222.e2, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26414879

RESUMO

BACKGROUND: Transmission of mucosal pathogens relies on their ability to bind to the surfaces of epithelial cells, to cross this thin barrier, and to gain access to target cells and tissues, leading to systemic infection. This implies that pathogen-specific immunity at mucosal sites is critical for the control of infectious agents using these routes to enter the body. Although mucosal delivery would ensure the best onset of protective immunity, most of the candidate vaccines are administered through the parenteral route. OBJECTIVE: The present study evaluates the feasibility of delivering the chemically bound p24gag (referred to as p24 in the text) HIV antigen through secretory IgA (SIgA) in nasal mucosae in mice. RESULTS: We show that SIgA interacts specifically with mucosal microfold cells present in the nasal-associated lymphoid tissue. p24-SIgA complexes are quickly taken up in the nasal cavity and selectively engulfed by mucosal dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin-positive dendritic cells. Nasal immunization with p24-SIgA elicits both a strong humoral and cellular immune response against p24 at the systemic and mucosal levels. This ensures effective protection against intranasal challenge with recombinant vaccinia virus encoding p24. CONCLUSION: This study represents the first example that underscores the remarkable potential of SIgA to serve as a carrier for a protein antigen in a mucosal vaccine approach targeting the nasal environment.


Assuntos
Proteína do Núcleo p24 do HIV/imunologia , Imunização/métodos , Imunoglobulina A Secretora/imunologia , Tecido Linfoide/imunologia , Mucosa Nasal/imunologia , Administração Intranasal , Animais , Citocinas/imunologia , Células Dendríticas , Feminino , Imunidade nas Mucosas , Imunoglobulina A/imunologia , Imunoglobulina G/imunologia , Tecido Linfoide/citologia , Camundongos Endogâmicos BALB C , Camundongos Knockout , Mucosa Nasal/citologia , Baço/imunologia , Vagina/imunologia
4.
Clin Immunol ; 173: 44-49, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27910805

RESUMO

The recent outbreak of Ebola virus disease in West Africa has led to more than 11,000 deaths, with a peak in mortality from August through December of 2014. A meeting convened by the World Health Organization (WHO) in September 2014, concluded that an urgent unmet need exists for efficacy and safety testing of the Ebola virus vaccine candidates and that clinical trials should be expedited. These vaccines could be used both in an outbreak setting and to provide long-term protection in populations at risk of sporadic outbreaks. A number of vaccines have been evaluated in phase 1 trials, but the two most advanced first-generation Ebola vaccine candidates are the live replicating vesicular stomatitis virus (rVSV) and the replication-defective chimpanzee adenovirus 3 (ChAd3). This review focuses on these two vaccines in clinical development and discusses the future opportunities and challenges faced in the licensure and deployment of Ebola virus vaccines.


Assuntos
Vacinas contra Ebola , Adenoviridae/genética , Adenoviridae/imunologia , Ebolavirus/genética , Ebolavirus/imunologia , Glicoproteínas/genética , Glicoproteínas/imunologia , Humanos , Vesiculovirus/genética , Vesiculovirus/imunologia , Proteínas Virais/genética , Proteínas Virais/imunologia
5.
Eur J Immunol ; 45(3): 773-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25412898

RESUMO

HIV transmission and spread in the host are based on the survival of the virus or infected cells present in mucosal secretions, and the virus' ability to cross the epithelial barrier and access immune target cells, which leads to systemic infection. Therefore, HIV-specific immunity at mucosal sites is critical for control of infection. Although mucosal delivery would ensure the best onset of protective immunity, most candidate vaccines are administered through the parenteral route. Remarkably, secretory IgA (SIgA) interacts specifically with mucosal microfold (M) cells present in gut-associated lymphoid tissues. Here we evaluate the feasibility of delivering chemically bound p24HIV antigen via SIgA into the intestinal mucosae in mice. After oral administration, p24-SIgA complexes are quickly delivered into the tissue and selectively captured by CX3CR1(+) dendritic cells. Oral immunization with p24gag linked to SIgA (p24-SIgA) adjuvanted with E. coli heat labile enterotoxin (HLT) elicits both humoral and cellular immune responses against p24 at the systemic and mucosal levels and induces efficient protection against rectal challenge with a recombinant vaccinia virus encoding gag. This is the first study which underscores the remarkable potential of SIgA to serve as a vaccine carrier for an HIV antigen in mucosal administration targeting the gastrointestinal environment.


