Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 224
Filtrer
1.
Virol J ; 20(1): 187, 2023 08 21.
Article de Anglais | MEDLINE | ID: mdl-37605141

RÉSUMÉ

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.


Sujet(s)
Vaccins antigrippaux , Grippe humaine , Vaccins à virosomes , Lavage bronchoalvéolaire , Sous-type H1N1 du virus de la grippe A , Sous-type H1N2 du virus de la grippe A , Sous-type H3N2 du virus de la grippe A , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/immunologie , Grippe humaine/immunologie , Rate/cytologie , Rate/immunologie , Vaccins combinés/administration et posologie , Vaccins à virosomes/administration et posologie , Vaccins à virosomes/immunologie , Virosomes/ultrastructure , Humains , Animaux , Souris
2.
Sci Rep ; 12(1): 3884, 2022 03 10.
Article de Anglais | MEDLINE | ID: mdl-35273217

RÉSUMÉ

Current SARS-CoV-2 vaccines are effective, but long-term protection is threatened by the emergence of virus variants. We generated a virosome vaccine containing the Beta spike protein and compared its immunogenicity in mice to a virosome vaccine containing the original Wuhan spike. Two administrations of the virosomes induced potent SARS-CoV-2 neutralizing antibodies in both vaccine groups. The level of autologous neutralization in Beta-vaccinated mice was similar to the level of autologous neutralization in Wuhan-vaccinated mice. However, heterologous neutralization to the Wuhan strain in Beta-vaccinated mice was 4.7-fold lower than autologous neutralization, whereas heterologous neutralization to the Beta strain in Wuhan-vaccinated mice was reduced by only 1.9-fold compared to autologous neutralization levels. In addition, neutralizing activity against the D614G, Alpha and Delta variants was also significantly lower after Beta spike vaccination than after Wuhan spike vaccination. Our results show that Beta spike vaccination induces inferior neutralization breadth. These results are informative for programs aimed to develop broadly active SARS-CoV-2 vaccines.


Sujet(s)
Vaccins contre la COVID-19/usage thérapeutique , SARS-CoV-2/immunologie , Glycoprotéine de spicule des coronavirus/immunologie , Animaux , Anticorps neutralisants/immunologie , Tests d'analyse de l'haleine , Vaccins contre la COVID-19/immunologie , Femelle , Souris , Souris de lignée BALB C , Tests de neutralisation , Vaccins à virosomes/immunologie , Vaccins à virosomes/usage thérapeutique
3.
Front Immunol ; 12: 747774, 2021.
Article de Anglais | MEDLINE | ID: mdl-34887855

RÉSUMÉ

The highly pathogenic avian influenza H5N1 viruses constantly evolve and give rise to novel variants that have caused widespread zoonotic outbreaks and sporadic human infections. Therefore, vaccines capable of eliciting broadly protective antibody responses are desired and under development. We here investigated the magnitude, kinetics and protective efficacy of the multi-faceted humoral immunity induced by vaccination in healthy adult volunteers with a Matrix M adjuvanted virosomal H5N1 vaccine. Vaccinees were given escalating doses of adjuvanted vaccine (1.5µg, 7.5µg, or 30µg), or a non-adjuvanted vaccine (30µg). An evaluation of sera from vaccinees against pseudotyped viruses covering all (sub)clades isolated from human H5N1 infections demonstrated that the adjuvanted vaccines (7.5µg and 30µg) could elicit rapid and robust increases of broadly cross-neutralizing antibodies against all clades. In addition, the adjuvanted vaccines also induced multifaceted antibody responses including hemagglutinin stalk domain specific, neuraminidase inhibiting, and antibody-dependent cellular cytotoxicity inducing antibodies. The lower adjuvanted dose (1.5µg) showed delayed kinetics, whilst the non-adjuvanted vaccine induced overall lower levels of antibody responses. Importantly, we demonstrate that human sera post vaccination with the adjuvanted (30µg) vaccine provided full protection against a lethal homologous virus challenge in mice. Of note, when combining our data from mice and humans we identified the neutralizing and neuraminidase inhibiting antibody titers as correlates of in vivo protection.


Sujet(s)
Anticorps antiviraux/sang , Anticorps neutralisants à large spectre/sang , Sous-type H5N1 du virus de la grippe A/immunologie , Vaccins antigrippaux/immunologie , Adulte , Animaux , Réactions croisées , Femelle , Humains , Immunisation passive , Vaccins antigrippaux/administration et posologie , Grippe humaine/immunologie , Grippe humaine/prévention et contrôle , Grippe humaine/virologie , Mâle , Souris , Souris de lignée BALB C , Adulte d'âge moyen , Sialidase/antagonistes et inhibiteurs , Sialidase/immunologie , Saponines de Quillaja/administration et posologie , Saponines de Quillaja/immunologie , Vaccins à virosomes/administration et posologie , Vaccins à virosomes/immunologie , Jeune adulte
4.
Front Immunol ; 12: 711997, 2021.
Article de Anglais | MEDLINE | ID: mdl-34326849