Assuntos
Vacinas contra a AIDS/farmacologia , Portadores de Fármacos/farmacologia , Proteína do Núcleo p24 do HIV/farmacologia , HIV-1/imunologia , Imunoglobulina A Secretora/farmacologia , Mucosa Intestinal/imunologia , Vacinas contra a AIDS/imunologia , Animais , Proteína do Núcleo p24 do HIV/imunologia , Imunidade Celular/efeitos dos fármacos , Imunidade Humoral/efeitos dos fármacos , Imunoglobulina A Secretora/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos
6.
PLoS Biol ; 11(9): e1001658, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24068891

RESUMO

Intestinal microfold (M) cells possess a high transcytosis capacity and are able to transport a broad range of materials including particulate antigens, soluble macromolecules, and pathogens from the intestinal lumen to inductive sites of the mucosal immune system. M cells are also the primary pathway for delivery of secretory IgA (SIgA) to the gut-associated lymphoid tissue. However, although the consequences of SIgA uptake by M cells are now well known and described, the mechanisms whereby SIgA is selectively bound and taken up remain poorly understood. Here we first demonstrate that both the Cα1 region and glycosylation, more particularly sialic acid residues, are involved in M cell-mediated reverse transcytosis. Second, we found that SIgA is taken up by M cells via the Dectin-1 receptor, with the possible involvement of Siglec-5 acting as a co-receptor. Third, we establish that transcytosed SIgA is taken up by mucosal CX3CR1⁺ dendritic cells (DCs) via the DC-SIGN receptor. Fourth, we show that mucosal and systemic antibody responses against the HIV p24-SIgA complexes administered orally is strictly dependent on the expression of Dectin-1. Having deciphered the mechanisms leading to specific targeting of SIgA-based Ag complexes paves the way to the use of such a vehicle for mucosal vaccination against various infectious diseases.


Assuntos
Complexo Antígeno-Anticorpo/metabolismo , Imunoglobulina A Secretora/imunologia , Mucosa Intestinal/imunologia , Lectinas Tipo C/metabolismo , Transcitose/imunologia , Animais , Complexo Antígeno-Anticorpo/imunologia , Antígenos CD/metabolismo , Antígenos de Diferenciação Mielomonocítica/metabolismo , Células CHO , Receptor 1 de Quimiocina CX3C , Células CACO-2 , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Cricetulus , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Glicosilação , Proteína do Núcleo p24 do HIV/imunologia , Células HeLa , Humanos , Imunoglobulina A/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Intestinos/citologia , Lectinas/metabolismo , Lectinas Tipo C/biossíntese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ácido N-Acetilneuramínico/química , Receptores de Superfície Celular/metabolismo , Receptores de Quimiocinas/metabolismo
7.
J Immunol ; 193(12): 5781-5, 2014 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-25392526

RESUMO

TLR ligands are critical activators of innate immunity and are being developed as vaccine adjuvants. However, their usefulness in conjunction with NOD-like receptor agonists remains poorly studied. In this study, we evaluated a new ligand that targets both TLR2 and NOD2 receptors. We assessed its ability to enhance dendritic cell maturation in vitro in addition to improving systemic and mucosal immune responses in mice. The chimeric NOD2/TLR2 ligand induced synergistic upregulation of dendritic cell maturation markers, costimulatory molecules, and secretion of proinflammatory cytokines compared with combinations of separate ligands. Furthermore, when coadministered with biodegradable nanoparticles carrying a model Ag, the ligand was able to induce high Ag-specific IgA and IgG titers at both systemic and mucosal sites after parenteral immunizations. These findings point out the potential utility of chimeric molecules TLR/NOD as adjuvants for vaccines to induce systemic and mucosal immune responses.


Assuntos
Adjuvantes Imunológicos/metabolismo , Ligantes , Proteína Adaptadora de Sinalização NOD2/metabolismo , Receptor 2 Toll-Like/metabolismo , Vacinas/imunologia , Acetilmuramil-Alanil-Isoglutamina/análogos & derivados , Acetilmuramil-Alanil-Isoglutamina/química , Acetilmuramil-Alanil-Isoglutamina/imunologia , Adjuvantes Imunológicos/química , Animais , Biomarcadores/metabolismo , Diferenciação Celular , Citocinas/biossíntese , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Proteína do Núcleo p24 do HIV/imunologia , Humanos , Imunidade nas Mucosas/imunologia , Imunoglobulina A/sangue , Imunoglobulina A/imunologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Lipopeptídeos/genética , Lipopeptídeos/imunologia , Camundongos , Fenótipo , Vacinas/genética
8.
Pharm Res ; 32(1): 311-20, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25248333

RESUMO

PURPOSE: Activation of immune cells through pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs) or NOD-like receptors (NLRs), has been identified as a key issue in the development of new efficient vaccine adjuvants. We report here on the elaboration and immunostimulatory potential of polylactide (PLA)-based micelles core-loaded with imiquimod TLR7 ligand and able to be further surface-functionalized with antigenic protein (HIV-1 Gag p24) for antigen delivery purpose. METHODS: Micelles prepared from poly(D,L-lactide)-b-poly(N-acryloxysuccinimide-co-N-vinylpyrrolidone) amphiphilic copolymer were incubated in the presence of imiquimod, leading to 1.2 wt% loading, and further conjugated to p24 antigen through reaction of p24 lysines and N-terminal amine with the N-succinimidyl pendant groups of the micelle corona. The impact of imiquimod encapsulation in the micelles on its immunostimulatory properties was investigated in vitro, by monitoring: (i) the NF-κB and mitogen-activated protein kinases (MAPK) pathways through experiments with RAW-Blue™ cells, a mouse macrophage cell line encoding an NF-κB/AP-1-inducible reporter construct; (ii) human dendritic cells (DCs) maturation markers by flow cytometry. RESULTS: RAW-Blue™ cells based experiments showed that imiquimod encapsulated in the micelles was much more efficient to activate the NF-κB and MAPK pathways than free imiquimod. Furthermore, encapsulated imiquimod was found to induce much higher maturation of DCs than the free analog. Finally, these immunostimulatory properties of the loaded imiquimod were shown to be conserved when the p24 antigen was coupled at the micelle surface. CONCLUSIONS: Taken together, these data regarding improved immunostimulatory efficiency suggest the strong potential of our micelle-based nano-system for vaccine delivery.


Assuntos
Adjuvantes Imunológicos/administração & dosagem , Aminoquinolinas/administração & dosagem , Portadores de Fármacos/química , Poliésteres/química , Povidona/análogos & derivados , Vacinas/administração & dosagem , Adjuvantes Imunológicos/química , Adjuvantes Imunológicos/farmacologia , Aminoquinolinas/química , Aminoquinolinas/imunologia , Aminoquinolinas/farmacologia , Animais , Linhagem Celular , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Liberação Controlada de Fármacos , Citometria de Fluxo , Proteína do Núcleo p24 do HIV/imunologia , Humanos , Imiquimode , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Micelas , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Tamanho da Partícula , Povidona/química , Propriedades de Superfície
9.
Front Immunol ; 14: 1100161, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36845117

RESUMO

Introduction: Pulmonary-resident memory T cells (TRM) and B cells (BRM) orchestrate protective immunity to reinfection with respiratory pathogens. Developing methods for the in situ detection of these populations would benefit both research and clinical settings. Methods: To address this need, we developed a novel in situ immunolabelling approach combined with clinic-ready fibre-based optical endomicroscopy (OEM) to detect canonical markers of lymphocyte tissue residency in situ in human lungs undergoing ex vivo lung ventilation (EVLV). Results: Initially, cells from human lung digests (confirmed to contain TRM/BRM populations using flow cytometry) were stained with CD69 and CD103/CD20 fluorescent antibodies and imaged in vitro using KronoScan, demonstrating it's ability to detect antibody labelled cells. We next instilled these pre-labelled cells into human lungs undergoing EVLV and confirmed they could still be visualised using both fluorescence intensity and lifetime imaging against background lung architecture. Finally, we instilled fluorescent CD69 and CD103/CD20 antibodies directly into the lung and were able to detect TRM/BRM following in situ labelling within seconds of direct intra-alveolar delivery of microdoses of fluorescently labelled antibodies. Discussion: In situ, no wash, immunolabelling with intra-alveolar OEM imaging is a novel methodology with the potential to expand the experimental utility of EVLV and pre-clinical models.