RÉSUMÉ

Influenza A virus is one of the most important zoonotic pathogens that can cause severe symptoms and has the potential to cause high number of deaths and great economic loss. Vaccination is still the best option to prevent influenza virus infection. Different types of influenza vaccines, including live attenuated virus vaccines, inactivated whole virus vaccines, virosome vaccines, split-virion vaccines and subunit vaccines have been developed. However, they have several limitations, such as the relatively high manufacturing cost and long production time, moderate efficacy of some of the vaccines in certain populations, and lack of cross-reactivity. These are some of the problems that need to be solved. Here, we summarized recent advances in the development and application of different types of influenza vaccines, including the recent development of viral vectored influenza vaccines. We also described the construction of other vaccines that are based on recombinant influenza viruses as viral vectors. Information provided in this review article might lead to the development of safe and highly effective novel influenza vaccines.


Sujet(s)
Vaccins antigrippaux , Développement de vaccin , Animaux , Poulets , Prévision , Vecteurs génétiques , Humains , Virus de la grippe A/génétique , Virus de la grippe A/immunologie , Grippe chez les oiseaux/prévention et contrôle , Grippe humaine/épidémiologie , Grippe humaine/prévention et contrôle , Grippe humaine/virologie , Maladies de la volaille/prévention et contrôle , Saisons , Développement de vaccin/tendances , Vaccins atténués , Vaccins inactivés , Vaccins sous-unitaires , Vaccins synthétiques , Vaccins à virosomes , Virion
5.
Biomed Res Int ; 2021: 8879277, 2021.
Article de Anglais | MEDLINE | ID: mdl-33575353

RÉSUMÉ

Newcastle disease (ND) is a highly fatal, infectious, viral disease, and despite immunization with live and inactivated vaccines, the disease is still endemic, causing heavy morbidity and mortality leading to huge economic losses to the poultry industry in Pakistan. Therefore, the present study was aimed for the first time in the country at using novel virosomal technology to develop the ND vaccine using an indigenous highly virulent strain of the virus. ND virosome was prepared using Triton X-100, and SM2 Bio-Beads were used to remove the detergent and reconstitute the viral membrane into virosome. Confirmation was done by transmission electron microscopy and protein analysis by SDS-PAGE. In vitro cell adhesion property was observed by incorporating green fluorescent protein (GFP), producing plasmid into virosome and in vitro cell culture assay. Sterility, safety, and stability of the vaccine were tested before in vivo evaluation of immunogenicity and challenge protection study in commercial broiler. The virosome vaccine was administered (30 µg/bird) at days 7 and 14 through the intranasal route in comparison with commercially available live and inactivated ND vaccines. Results revealed significantly high (p < 0.05) and clinically protective hemagglutination inhibition (HI) antibody titers at 7, 14, 21, and 28 days postimmunization with the virosome vaccine in comparison to the negative control. The GMTs were comparable to live and inactivated vaccines with nonsignificant (p > 0.05) differences throughout the experiment. Antibody levels increased in all vaccinated groups gradually from the 7th day and were maximum at 28th-day postvaccination. In the virosome-administered group, GMT was 83.18 and 77.62 at 21st and 28th-days postvaccination, respectively. Challenge revealed 100%, 90%, and 80% protection in virosome, live, and inactivated vaccinated groups, respectively. Under given experimental conditions, we can conclude that ND virosome vaccine prepared from the indigenous virus was found to be safe and immunogenic.


Sujet(s)
Maladie de Newcastle , Maladies de la volaille , Vaccins à virosomes , Animaux , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Poulets , Maladie de Newcastle/immunologie , Maladie de Newcastle/prévention et contrôle , Virus de la maladie de Newcastle/génétique , Virus de la maladie de Newcastle/immunologie , Pakistan , Maladies de la volaille/immunologie , Maladies de la volaille/prévention et contrôle , Vaccins à virosomes/composition chimique , Vaccins à virosomes/immunologie , Vaccins à virosomes/métabolisme , Virosomes/immunologie
6.
Pharm Res ; 35(9): 172, 2018 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-29971500

RÉSUMÉ

PURPOSE: Characterization of virosomes, in late stage preclinical development as vaccines for Respiratory Syncytial Virus (RSV), with a membrane-incorporated synthetic monophosphoryl lipid A, 3D-PHAD® adjuvant. METHODS: Virosomes were initially formed by contacting a lipid film containing 3D-PHAD® with viral membranes solubilized with the short chain phospholipid DCPC, followed by dialysis, later by adding solubilized 3D-PHAD to viral membranes, or to preformed virosomes from DMSO. RESULTS: Virosomes formed from lipid films contained the membrane glycoproteins G and F, at similar F to G ratios but lower concentrations than in virus, and the added lipids, but only a fraction of the 3D-PHAD®. By single particle tracking (SPT), the virosome size distribution resembled that seen by cryo-electron microscopy, but dynamic light scattering showed much larger particles. These differences were caused by small virosome aggregates. Measured by SPT, virosomes were stable for 300 days. 3DPHAD ® incorporation in virosomes could be enhanced by providing the adjuvant from DCPC solubilized stock, but also by adding DMSO dissolved adjuvant to pre-formed virosomes. Virosomes with 0.1 mg/mg of 3D-PHAD®/viral protein from DMSO induced antibody titers similar to those by virosomes containing 0.2 mg/mg of DCPC-solubilized 3D-PHAD®. CONCLUSIONS: Stable 3D-PHAD® adjuvanted RSV virosomes can be formulated.