Assuntos
Memória Imunológica , Pulmão , Humanos , Pulmão/diagnóstico por imagem , Linfócitos T CD8-Positivos , Linfócitos
10.
Nat Commun ; 14(1): 1309, 2023 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-36894558

RESUMO

The rapid spread of the SARS-CoV-2 Omicron subvariants, despite the implementation of booster vaccination, has raised questions about the durability of protection conferred by current vaccines. Vaccine boosters that can induce broader and more durable immune responses against SARS-CoV-2 are urgently needed. We recently reported that our Beta-containing protein-based SARS-CoV-2 spike booster vaccine candidates with AS03 adjuvant (CoV2 preS dTM-AS03) elicited robust cross-neutralizing antibody responses at early timepoints against SARS-CoV-2 variants of concern in macaques primed with mRNA or protein-based subunit vaccine candidates. Here we demonstrate that the monovalent Beta vaccine with AS03 adjuvant induces durable cross-neutralizing antibody responses against the prototype strain D614G as well as variants Delta (B.1.617.2), Omicron (BA.1 and BA.4/5) and SARS-CoV-1, that are still detectable in all macaques 6 months post-booster. We also describe the induction of consistent and robust memory B cell responses, independent of the levels measured post-primary immunization. These data suggest that a booster dose with a monovalent Beta CoV2 preS dTM-AS03 vaccine can induce robust and durable cross-neutralizing responses against a broad spectrum of variants.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Humanos , SARS-CoV-2/genética , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Anticorpos Amplamente Neutralizantes , Subunidades Proteicas , Macaca , Primatas , Anticorpos Antivirais , Anticorpos Neutralizantes
11.
Commun Med (Lond) ; 3(1): 75, 2023 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-37237062

RESUMO

BACKGROUND: Since the beginning of the COVID-19 pandemic, several variants of concern (VOC) have emerged for which there is evidence of an increase in transmissibility, more severe disease, and/or reduced vaccine effectiveness. Effective COVID-19 vaccine strategies are required to achieve broad protective immunity against current and future VOC. METHODS: We conducted immunogenicity and challenge studies in macaques and hamsters using a bivalent recombinant vaccine formulation containing the SARS-CoV-2 prefusion-stabilized Spike trimers of the ancestral D614 and the variant Beta strains with AS03 adjuvant (CoV2 preS dTM-AS03) in a primary immunization setting. RESULTS: We show that a primary immunization with the bivalent CoV2 preS dTM-AS03 elicits broader and durable (1 year) neutralizing antibody responses against VOC including Omicron BA.1 and BA.4/5, and SARS-CoV-1 as compared to the ancestral D614 or Beta variant monovalent vaccines in naïve non-human primates. In addition, the bivalent formulation confers protection against viral challenge with SARS-CoV-2 prototype D614G strain as well as Alpha and Beta variant strains in hamsters. CONCLUSIONS: Our findings demonstrate the potential of a Beta-containing bivalent CoV2 preS dTM-AS03 formulation to provide broad and durable immunogenicity, as well as protection against VOC in naïve populations.


SARS-CoV-2 has changed over time, resulting in different forms of the virus called variants. These variants compromise the protection offered by the COVID-19 vaccines, which trigger an immune response against the viral Spike protein that allows the virus to attach and infect human cells, since their spike proteins are different. Here, we developed and tested a vaccine containing two different Spike proteins, one from the original Wuhan strain and another from the Beta variant. In macaques, the vaccine leads to the production of antibodies able to stop all variants tested from infecting human cells, including Omicron, with stable levels over one year. In hamsters, the vaccine protected against infection with the ancestral virus and the Alpha and Beta variants. Our findings have important implications for vaccine control of existing and future SARS-CoV-2 variants of concern.

12.
Front Microbiol ; 14: 1065609, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37350788

RESUMO

The development of virus-like particle (VLP) based vaccines for human papillomavirus, hepatitis B and hepatitis E viruses represented a breakthrough in vaccine development. However, for dengue and COVID-19, technical complications, such as an incomplete understanding of the requirements for protective immunity, but also limitations in processes to manufacture VLP vaccines for enveloped viruses to large scale, have hampered VLP vaccine development. Selecting the right adjuvant is also an important consideration to ensure that a VLP vaccine induces protective antibody and T cell responses. For diseases like COVID-19 and dengue fever caused by RNA viruses that exist as families of viral variants with the potential to escape vaccine-induced immunity, the development of more efficacious vaccines is also necessary. Here, we describe the development and characterisation of novel VLP vaccine candidates using SARS-CoV-2 and dengue virus (DENV), containing the major viral structural proteins, as protypes for a novel approach to produce VLP vaccines. The VLPs were characterised by Western immunoblot, enzyme immunoassay, electron and atomic force microscopy, and in vitro and in vivo immunogenicity studies. Microscopy techniques showed proteins self-assemble to form VLPs authentic to native viruses. The inclusion of the glycolipid adjuvant, α-galactosylceramide (α-GalCer) in the vaccine formulation led to high levels of natural killer T (NKT) cell stimulation in vitro, and strong antibody and memory CD8+ T cell responses in vivo, demonstrated with SARS-CoV-2, hepatitis C virus (HCV) and DEN VLPs. This study shows our unique vaccine formulation presents a promising, and much needed, new vaccine platform in the fight against infections caused by enveloped RNA viruses.

13.
Nat Commun ; 13(1): 1699, 2022 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-35361754

RESUMO

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants that partly evade neutralizing antibodies raises concerns of reduced vaccine effectiveness and increased infection. We previously demonstrated that the SARS-CoV-2 spike protein vaccine adjuvanted with AS03 (CoV2 preS dTM-AS03) elicits robust neutralizing antibody responses in naïve subjects. Here we show that, in macaques primed with mRNA or protein-based subunit vaccine candidates, one booster dose of CoV2 preS dTM-AS03 (monovalent D614 or B.1.351, or bivalent D614 + B.1.351 formulations), significantly boosts the pre-existing neutralizing antibodies against the parental strain from 177- to 370-fold. Importantly, the booster dose elicits high and persistent cross-neutralizing antibodies covering five former or current SARS-CoV-2 variants of concern (Alpha, Beta, Gamma, Delta and Omicron) and, unexpectedly, SARS-CoV-1. Interestingly, we show that the booster specifically increases the functional antibody responses as compared to the receptor binding domain (RBD)-specific responses. Our findings show that these vaccine candidates, when used as a booster, have the potential to offer cross-protection against a broad spectrum of variants. This has important implications for vaccine control of SARS-CoV-2 variants of concern and informs on the benefit of a booster with the vaccine candidates currently under evaluation in clinical trials.


Assuntos
COVID-19 , Vacinas Virais , Animais , Anticorpos Antivirais , COVID-19/prevenção & controle , Vacinas contra COVID-19 , Humanos , Primatas , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus
14.
NPJ Vaccines ; 7(1): 74, 2022 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-35773301

RESUMO

Respiratory syncytial virus (RSV) G glycoprotein has recently reemerged as a vaccine antigen due to its ability to elicit potent neutralizing antibodies and ameliorate disease in animal models. Here we designed three constructs to display the G central conserved domain (Gcc) focused on inducing broad and potent neutralizing antibodies. One construct displaying Gcc from both RSV subgroups trimerized via a C-terminal foldon (Gcc-Foldon) was highly immunogenic in mice and in MIMIC, a pre-immune human in vitro model. To explore an optimal RSV vaccine, we combined the Gcc-Foldon antigen with a stabilized pre-fusion-F nanoparticle (pre-F-NP) as a bivalent vaccine and detected no antigenic interference between the two antigens in the MIMIC model. In RSV-primed macaques, the bivalent vaccine elicited potent humoral responses. Furthermore, both Gcc-Foldon and the bivalent vaccine conferred effective protection against RSV challenge in mice. This two-component vaccine could potentially provide effective protection against RSV infection in humans and warrants further clinical evaluation.

15.
Hum Vaccin Immunother ; 17(7): 2336-2348, 2021 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-33427044

RESUMO

Adjuvants are central to the efficacy of subunit vaccines. Although several new adjuvants have been approved in human vaccines over the last decade, the panel of adjuvants in licensed human vaccines remains small. There is still a need for novel adjuvants that can be safely used in humans, easy to source and to formulate with a wide range of antigens and would be broadly applicable to a wide range of vaccines. In this article, using the Respiratory Syncytial Virus (RSV) nanoparticulate prefusion F model antigen developed by Sanofi, we demonstrate in the macaque model that the polyacrylate (PAA)-based adjuvant SPA09 is well tolerated and increases vaccine antigen-specific humoral immunity (sustained neutralizing antibodies, memory B cells and mucosal immunity) and elicits strong TH1-type responses (based on IFNγ and IL-2 ELISpots) in a dose-dependent manner. These data warrant further development of the SPA09 adjuvant for evaluation in clinical trials.