Sujet(s)
Adjuvants immunologiques/composition chimique , Infections à virus respiratoire syncytial/prévention et contrôle , Vaccins contre les virus respiratoires syncytiaux/composition chimique , Virus respiratoires syncytiaux/immunologie , Adjuvants immunologiques/administration et posologie , Adjuvants immunologiques/pharmacologie , Animaux , Chlorocebus aethiops , Préparation de médicament , Femelle , Humains , Immunisation , Souris de lignée BALB C , Infections à virus respiratoire syncytial/immunologie , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Vaccins contre les virus respiratoires syncytiaux/pharmacologie , Vaccins à virosomes/administration et posologie , Vaccins à virosomes/composition chimique , Vaccins à virosomes/pharmacologie , Cellules Vero
7.
Curr Opin Pharmacol ; 41: 34-41, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-29677646

RÉSUMÉ

Immunosenescence contributes to increased incidence and severity of many infections in old age and is responsible for impaired immunogenicity and efficacy of vaccines. Adjuvants are one strategy to enhance immunogenicity of vaccines. The oil-in-water emulsions MF59TM and AS03, as well as a virosomal vaccine have been licensed in seasonal or pandemic influenza vaccines and are/were used successfully in the elderly. AS01, a liposome-based adjuvant comprising two immunostimulants has recently been approved in a recombinant protein vaccine for older adults, which showed very high efficacy against herpes zoster in clinical trials. Several adjuvants for use in the older population are in clinical and preclinical development and will hopefully improve vaccines for this age group in the future.


Sujet(s)
Adjuvants immunologiques/administration et posologie , Immunogénicité des vaccins , Immunosénescence , Vaccination/méthodes , Sujet âgé , Vaccin contre le zona/immunologie , Humains , Vaccins antigrippaux/immunologie , Vaccins à virosomes/immunologie , Infection à virus varicelle-zona/prévention et contrôle
8.
Vaccine ; 34(44): 5262-5272, 2016 10 17.
Article de Anglais | MEDLINE | ID: mdl-27667332

RÉSUMÉ

BACKGROUND: Influenza remains a significant problem in elderly despite widespread vaccination coverage. This randomized, phase-I study in elderly compared different strategies of improving vaccine immunogenicity. METHODS: A total of 370 healthy participants (⩾65years) were randomized equally 1:1:1:1:1:1 to six influenza vaccine treatments (approximately 60-63 participants per treatment arm) at day 1 that consisted of three investigational virosomal vaccine formulations at doses of 7.5, 15, and 45µg HA antigen/strain administered intradermally (ID) by MicronJet600™ microneedle device (NanoPass Technologies) or intramuscularly (IM), and three comparator registered seasonal vaccines; Inflexal V™ (Janssen) and MF59 adjuvanted Fluad™ (Novartis) administered IM and Intanza™ (Sanofi Pasteur) administered ID via Soluvia™ prefilled microinjection system (BD). Serological evaluations were performed at days 22 and 90 and safety followed-up for 6months. RESULTS: Intradermal delivery of virosomal vaccine using MicronJet600™ resulted in significantly higher immunogenicity than the equivalent dose of virosomal Inflexal V™ administered intramuscularly across most of the parameters and strains, as well as in some of the readouts and strains as compared with the 45µg dose of virosomal vaccine formulation. Of 370 participants, 300 (81.1%) reported ⩾1 adverse event (AE); more participants reported solicited local AEs (72.2%) than solicited systemic AEs (12.2%). CONCLUSIONS: Intradermal delivery significantly improved influenza vaccine immunogenicity compared with intramuscular delivery. Triple dose (45µg) virosomal vaccine did not demonstrate any benefit on vaccine's immunogenicity over 15µg commercial presentation. All treatments were generally safe and well-tolerated.


Sujet(s)
Vieillissement/immunologie , Immunogénicité des vaccins , Vaccins antigrippaux/administration et posologie , Grippe humaine/prévention et contrôle , Adjuvants immunologiques , Sujet âgé , Sujet âgé de 80 ans ou plus , Anticorps antiviraux/sang , Relation dose-réponse (immunologie) , Voies d'administration de substances chimiques et des médicaments , Femelle , Humains , Vaccins antigrippaux/effets indésirables , Vaccins antigrippaux/immunologie , Grippe humaine/immunologie , Injections intradermiques , Injections musculaires , Mâle , Vaccins à virosomes/administration et posologie , Vaccins à virosomes/effets indésirables , Vaccins à virosomes/immunologie
9.
Pediatr Infect Dis J ; 35(7): e220-8, 2016 07.
Article de Anglais | MEDLINE | ID: mdl-27093164

RÉSUMÉ

BACKGROUND: The aim of this open-label, active-controlled, parallel group, phase 2 follow-up study was to assess the long-term immunogenicity of Epaxal Junior, the pediatric dose of an aluminum-free virosomal inactivated hepatitis A virus (HAV) vaccine, in children receiving routine childhood vaccines (RCV). METHODS: Healthy children (12-15 months old, ≥8 kg weight) were randomized (1:1:1) to group A: Epaxal Junior + RCV (day 1); group B: Epaxal Junior (day 1) + RCV (day 29) and group C: Havrix 720 + RCV (day 1). All 3 groups received 2 doses of HAV vaccines 6 months apart. Children who completed the primary study were followed up from 18 months to 7.5 years post booster. RESULTS: Of 291/327 randomized children who had completed the primary study, 157 were followed for the 7.5-year analysis (group A: 50; group B: 54; and group C: 53). Of these, 152 children had protective levels of anti-HAV antibodies [≥10 mIU/mL; 98% (group A); 96.3% (group B); 96.2% (group C)]. Anti-HAV geometric mean concentrations were similar in groups A and B at all the time points (1.5-, 2.5-, 3.5-, 5.25- and 7.5-year time point) but slightly lower in group C. Predictions of the median duration of persistence of seroprotective antibody levels, using the linear mixed model were similar in all groups: (group A: 19.1 years, group B: 18.7 years, group C: 17.3 years). CONCLUSIONS: Immunization with Epaxal Junior administered with RCVs at 12 months elicited protective response beyond 7.5 years in almost all children. Assessing the kinetic of anti-HAV antibody titers decline over time, the moment to reach antibody concentrations below the accepted protective level may occur earlier than previously estimated.


Sujet(s)
Vaccins anti-hépatite A/administration et posologie , Vaccins anti-hépatite A/sang , Femelle , Études de suivi , Hépatite A/immunologie , Hépatite A/prévention et contrôle , Anticorps de l'hépatite A/sang , Vaccins anti-hépatite A/effets indésirables , Vaccins anti-hépatite A/immunologie , Virus de l'hépatite A humaine/immunologie , Humains , Calendrier vaccinal , Rappel de vaccin , Nourrisson , Mâle , Vaccins inactivés/administration et posologie , Vaccins inactivés/effets indésirables , Vaccins inactivés/sang , Vaccins inactivés/immunologie , Vaccins à virosomes/administration et posologie , Vaccins à virosomes/effets indésirables , Vaccins à virosomes/sang , Vaccins à virosomes/immunologie
10.
Arch Virol ; 161(3): 705-10, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26666439

RÉSUMÉ

An optimized VP2 gene from the current prevalent CPV strain (new CPV-2a) in China was expressed in a baculovirus expression system. It was found that the VP2 proteins assembled into virus-like particles (VLPs) with antigenic properties similar to those of natural CPV and with an especially high hemagglutination (HA) titer (1:2(20)). Dogs intramuscularly or orally immunized with VLPs produced antibodies against CPV with >1:80 hemagglutination inhibition (HI) units for at least 3 months. The CPV VLPs could be considered for use as a vaccine against CPV or as a platform for research on chimeric VLP vaccines against other diseases.


Sujet(s)
Baculoviridae , Vecteurs génétiques , Parvovirus canin/génétique , Protéines virales structurales/métabolisme , Virosomes/métabolisme , Administration par voie orale , Animaux , Anticorps antiviraux/sang , Chine , Chiens , Injections musculaires , Multimérisation de protéines , Vaccins à virosomes/administration et posologie , Vaccins à virosomes/génétique , Vaccins à virosomes/immunologie , Protéines virales structurales/génétique , Vaccins antiviraux/administration et posologie , Vaccins antiviraux/génétique , Vaccins antiviraux/immunologie , Virosomes/génétique
11.
PLoS One ; 10(9): e0135723, 2015.
Article de Anglais | MEDLINE | ID: mdl-26402787

RÉSUMÉ

There is a constant threat of zoonotic influenza viruses causing a pandemic outbreak in humans. It is virtually impossible to predict which virus strain will cause the next pandemic and it takes a considerable amount of time before a safe and effective vaccine will be available once a pandemic occurs. In addition, development of pandemic vaccines is hampered by the generally poor immunogenicity of avian influenza viruses in humans. An effective pre-pandemic vaccine is therefore required as a first line of defense. Broadening of the protective efficacy of current seasonal vaccines by adding an adjuvant may be a way to provide such first line of defense. Here we evaluate whether a seasonal trivalent virosomal vaccine (TVV) adjuvated with the saponin-based adjuvant Matrix-M (MM) can confer protection against avian influenza H5 and H7 virus strains in mice and ferrets. We demonstrate that mice were protected from death against challenges with H5N1 and H7N7, but that the protection was not complete as evidenced by severe clinical signs. In ferrets, protection against H7N9 was not observed. In contrast, reduced upper and lower respiratory tract viral loads and reduced lung pathology, was achieved in H5N1 challenged ferrets. Together these results suggest that, at least to some extent, Matrix-M adjuvated seasonal virosomal influenza vaccine can serve as an interim measure to decrease morbidity and mortality associated with a pandemic outbreak.


Sujet(s)
Adjuvants immunologiques , Virus de la grippe A/immunologie , Vaccins antigrippaux/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Protéines de la matrice virale/immunologie , Animaux , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Réactions croisées/immunologie , Modèles animaux de maladie humaine , Femelle , Furets , Immunisation , Virus de la grippe A/classification , Vaccins antigrippaux/administration et posologie , Souris , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/virologie , Vaccins à virosomes , Réplication virale
12.
Enferm Infecc Microbiol Clin ; 33(7): 480-90, 2015.
Article de Espagnol | MEDLINE | ID: mdl-26232121

RÉSUMÉ

Seasonal influenza is an annual challenge for health-care systems, due to factors such as co-circulation of 2 influenza A subtypes jointly with 2 influenza B lineages; the antigenic drift of these virus, which eludes natural immunity, as well as immunity conferred by vaccination; together with influenza impact in terms of morbidity and mortality. Influenza vaccines have been available for more than 70 years and they have progressed in formulation, production and delivery route. Recommendations on vaccination are focused on those with a higher probability of severe disease, and have a progressively wider coverage, and classically based on inactivated vaccines, but with an increasing importance of attenuated live vaccines. More inactivated vaccines are becoming available, from adyuvanted and virosomal vaccines to intradermal delivery, cell-culture or quadrivalent. Overall vaccine effectiveness is about 65%, but varies depending on characteristics of vaccines, virus, population and the outcomes to be prevented, and ranges from less than 10% to almost 90%. Future challenges are formulations that confer more extensive and lasting protection, as well as increased vaccination coverage, especially in groups such as pregnant women and health-care professionals, as well as being extended to paediatrics.


Sujet(s)
Vaccins antigrippaux/immunologie , Grippe humaine/prévention et contrôle , Efficacité du vaccin , Adolescent , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Animaux , Variation des antigènes , Enfant , Enfant d'âge préscolaire , Épidémies de maladies/prévention et contrôle , Femelle , Prévision , Humains , Calendrier vaccinal , Nourrisson , Virus de la grippe A/immunologie , Virus influenza B/immunologie , Vaccins antigrippaux/classification , Grippe humaine/épidémiologie , Mâle , Adulte d'âge moyen , Guides de bonnes pratiques cliniques comme sujet , Grossesse , Vaccins atténués , Vaccins inactivés , Vaccins à virosomes , Jeune adulte , Zoonoses
13.
PLoS One ; 10(4): e0124392, 2015.
Article de Anglais | MEDLINE | ID: mdl-25909814

RÉSUMÉ

We have previously identified 17 biomarker genes which were upregulated by whole virion influenza vaccines, and reported that gene expression profiles of these biomarker genes had a good correlation with conventional animal safety tests checking body weight and leukocyte counts. In this study, we have shown that conventional animal tests showed varied and no dose-dependent results in serially diluted bulk materials of influenza HA vaccines. In contrast, dose dependency was clearly shown in the expression profiles of biomarker genes, demonstrating higher sensitivity of gene expression analysis than the current animal safety tests of influenza vaccines. The introduction of branched DNA based-concurrent expression analysis could simplify the complexity of multiple gene expression approach, and could shorten the test period from 7 days to 3 days. Furthermore, upregulation of 10 genes, Zbp1, Mx2, Irf7, Lgals9, Ifi47, Tapbp, Timp1, Trafd1, Psmb9, and Tap2, was seen upon virosomal-adjuvanted vaccine treatment, indicating that these biomarkers could be useful for the safety control of virosomal-adjuvanted vaccines. In summary, profiling biomarker gene expression could be a useful, rapid, and highly sensitive method of animal safety testing compared with conventional methods, and could be used to evaluate the safety of various types of influenza vaccines, including adjuvanted vaccine.


Sujet(s)
Adjuvants immunologiques/pharmacologie , Vaccins antigrippaux/pharmacologie , Adjuvants immunologiques/administration et posologie , Adjuvants immunologiques/effets indésirables , Animaux , Technique d'amplification de signal par ADN branché/méthodes , Analyse de profil d'expression de gènes/méthodes , Marqueurs génétiques , Humains , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/effets indésirables , Mâle , Contrôle de qualité , Rats , Rat Wistar , Sécurité , Régulation positive , Vaccins à virosomes/administration et posologie , Vaccins à virosomes/effets indésirables , Vaccins à virosomes/pharmacologie
14.
Pharm Res ; 32(4): 1505-15, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25344321

RÉSUMÉ

PURPOSE: Influenza CD8(+) T-cell epitopes are conserved amongst influenza strains and can be recognized by influenza-specific cytotoxic T-cells (CTLs), which can rapidly clear infected cells. An influenza peptide vaccine that elicits these CTLs would therefore be an alternative to current influenza vaccines, which are not cross-reactive. However, peptide antigens are poorly immunogenic due to lack of delivery to antigen presenting cells, and therefore need additional formulation with a suitable delivery system. In this study, the potential of virosomes as a delivery system for an influenza T-cell peptide was investigated. METHODS: The conserved human HLA-A2.1 influenza T-cell epitope M158-66 was formulated with virosomes. The immunogenicity and protective effect of the peptide-loaded virosomes was assessed in HLA-A2 transgenic mice. Delivery properties of the virosomes were studied in mice and in in vitro dendritic cell cultures. RESULTS: Immunization of HLA-A2.1 transgenic C57BL/6 mice with peptide-loaded virosomes in the presence of the adjuvant CpG-ODN 1826 increased the number of peptide-specific CTLs. Vaccination with adjuvanted peptide-loaded virosomes reduced weight loss in mice after heterologous influenza infection. Association with fusion-active virosomes was found to be crucial for antigen uptake by dendritic cells, and subsequent induction of CTLs in mice. CONCLUSIONS: These results show that influenza virosomes loaded with conserved influenza epitopes could be the basis of a novel cross-protective influenza vaccine.


Sujet(s)
Adjuvants immunologiques/composition chimique , Déterminants antigéniques des lymphocytes T/immunologie , Antigène HLA-A2/immunologie , Vaccins antigrippaux/administration et posologie , Oligodésoxyribonucléotides/composition chimique , Animaux , Antigène HLA-A2/génétique , Humains , Vaccins antigrippaux/composition chimique , Souris de lignée C57BL , Souris transgéniques , Fragments peptidiques/immunologie , Vaccins à virosomes/administration et posologie , Vaccins à virosomes/composition chimique , Protéines de la matrice virale/immunologie , Virosomes
15.
Pediatr Infect Dis J ; 34(4): e85-91, 2015 Apr.
Article de Anglais | MEDLINE | ID: mdl-25389920

RÉSUMÉ

BACKGROUND: The pediatric dose of the virosomal hepatitis A vaccine Epaxal, Epaxal Junior, is safe and immunogenic in children from 1 to 17 years of age. The present study investigated the long-term immunogenicity of Epaxal Junior. The standard doses of Epaxal and aluminum-adsorbed hepatitis A vaccine (Havrix Junior) were used as comparators. METHODS: A total of 271 children who had completed a 0/6-month immunization schedule (priming and booster dose) participated in this follow-up study. Anti-hepatitis A virus (HAV) antibody levels were measured using a microparticle enzyme immunoassay (HAVAB 2.0 Quantitative; Abbott Diagnostics, Wiesbaden, Germany) starting at 18 months following the second dose, and then yearly until 66 months (ie, 5.5 years) after the second dose. RESULTS: All subjects tested at Month 66 still had protective anti-HAV antibodies (≥10 mIU/mL). Antibody titers were generally lower in subjects 1-7 years old than in subjects 8-17 years old and higher in females 11-17 years old than in males 11-17 years old. In addition, an age-dependent decay was observed, that is, antibody decreased more rapidly in younger than in older children. CONCLUSIONS: Vaccination of children with two doses of Epaxal Junior confers a real-time protection of at least 5.5 years. This protection is estimated to last approximately 25 years. Younger children showed lower antibody titers and a faster antibody decline than older children. Additional follow-up studies are needed beyond 5.5 years to further assess the long-term immunogenicity of Epaxal Junior.


Sujet(s)
Anticorps de l'hépatite A/sang , Vaccins anti-hépatite A/administration et posologie , Vaccins anti-hépatite A/immunologie , Vaccination/méthodes , Vaccins à virosomes/administration et posologie , Vaccins à virosomes/immunologie , Adolescent , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Dosage immunologique , Nourrisson , Études longitudinales , Facteurs temps
16.
Hum Vaccin Immunother ; 10(7): 1969-73, 2014.
Article de Anglais | MEDLINE | ID: mdl-25424806

RÉSUMÉ

Acute disseminated encephalomyelitis (ADEM) is an inflammatory, usually monophasic, immune mediate, demyelinating disease of the central nervous system which involves the white matter. ADEM is more frequent in children and usually occurs after viral infections, but may follow vaccinations, bacterial infections, or may occur without previous events. Only 5% of cases of ADEM are preceded by vaccination within one month prior to symptoms onset. The diagnosis of ADEM requires both multifocal involvement and encephalopathy and specific demyelinating lesions of white matter. Overall prognosis of ADEM patients is often favorable, with full recovery reported in 23% to 100% of patients from pediatric cohorts, and more severe outcome in adult patients. We describe the first case of ADEM occurred few days after administration of virosomal seasonal influenza vaccine. The patient, a 59-year-old caucasic man with unremarkable past medical history presented at admission decreased alertness, 10 days after flu vaccination. During the 2 days following hospitalization, his clinical conditions deteriorated with drowsiness and fever until coma. The magnetic resonance imaging of the brain showed multiple and symmetrical white matter lesions in both cerebellar and cerebral hemispheres, suggesting demyelinating disease with inflammatory activity, compatible with ADEM. The patient was treated with high dose of steroids and intravenous immunoglobulin with relevant sequelae and severe neurological outcomes.


Sujet(s)
Encéphalomyélite aigüe disséminée/complications , Encéphalomyélite aigüe disséminée/diagnostic , Vaccins antigrippaux/effets indésirables , Maladies du système nerveux/diagnostic , Anti-inflammatoires/usage thérapeutique , Encéphale/imagerie diagnostique , Encéphale/anatomopathologie , Encéphalomyélite aigüe disséminée/traitement médicamenteux , Humains , Immunoglobulines par voie veineuse/usage thérapeutique , Facteurs immunologiques/usage thérapeutique , Vaccins antigrippaux/administration et posologie , Imagerie par résonance magnétique , Mâle , Adulte d'âge moyen , Radiographie , Stéroïdes/usage thérapeutique , Vaccins à virosomes/administration et posologie , Vaccins à virosomes/effets indésirables
17.
Hum Vaccin Immunother ; 10(7): 2089-97, 2014.
Article de Anglais | MEDLINE | ID: mdl-25424821

RÉSUMÉ

As India is transitioning from high to intermediate hepatitis A endemicity, the need for hepatitis A vaccination programs increases. This study investigated the immunogenicity and safety of a virosomal hepatitis A vaccine (HAVpur Junior) compared with an aluminum-adsorbed hepatitis A vaccine (Havrix 720 Junior) in Indian children. Healthy children aged 18-47 months, stratified by age, were randomized to either HAVpur Junior or Havrix 720 Junior. The first dose of vaccine was administered on Day 1 and the second (booster) dose 6 months later. Antibodies against hepatitis A virus (HAV) were measured using a microparticle enzyme immunoassay. The primary objective assessed non-inferiority of HAVpur Junior to Havrix 720 Junior in terms of seroprotection rates (≥ 10 mIU/mL anti-HAV antibodies) at 1 month after the first vaccination. Non-inferiority was demonstrated if the lower limit of the 90% confidence interval of the group difference was greater than -10%. Local and systemic adverse events were recorded. The seroprotection rate at 1 month was 95.9% in the HAVpur Junior group and 96.6% in the Havrix 720 Junior group. As the lower limit of the 90% confidence interval of the group difference was greater than -10% (-4.7), non-inferiority of HAVpur Junior to Havrix 720 Junior was established. The overall incidence of adverse events (solicited and unsolicited) after each vaccination was similar in both groups. In conclusion, the aluminum-free virosomal vaccine HAVpur Junior induced a similar immune response to Havrix 720 Junior in healthy Indian children aged 18 to 47 months. Both vaccines were well tolerated. The study shows that the low-dose virosomal HAV vaccine is consistently efficacious and well tolerated in children of all age groups and is suitable for inclusion into Indian childhood vaccination schedules.


Sujet(s)
Anticorps de l'hépatite A/sang , Vaccins anti-hépatite A/effets indésirables , Vaccins anti-hépatite A/immunologie , Hépatite A/prévention et contrôle , Adjuvants immunologiques/administration et posologie , Hydroxyde d'aluminium/administration et posologie , Enfant d'âge préscolaire , Effets secondaires indésirables des médicaments/épidémiologie , Effets secondaires indésirables des médicaments/anatomopathologie , Femelle , Volontaires sains , Vaccins anti-hépatite A/administration et posologie , Humains , Techniques immunoenzymatiques , Inde , Nourrisson , Mâle , Vaccins à virosomes/administration et posologie , Vaccins à virosomes/effets indésirables , Vaccins à virosomes/immunologie
18.
Hum Vaccin Immunother ; 10(8): 2408-16, 2014.
Article de Anglais | MEDLINE | ID: mdl-25424948

RÉSUMÉ

T cellular responses play a significant role in mediating protective immune responses against influenza in humans. In the current study, we evaluated the ability of a candidate virosomal H5N1 vaccine adjuvanted with Matrix M(TM) to induce CD4(+) and CD8(+) T cell responses in a phase 1 clinical trial. We vaccinated 60 healthy adult volunteers (at days 0 and 21) with 30 µg haemagglutinin (HA) alone or 1.5, 7.5, or 30 µg HA formulated with Matrix M(TM). To evaluate the T cellular responses, lymphocytes were stimulated in vitro with homologous (A/Vietnam/1194/2004 [H5N1]) and heterologous H5N1 (A/Anhui/1/05 or A/Bar-headed Goose/Qinghai/1A/05) antigens. The antigen-specific cytokine responses were measured by intracellular cytokine staining and by multiplex (Luminex) assays. An increase in CD4(+) Th1 and Th2 cytokines was detected 21 days after the first vaccine dose. No increase in Th cytokine responses was observed after the second dose, although it is possible that the cytokine levels peaked earlier than sampling point at day 42. Formulation with the Matrix M(TM) adjuvant augmented both the homologous and cross-reactive cytokine response. Antigen-specific CD8(+) T cell responses were detected only in a few vaccinated individuals. The concentrations of Th1 and to a lesser extent, Th2 cytokines at 21 days post-vaccination correlated moderately with subsequent days 35 and 180 serological responses as measured by the microneutralisation, haemagglutination inhibition, and single radial hemolysis assays. Results presented here show that the virosomal H5N1 vaccine induced balanced Th1/Th2 cytokine responses and that Matrix M(TM) is a promising adjuvant for future development of candidate pandemic influenza vaccines.


Sujet(s)
Adjuvants immunologiques/administration et posologie , Lymphocytes T CD4+/immunologie , Sous-type H5N1 du virus de la grippe A/immunologie , Vaccins antigrippaux/immunologie , Lymphocytes auxiliaires Th1/immunologie , Lymphocytes auxiliaires Th2/immunologie , Adulte , Lymphocytes T CD8+/immunologie , Cytokines/métabolisme , Femelle , Volontaires sains , Humains , Vaccins antigrippaux/administration et posologie , Mâle , Vaccins à virosomes/administration et posologie , Vaccins à virosomes/immunologie
19.
Vaccine ; 32(42): 5447-54, 2014 Sep 22.
Article de Anglais | MEDLINE | ID: mdl-25131737

RÉSUMÉ

BACKGROUND: The use of intradermal vaccination or virosomal vaccines could increase protection against influenza among the vulnerable population of older adults. Studies assessing the comparative effectiveness of these two influenza vaccine types in this age group are lacking. METHODS: We conducted a retrospective cohort study to estimate the comparative effectiveness of intradermal seasonal trivalent-influenza vaccine (TIV) delivered by a microneedle injection system and a virosomal-TIV intramuscularly delivered for prevention of influenza hospitalization in non-institutionalized adults aged ≥65 years. We obtained administrative data on immunization status and influenza hospitalization for the 2011-2012 influenza season, and used Cox regression models to assess comparative effectiveness. We estimated crude and adjusted (age, sex, comorbidity, pharmaceutical claims, recent pneumococcal vaccination and number of hospitalizations for all causes other than influenza between the previous and current influenza seasons) hazard ratios (HR). RESULTS: Overall, 164,021 vaccinated subjects were evaluated. There were 127 hospitalizations for influenza among 62,058 subjects, contributing 914,740 person-weeks at risk in the virosomal-TIV group, and 133 hospitalizations for influenza among 101,963 subjects, contributing 1,504,570 person-weeks at risk in the intradermal-TIV group. The crude HR of intradermal-TIV relative to virosomal-TIV was 0.64 (95% confidence interval (CI): 0.50-0.81), and the adjusted Cox estimated HR was 0.67 (95% CI: 0.52-0.85). CONCLUSIONS: During the 2011-2012 influenza season the risk of hospitalization for influenza was reduced by 33% in non-institutionalized elderly adults who were vaccinated with intradermal-TIV compared with virosomal-TIV.


Sujet(s)
Vaccins antigrippaux/usage thérapeutique , Grippe humaine/prévention et contrôle , Sujet âgé , Sujet âgé de 80 ans ou plus , Femelle , Hospitalisation/statistiques et données numériques , Humains , Vaccins antigrippaux/classification , Mâle , Modèles des risques proportionnels , Études rétrospectives , Espagne , Vaccins à virosomes/usage thérapeutique
20.
Expert Rev Vaccines ; 13(8): 1059-66, 2014 Aug.
Article de Anglais | MEDLINE | ID: mdl-24972949

RÉSUMÉ

AIM: To clarify the immunogenicity and safety of influenza vaccine in patients with end-stage kidney disease (ESKD) on dialysis or who have received a kidney transplant. METHODS: Sixty adolescents and young adults with ESKD (25 on hemodialysis and 35 kidney transplant recipients) were randomized 1:1 to receive a traditional trivalent split virion vaccine (TIIV) or a virosome-adjuvanted trivalent inactivated influenza vaccine (VATIIV). The immunogenicity and safety of the two vaccines was evaluated and compared with the findings observed in 30 healthy subjects of similar age and gender distribution who received TIIV. RESULTS: The results indicate that the immune response of the patients to TIIV and VATIIV were similar. The administered vaccines were safe and well tolerated, and no advantage was found with the use of VATIIV. CONCLUSION: Given the potential clinical relevance of influenza in patients with ESKD, these findings support the official recommendation that they should receive annual influenza vaccinations.


Sujet(s)
Immunisation/méthodes , Vaccins antigrippaux/administration et posologie , Grippe humaine/prévention et contrôle , Transplantation rénale , Dialyse rénale , Insuffisance rénale , Adolescent , Anticorps antiviraux/sang , Effets secondaires indésirables des médicaments/épidémiologie , Femelle , Humains , Immunisation/effets indésirables , Vaccins antigrippaux/effets indésirables , Vaccins antigrippaux/immunologie , Mâle , Études prospectives , Méthode en simple aveugle , Vaccins sous-unitaires/administration et posologie , Vaccins sous-unitaires/immunologie , Vaccins à virosomes/administration et posologie , Vaccins à virosomes/immunologie , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...