Assuntos
Infecções por Vírus Respiratório Sincicial , Vacinas contra Vírus Sincicial Respiratório , Adjuvantes Imunológicos , Animais , Anticorpos Neutralizantes , Anticorpos Antivirais , Humanos , Imunidade Celular , Imunidade Humoral , Macaca fascicularis
16.
Front Vet Sci ; 8: 679248, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34113679

RESUMO

Bronchoalveolar lavage, or BAL, is a minimally invasive procedure frequently used for clinical and non-clinical research, allowing studies of the respiratory system. Macaques are the most widely used non-human primate models in biomedical research. However, very little information is available in the literature concerning BAL cytology in macaques. The purpose of this study was to establish BAL reference values and document an atlas of BAL cytology from healthy cynomolgus macaques. BALs were obtained from 30 macaques and BAL fluid differential cell counts based on 400 nucleated cells/BAL sample were performed by a board-certified clinical pathologist. Results were analyzed using Reference Value Advisor macroinstructions and the effect of blood and oropharyngeal contaminations was investigated. Overall, nucleated cells interval percentages in BAL fluids were 55.8 to 93.7 for macrophages, 1.8 to 37.1 for lymphocytes, 0.4 to 8.7 for neutrophils, and 0.4 to 9.8 for eosinophils. Mild oropharyngeal contamination did not affect BAL differential cell counts, whilst a slight but significant increase of the percentage of lymphocytes was observed in samples with mild blood contamination. Mucus and variable numbers of ciliated epithelial cells were commonly present. Rarely, multinucleated macrophages and mastocytes were also observed. The reference intervals established in this study provide a useful baseline for the assessment of BAL cytological data in cynomolgus macaques.

17.
Front Immunol ; 12: 738955, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34603321

RESUMO

There is increasing evidence that lung-resident memory T and B cells play a critical role in protecting against respiratory reinfection. With a unique transcriptional and phenotypic profile, resident memory lymphocytes are maintained in a quiescent state, constantly surveying the lung for microbial intruders. Upon reactivation with cognate antigen, these cells provide rapid effector function to enhance immunity and prevent infection. Immunization strategies designed to induce their formation, alongside novel techniques enabling their detection, have the potential to accelerate and transform vaccine development. Despite most data originating from murine studies, this review will discuss recent insights into the generation, maintenance and characterisation of pulmonary resident memory lymphocytes in the context of respiratory infection and vaccination using recent findings from human and non-human primate studies.


Assuntos
Infecções Bacterianas/prevenção & controle , Memória Imunológica , Pulmão/imunologia , Células B de Memória/imunologia , Células T de Memória/imunologia , Infecções Respiratórias/imunologia , Viroses/prevenção & controle , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/metabolismo , Infecções Bacterianas/microbiologia , Vacinas Bacterianas/administração & dosagem , Vacinas Bacterianas/imunologia , Interações Hospedeiro-Patógeno , Humanos , Pulmão/metabolismo , Pulmão/microbiologia , Pulmão/virologia , Células B de Memória/metabolismo , Células B de Memória/microbiologia , Células B de Memória/virologia , Células T de Memória/metabolismo , Células T de Memória/microbiologia , Células T de Memória/virologia , Fenótipo , Infecções Respiratórias/microbiologia , Infecções Respiratórias/prevenção & controle , Infecções Respiratórias/virologia , Vacinação , Vacinas Virais/administração & dosagem , Vacinas Virais/imunologia , Viroses/imunologia , Viroses/metabolismo , Viroses/microbiologia
18.
NPJ Vaccines ; 5(1): 19, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194996

RESUMO

The recent spread of Zika virus (ZIKV) through the Americas and Caribbean and its devastating consequences for pregnant women and their babies have driven the search for a safe and efficacious ZIKV vaccine. Among the vaccine candidates, a first-generation ZIKV purified inactivated vaccine (ZPIV), adjuvanted with aluminum hydroxide, developed by the Walter Reed Army Institute of Research (WRAIR), has elicited high seroconversion rates in participants in three phase-I clinical trials. In collaboration with the WRAIR, Sanofi Pasteur (SP) optimized the production scale, culture and purification conditions, and increased the regulatory compliance, both of which are critical for clinical development and licensure of this vaccine. Using a clinical batch of the first-generation ZPIV as a benchmark, we report that different doses of the optimized vaccine (ZPIV-SP) elicited sustained neutralizing antibodies, specific T- and memory B-cells, and provided complete protection against a ZIKV challenge in cynomolgus macaques. These data provide evidence that the ZPIV-SP vaccine performs at least as well as the ZPIV vaccine, and provide support for continued development in the event of future ZIKV outbreaks.

19.
Sci Immunol ; 5(47)2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32358170

RESUMO

A stabilized form of the respiratory syncytial virus (RSV) fusion (F) protein has been explored as a vaccine to prevent viral infection because it presents several potent neutralizing epitopes. Here, we used a structure-based rational design to optimize antigen presentation and focus antibody (Ab) responses to key epitopes on the pre-fusion (pre-F) protein. This protein was fused to ferritin nanoparticles (pre-F-NP) and modified with glycans to mask nonneutralizing or poorly neutralizing epitopes to further focus the Ab response. The multimeric pre-F-NP elicited durable pre-F-specific Abs in nonhuman primates (NHPs) after >150 days and elicited potent neutralizing Ab (NAb) responses in mice and NHPs in vivo, as well as in human cells evaluated in the in vitro MIMIC system. This optimized pre-F-NP stimulated a more potent Ab response than a representative pre-F trimer, DS-Cav1. Collectively, this pre-F vaccine increased the generation of NAbs targeting the desired pre-F conformation, an attribute that facilitates the development of an effective RSV vaccine.


Assuntos
Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Nanopartículas/química , Vacinas contra Vírus Sincicial Respiratório/imunologia , Vírus Sincicial Respiratório Humano/química , Proteínas Virais de Fusão/imunologia , Animais , Formação de Anticorpos , Antígenos Virais/imunologia , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Vacinas contra Vírus Sincicial Respiratório/química , Vírus Sincicial Respiratório Humano/imunologia , Proteínas Virais de Fusão/química
20.
Vaccine ; 37(37): 5567-5577, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31399277

RESUMO

Seasonal influenza virus infections cause significant morbidity and mortality every year. Annual influenza virus vaccines are effective but only when well matched with circulating strains. Therefore, there is an urgent need for better vaccines that induce broad protection against drifted seasonal and emerging pandemic influenza viruses. One approach to design such vaccines is based on targeting conserved regions of the influenza virus hemagglutinin. Sequential vaccination with chimeric hemagglutinin constructs can refocus antibody responses towards the conserved immunosubdominant stalk domain of the hemagglutinin, rather than the variable immunodominant head. A complementary approach for a universal influenza A virus vaccine is to induce T-cell responses to conserved internal influenza virus antigens. For this purpose, replication deficient recombinant viral vectors based on Chimpanzee Adenovirus Oxford 1 and Modified Vaccinia Ankara virus are used to express the viral nucleoprotein and the matrix protein 1. In this study, we combined these two strategies and evaluated the efficacy of viral vectors expressing both chimeric hemagglutinin and nucleoprotein plus matrix protein 1 in a mouse model against challenge with group 2 influenza viruses including H3N2, H7N9 and H10N8. We found that vectored vaccines expressing both sets of antigens provided enhanced protection against H3N2 virus challenge when compared to vaccination with viral vectors expressing only one set of antigens. Vaccine induced antibody responses against divergent group 2 hemagglutinins, nucleoprotein and matrix protein 1 as well as robust T-cell responses to the nucleoprotein and matrix protein 1 were detected. Of note, it was observed that while antibodies to the H3 stalk were already boosted to high levels after two vaccinations with chimeric hemagglutinins (cHAs), three exposures were required to induce strong reactivity across subtypes. Overall, these results show that a combinations of different universal influenza virus vaccine strategies can induce broad antibody and T-cell responses and can provide increased protection against influenza.


Assuntos
Vetores Genéticos , Glicoproteínas de Hemaglutininação de Vírus da Influenza/imunologia , Imunização , Infecções por Orthomyxoviridae/prevenção & controle , Proteínas de Ligação a RNA/imunologia , Vacinas de DNA/imunologia , Proteínas do Core Viral/imunologia , Proteínas da Matriz Viral/imunologia , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Citotoxicidade Celular Dependente de Anticorpos , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Modelos Animais de Doenças , Feminino , Vetores Genéticos/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Imunidade Celular , Camundongos , Proteínas do Nucleocapsídeo , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Vacinas de DNA/genética , Proteínas do Core Viral/genética , Proteínas da Matriz Viral/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa