Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.094
Filtrer
1.
Hum Vaccin Immunother ; 20(1): 2383504, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-39118413

RÉSUMÉ

The Ad26.RSV.preF/RSV preF protein vaccine has previously demonstrated efficacyin protecting older adults against respiratory syncytial virus (RSV)-related lower respiratory tract disease in a phase 2b study. This study compared the immunogenicity of vaccine clinical trial material (CTM) representative of phase 2b clinical studies with CTM used in phase 3 clinical studies. A total of 248 adults aged 60-75 years, randomized in a 1:1 ratio, received one dose of either phase 3 CTM or phase 2b CTM. Solicited adverse events (AEs), unsolicited AEs, and serious AEs (SAEs) were assessed for 7-d, 28-d, and 6-month periods post-vaccination, respectively. RSV preF-ELISA antibody titers and RSV neutralizing titers were measured before and 14 d after vaccination. The phase 3 CTM-induced preF-ELISA response at Day 15, in terms of geometric mean titer, was shown to be non-inferior to that induced by phase 2b CTM. The RSV neutralizing antibody titers were also similar in the two groups at Day 15. The safety profile in terms of solicited AEs, unsolicited AEs, or SAEs was in general similar between the phase 3 CTM and phase 2b CTM groups, and solicited AEs were mostly mild to moderate in intensity. No related SAEs were reported, and no safety concerns were identified.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Vaccins contre les virus respiratoires syncytiaux , Humains , Adulte d'âge moyen , Sujet âgé , Mâle , Femelle , Anticorps antiviraux/sang , Anticorps neutralisants/sang , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/effets indésirables , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/immunologie , Immunogénicité des vaccins , Test ELISA , Virus respiratoire syncytial humain/immunologie , Effets secondaires indésirables des médicaments/épidémiologie
3.
MMWR Morb Mortal Wkly Rep ; 73(32): 696-702, 2024 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-39146277

RÉSUMÉ

Respiratory syncytial virus (RSV) is a major cause of respiratory illness and hospitalization in older adults during fall and winter in the United States. The 2023-2024 RSV season was the first during which RSV vaccination was recommended for U.S. adults aged ≥60 years, using shared clinical decision-making. On June 26, 2024, the Advisory Committee on Immunization Practices voted to update this recommendation as follows: a single dose of any Food and Drug Administration-approved RSV vaccine (Arexvy [GSK]; Abrysvo [Pfizer]; or mResvia [Moderna]) is now recommended for all adults aged ≥75 years and for adults aged 60-74 years who are at increased risk for severe RSV disease. Adults who have previously received RSV vaccine should not receive another dose. This report summarizes the evidence considered for these updated recommendations, including postlicensure data on vaccine effectiveness and safety, and provides clinical guidance for the use of RSV vaccines in adults aged ≥60 years. These updated recommendations are intended to maximize RSV vaccination coverage among persons most likely to benefit, by clarifying who is at highest risk and by reducing implementation barriers associated with the previous shared clinical decision-making recommendation. Continued postlicensure monitoring will guide future recommendations.


Sujet(s)
Comités consultatifs , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Humains , Sujet âgé , États-Unis , Infections à virus respiratoire syncytial/prévention et contrôle , Adulte d'âge moyen , Vaccins contre les virus respiratoires syncytiaux/administration et posologie ,
4.
Hum Vaccin Immunother ; 20(1): 2365804, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-39137331

RÉSUMÉ

Respiratory syncytial virus (RSV) is the main cause of low respiratory tract infections in infants under one year of age. In the 2023/2024 season, the monoclonal antibody nirsevimab was available to protect children from RSV, and Spain has become one of the first countries worldwide to implement this strategy. It is essential to evaluate the results of this first campaign and different characteristics of the immunized population in order to plan next campaigns, especially for countries that are going to include this immunization. Our coverage was high (91.5% for those born during the season and 88.3% globally). For those born during the season, only 4.9% preferred not to immunize at the maternity hospital, which meant an average delay of 27.45 days. We observed a lower coverage in the population of immigrant origin. There was a rapid pace of immunization, since for those born before the beginning of the campaign the mean to be immunized was 15.63 days, without differences between healthy and at-risk children. This allows immunization before the RSV season (90% of the catch-up children had been immunized on November 3). The average age at which all the immunized children have received nirsevimab was lower in healthy children compared to those with risk conditions (49.65 versus 232.85 days). For those born during the campaign, the average age was also lower in healthy children (3.14 versus 14.58 days). In conclusion, we consider that the implementation of the immunization strategy with nirsevimab in the Region of Murcia, Spain, has been a success.


Sujet(s)
Infections à virus respiratoire syncytial , Humains , Espagne , Infections à virus respiratoire syncytial/prévention et contrôle , Nourrisson , Femelle , Mâle , Programmes de vaccination , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/administration et posologie , Nouveau-né , Virus respiratoire syncytial humain/immunologie , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/administration et posologie
5.
Front Immunol ; 15: 1406929, 2024.
Article de Anglais | MEDLINE | ID: mdl-39114655

RÉSUMÉ

Numerous enveloped viruses, such as coronaviruses, influenza, and respiratory syncytial virus (RSV), utilize class I fusion proteins for cell entry. During this process, the proteins transition from a prefusion to a postfusion state, undergoing substantial and irreversible conformational changes. The prefusion conformation has repeatedly shown significant potential in vaccine development. However, the instability of this state poses challenges for its practical application in vaccines. While non-native disulfides have been effective in maintaining the prefusion structure, identifying stabilizing disulfide bonds remains an intricate task. Here, we present a general computational approach to systematically identify prefusion-stabilizing disulfides. Our method assesses the geometric constraints of disulfide bonds and introduces a ranking system to estimate their potential in stabilizing the prefusion conformation. We hypothesized that disulfides restricting the initial stages of the conformational switch could offer higher stability to the prefusion state than those preventing unfolding at a later stage. The implementation of our algorithm on the RSV F protein led to the discovery of prefusion-stabilizing disulfides that supported our hypothesis. Furthermore, the evaluation of our top design as a vaccine candidate in a cotton rat model demonstrated robust protection against RSV infection, highlighting the potential of our approach for vaccine development.


Sujet(s)
Disulfures , Protéines de fusion virale , Disulfures/composition chimique , Animaux , Protéines de fusion virale/immunologie , Protéines de fusion virale/composition chimique , Humains , Infections à virus respiratoire syncytial/immunologie , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/virologie , Stabilité protéique , Conception assistée par ordinateur , Conformation des protéines , Virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/immunologie , Rats , Modèles moléculaires
6.
Virology ; 598: 110194, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39096774

RÉSUMÉ

RSV infection remains a serious threat to the children all over the world, especially, in the low-middle income countries. Vaccine delivery via the mucosa holds great potential for inducing local immune responses in the respiratory tract. Previously, we reported the development of highly immunogenic RSV virus-like-particles (RSV-VLPs) based on the conformationally stable prefusogenic-F protein (preFg), glycoprotein and matrix protein. Here, to explore whether mucosal delivery of RSV-VLPs is an effective strategy to induce RSV-specific mucosal and systemic immunity, RSV-VLPs were administered via the nasal, sublingual and pulmonary routes to BALB/c mice. The results demonstrate that immunization with the VLPs via the mucosal routes induced minimal mucosal response and yet facilitated modest levels of serum IgG antibodies, enhanced T cell responses and the expression of the lung-homing marker CXCR3 on splenocytes. Immunization with VLPs via all three mucosal routes provided protection against RSV challenge with no signs of RSV induced pathology.


Sujet(s)
Anticorps antiviraux , Souris de lignée BALB C , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Vaccins à pseudo-particules virales , Protéines de fusion virale , Protéines de la matrice virale , Animaux , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/immunologie , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Souris , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Vaccins à pseudo-particules virales/immunologie , Vaccins à pseudo-particules virales/administration et posologie , Protéines de fusion virale/immunologie , Protéines de fusion virale/génétique , Protéines de fusion virale/administration et posologie , Femelle , Protéines de la matrice virale/immunologie , Protéines de la matrice virale/administration et posologie , Protéines de la matrice virale/génétique , Immunité muqueuse , Immunoglobuline G/sang , Immunoglobuline G/immunologie , Virus respiratoire syncytial humain/immunologie , Poumon/virologie , Poumon/immunologie , Glycoprotéines/immunologie , Glycoprotéines/administration et posologie , Administration par voie muqueuse , Virus respiratoires syncytiaux/immunologie , Lymphocytes T/immunologie
8.
JAMA Netw Open ; 7(7): e2419268, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38976271

RÉSUMÉ

Importance: A nonadjuvanted bivalent respiratory syncytial virus (RSV) prefusion F (RSVpreF [Pfizer]) protein subunit vaccine was newly approved and recommended for pregnant individuals at 32 0/7 to 36 6/7 weeks' gestation during the 2023 to 2024 RSV season; however, clinical vaccine data are lacking. Objective: To evaluate the association between prenatal RSV vaccination status and perinatal outcomes among patients who delivered during the vaccination season. Design, Setting, and Participants: This retrospective observational cohort study was conducted at 2 New York City hospitals within 1 health care system among patients who gave birth to singleton gestations at 32 weeks' gestation or later from September 22, 2023, to January 31, 2024. Exposure: Prenatal RSV vaccination with the RSVpreF vaccine captured from the health system's electronic health records. Main Outcome and Measures: The primary outcome is preterm birth (PTB), defined as less than 37 weeks' gestation. Secondary outcomes included hypertensive disorders of pregnancy (HDP), stillbirth, small-for-gestational age birth weight, neonatal intensive care unit (NICU) admission, neonatal respiratory distress with NICU admission, neonatal jaundice or hyperbilirubinemia, neonatal hypoglycemia, and neonatal sepsis. Logistic regression models were used to estimate odds ratios (ORs), and multivariable logistic regression models and time-dependent covariate Cox regression models were performed. Results: Of 2973 pregnant individuals (median [IQR] age, 34.9 [32.4-37.7] years), 1026 (34.5%) received prenatal RSVpreF vaccination. Fifteen patients inappropriately received the vaccine at 37 weeks' gestation or later and were included in the nonvaccinated group. During the study period, 60 patients who had evidence of prenatal vaccination (5.9%) experienced PTB vs 131 of those who did not (6.7%). Prenatal vaccination was not associated with an increased risk for PTB after adjusting for potential confounders (adjusted OR, 0.87; 95% CI, 0.62-1.20) and addressing immortal time bias (hazard ratio [HR], 0.93; 95% CI, 0.64-1.34). There were no significant differences in pregnancy and neonatal outcomes based on vaccination status in the logistic regression models, but an increased risk of HDP in the time-dependent model was seen (HR, 1.43; 95% CI, 1.16-1.77). Conclusions and Relevance: In this cohort study of pregnant individuals who delivered at 32 weeks' gestation or later, the RSVpreF vaccine was not associated with an increased risk of PTB and perinatal outcomes. These data support the safety of prenatal RSVpreF vaccination, but further investigation into the risk of HDP is warranted.


Sujet(s)
Naissance prématurée , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Humains , Femelle , Grossesse , Études rétrospectives , Adulte , Infections à virus respiratoire syncytial/prévention et contrôle , Nouveau-né , Vaccins contre les virus respiratoires syncytiaux/effets indésirables , New York (ville)/épidémiologie , Naissance prématurée/épidémiologie , Issue de la grossesse/épidémiologie , Complications infectieuses de la grossesse/prévention et contrôle , Vaccination/statistiques et données numériques , Mâle
9.
Front Immunol ; 15: 1376395, 2024.
Article de Anglais | MEDLINE | ID: mdl-38975350

RÉSUMÉ

Influenza A Virus (IAV) and Respiratory Syncytial Virus (RSV) are both responsible for millions of severe respiratory tract infections every year worldwide. Effective vaccines able to prevent transmission and severe disease, are important measures to reduce the burden for the global health system. Despite the strong systemic immune responses induced upon current parental immunizations, this vaccination strategy fails to promote a robust mucosal immune response. Here, we investigated the immunogenicity and efficacy of a mucosal adenoviral vector vaccine to tackle both pathogens simultaneously at their entry site. For this purpose, BALB/c mice were immunized intranasally with adenoviral vectors (Ad) encoding the influenza-derived proteins, hemagglutinin (HA) and nucleoprotein (NP), in combination with an Ad encoding for the RSV fusion (F) protein. The mucosal combinatory vaccine induced neutralizing antibodies as well as local IgA responses against both viruses. Moreover, the vaccine elicited pulmonary CD8+ and CD4+ tissue resident memory T cells (TRM) against the immunodominant epitopes of RSV-F and IAV-NP. Furthermore, the addition of Ad-TGFß or Ad-CCL17 as mucosal adjuvant enhanced the formation of functional CD8+ TRM responses against the conserved IAV-NP. Consequently, the combinatory vaccine not only provided protection against subsequent infections with RSV, but also against heterosubtypic challenges with pH1N1 or H3N2 strains. In conclusion, we present here a potent combinatory vaccine for mucosal applications, which provides protection against two of the most relevant respiratory viruses.


Sujet(s)
Anticorps antiviraux , Immunité muqueuse , Virus de la grippe A , Vaccins antigrippaux , Souris de lignée BALB C , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Animaux , Souris , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/immunologie , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Anticorps antiviraux/immunologie , Virus de la grippe A/immunologie , Femelle , Infections à Orthomyxoviridae/immunologie , Infections à Orthomyxoviridae/prévention et contrôle , Virus respiratoires syncytiaux/immunologie , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Vaccins combinés/immunologie , Vaccins combinés/administration et posologie , Humains , Adenoviridae/immunologie , Adenoviridae/génétique , Vecteurs génétiques
10.
J Pediatric Infect Dis Soc ; 13(Supplement_2): S125-S130, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38995087

RÉSUMÉ

This review article will summarize the vaccines and monoclonal antibodies currently under evaluation for the prevention of RSV disease in older infants, toddlers and young children. We will review the rationale for passive protection during the first months of life, and the role of active immunization afterwards, either with live attenuated, protein-based or mRNA vaccines.


Sujet(s)
Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Humains , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/immunologie , Nourrisson , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/usage thérapeutique , Enfant d'âge préscolaire , Immunisation passive , Anticorps monoclonaux/usage thérapeutique , Vaccins atténués/immunologie , Virus respiratoire syncytial humain/immunologie
12.
J Pediatric Infect Dis Soc ; 13(Supplement_2): S110-S114, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38995085

RÉSUMÉ

Newborns and young infants are at risk for severe respiratory syncytial virus (RSV) lower respiratory tract infection. Passive immunity is the mainstay of infection prevention in this cohort. Transplacental transfer of maternal antibodies provides the newborn with immediate protection from life-threatening infections, however, is dependent upon gestational age, birth weight, mother's age, recent maternal vaccination, maternal nutritional status, maternal immunocompetence and medical conditions, and placental integrity. Efficient transplacental transfer of RSV-neutralizing antibodies have led to the development and approval of maternal RSV immunization for the protection of the newborn. Additionally, administration of RSV-specific antibodies to infants leads to high serum titers of RSV-neutralizing antibodies and further protection from severe disease.


Sujet(s)
Anticorps antiviraux , Immunité acquise d'origine maternelle , Immunisation passive , Infections à virus respiratoire syncytial , Humains , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/immunologie , Immunisation passive/méthodes , Nouveau-né , Nourrisson , Femelle , Anticorps antiviraux/sang , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Virus respiratoire syncytial humain/immunologie , Grossesse , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/administration et posologie
13.
J Med Virol ; 96(7): e29308, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39007405

RÉSUMÉ

Respiratory syncytial virus (RSV) remains the primary cause of lower respiratory tract infections, particularly in infants and the elderly. In this study, we employed reverse genetics to generate a chimeric influenza virus expressing neuraminidase-3F protein conjugate with three repeats of the RSV F protein protective epitope inserted into the NA gene of A/California/7/2009 ca (CA/AA ca), resulting in rFlu/RSV/NA-3F (hereafter, rFRN3). The expression of NA-3F protein was confirmed by Western blotting. The morphology and temperature-sensitive phenotype of rFRN3 were similar to CA/AA ca. Its immunogenicity and protective efficiency were evaluated in BALB/c mice and cotton rats. Intranasal administration of rFRN3 elicited robust humoral, cellular, and to some extent, mucosal immune responses. Compared to controls, rFRN3 protected animals from RSV infection, attenuated lung injury, and reduced viral titers in the nose and lungs post-RSV challenge. These results demonstrate that rFRN3 can trigger RSV-specific immune responses and thus exhibits potent protective efficacy. The "dual vaccine" approach of a cold-adapted influenza vector RSV vaccine will improve the prophylaxis of influenza and RSV infection. rFRN3 thus warrants further clinical investigations as a candidate RSV vaccine.


Sujet(s)
Anticorps antiviraux , Vecteurs génétiques , Souris de lignée BALB C , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Sigmodontinae , Animaux , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/immunologie , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Vaccins contre les virus respiratoires syncytiaux/génétique , Femelle , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Souris , Vecteurs génétiques/génétique , Vecteurs génétiques/immunologie , Poumon/virologie , Poumon/immunologie , Poumon/anatomopathologie , Administration par voie nasale , Basse température , Vaccins synthétiques/immunologie , Vaccins synthétiques/administration et posologie , Vaccins synthétiques/génétique , Modèles animaux de maladie humaine , Protéines de fusion virale/immunologie , Protéines de fusion virale/génétique , Immunité muqueuse , , Charge virale
15.
Expert Rev Vaccines ; 23(1): 715-729, 2024.
Article de Anglais | MEDLINE | ID: mdl-39042099

RÉSUMÉ

INTRODUCTION: The use of novel adjuvants in human vaccines continues to expand as their contribution to preventing disease in challenging populations and caused by complex pathogens is increasingly understood. AS01 is a family of liposome-based vaccine Adjuvant Systems containing two immunostimulants: 3-O-desacyl-4'-monophosphoryl lipid A and the saponin QS-21. AS01-containing vaccines have been approved and administered to millions of individuals worldwide. AREAS COVERED: Here, we report advances in our understanding of the mode of action of AS01 that contributed to the development of efficacious vaccines preventing disease due to malaria, herpes zoster, and respiratory syncytial virus. AS01 induces early innate immune activation that induces T cell-mediated and antibody-mediated responses with optimized functional characteristics and induction of immune memory. AS01-containing vaccines appear relatively impervious to baseline immune status translating into high efficacy across populations. Currently licensed AS01-containing vaccines have shown acceptable safety profiles in clinical trials and post-marketing settings. EXPERT OPINION: Initial expectations that adjuvantation with AS01 could support effective vaccine responses and contribute to disease control have been realized. Investigation of the utility of AS01 in vaccines to prevent other challenging diseases, such as tuberculosis, is ongoing, together with efforts to fully define its mechanisms of action in different vaccine settings.


Adjuvants are added to vaccines to increase the immune response produced after vaccination. Adjuvant Systems contain two or more molecules that stimulate the immune system. AS01 is an Adjuvant System that contains two components, MPL and QS-21, that stimulate the immune system. AS01 is included in three approved vaccines: a malaria vaccine for children, a herpes zoster vaccine for older adults, and a respiratory syncytial virus vaccine also for older adults. Vaccines containing AS01 have been extensively evaluated in clinical trials and administered to millions of individuals during market use. These vaccines are effective in preventing disease and have acceptable safety in different age groups. Experiments have been done to investigate how AS01 works in vaccines to produce an efficient immune response that helps to protect against the disease being targeted. A key effect of AS01 is to encourage specific immune cells to produce chemicals that stimulate the immune system. We now know that this effect is due to co-operation between MPL and QS-21. Experiments have shown that AS01 induces a sophisticated immune 'gene signature' in blood within 24 h after vaccination, and people who developed this 'gene signature' had a stronger response to vaccination. AS01 seems to be able to stimulate the immune system of most people ­ even if they are older or have a weakened immune system. This means that AS01 could be included in other vaccines against other challenging diseases, such as tuberculosis, or could be used in the treatment of some disease, such as chronic hepatitis B.


Sujet(s)
Adjuvants immunologiques , Adjuvants vaccinaux , Saponines , Humains , Saponines/immunologie , Saponines/pharmacologie , Adjuvants immunologiques/administration et posologie , Adjuvants immunologiques/pharmacologie , Lipide A/analogues et dérivés , Lipide A/immunologie , Lipide A/pharmacologie , Animaux , Immunité innée/effets des médicaments et des substances chimiques , Vaccins contre les virus respiratoires syncytiaux/immunologie , Liposomes , Paludisme/prévention et contrôle , Paludisme/immunologie , Vaccins contre le paludisme/immunologie , Vaccins contre le paludisme/administration et posologie , Association médicamenteuse
16.
J Infect Public Health ; 17(8): 102492, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-39002465

RÉSUMÉ

BACKGROUND: A new monoclonal antibody (nirsevimab; Beyfortus®) and a bivalent prefusion RSV vaccine (Abrysvo®) for maternal immunization have been approved recently. This is a modelling study to estimate the potential impact of different immunization programs with these products on RSV-bronchiolitis. METHODS: Population-based real-world data from primary care and hospitalizations were considered. RSV bronchiolitis dynamics in absence of these immunization scenarios were explained by a multivariate age-structured Bayesian model. Then, the potential impact was simulated under different assumptions including the most recent clinical trial data. Differences in endpoints, populations, and timeframes between trials make the two products' efficacy difficult to compare. RESULTS: A seasonal with catch-up program, assuming a constant effectiveness of 79.5 % during the first 5 months followed by a linear decay to 0 by month 10 with nirsevimab, would prevent between 5121 and 8846 RSV bronchiolitis per 100,000 infants-years. Assuming 77.3 % effectiveness with the same decay, between 976 and 1686 RSV-hospitalizations per 100,000 infants-years could be prevented depending on the uptake. A year-round maternal immunization program, with 51 % of effectiveness during the first 6 months followed by a linear decay to 0 by month 10 would prevent between 3246 and 5606 RSV bronchiolitis cases per 100,000 infants-years. Assuming 56.9 % effectiveness with the same decay, between 713 and 1231 RSV-hospitalizations per 100,000 infants-years could be prevented. CONCLUSIONS: Our results suggest that each strategy would effectively reduce RSV-bronchiolitis.


Sujet(s)
Hospitalisation , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Humains , Infections à virus respiratoire syncytial/prévention et contrôle , Nourrisson , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Femelle , Hospitalisation/statistiques et données numériques , Mâle , Bronchiolite/prévention et contrôle , Bronchiolite virale/prévention et contrôle , Anticorps monoclonaux humanisés/usage thérapeutique , Nouveau-né , Programmes de vaccination , Anticorps monoclonaux/usage thérapeutique
17.
J Infect Dis ; 230(1): e102-e110, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-39052726

RÉSUMÉ

BACKGROUND: The recently approved AS01E-adjuvanted respiratory syncytial virus (RSV) prefusion F protein-based vaccine for older adults (RSVPreF3 OA) demonstrated high efficacy against RSV-related disease in ≥60-year-olds. METHODS: This ongoing phase 3 study in ≥60-year-olds evaluates immune persistence until 3 years after RSVPreF3 OA vaccination. Here, we describe interim results on humoral and cell-mediated immunogenicity, reactogenicity, and safety until 1 year post-dose 1. RESULTS: In total, 1653 participants were vaccinated. One month post-dose 1, neutralization titers increased 10.5-fold (RSV-A) and 7.8-fold (RSV-B) vs pre-dose 1. Titers then declined to levels 4.4-fold (RSV-A) and 3.5-fold (RSV-B) above pre-dose 1 at month 6 and remained 3.1-fold (RSV-A) and 2.3-fold (RSV-B) above pre-dose 1 levels after 1 year. RSVPreF3-binding immunoglobulin G levels and CD4+ T-cell frequencies showed similar kinetics. Solicited administration-site and systemic adverse events (mostly mild to moderate and transient) were reported by 62.2% and 49.5% of participants. Serious adverse events were reported by 3.9% of participants within 6 months post-dose 1; 1 case was considered vaccine related. CONCLUSIONS: One RSVPreF3 OA dose elicited cell-mediated and RSV-A- and RSV-B-specific humoral immune responses that declined over time but remained above pre-dose 1 levels for at least 1 year. The vaccine was well tolerated with an acceptable safety profile. Clinical Trials Registration. NCT04732871 (ClinicalTrials.gov).


Respiratory syncytial virus (RSV) is a major cause of illness and hospitalization in older adults. An RSV vaccine for older adults developed by GSK was recently approved. The vaccine was well tolerated and provided protection against RSV disease in adults aged ≥60 years during at least 1 RSV season. In this ongoing study, we are evaluating the magnitude and durability of the immune response, as well as vaccine safety, until 3 years after vaccination of adults aged ≥60 years from 5 countries. Here, we report the results of an interim analysis until 1 year after vaccination with 1 dose. In total, 1653 participants were vaccinated. We found that the vaccine induced a strong immune response that was evident 1 month after vaccination, after which it declined but persisted for at least 1 year. Study participants most often reported pain at the injection site, muscle pain, tiredness, and headache as adverse reactions, which were mostly mild to moderate and of short duration. One serious adverse reaction was considered related to the vaccine. The long-term immune response that was observed in this study is consistent with the vaccine providing protection during at least 1 RSV season.


Sujet(s)
Anticorps antiviraux , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Virus respiratoire syncytial humain , Humains , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Vaccins contre les virus respiratoires syncytiaux/effets indésirables , Mâle , Femelle , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/immunologie , Anticorps antiviraux/sang , Sujet âgé , Adulte d'âge moyen , Virus respiratoire syncytial humain/immunologie , Protéines de fusion virale/immunologie , Protéines de fusion virale/administration et posologie , Anticorps neutralisants/sang , Immunogénicité des vaccins , Sujet âgé de 80 ans ou plus , Adjuvants vaccinaux/administration et posologie
20.
Antiviral Res ; 229: 105960, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38986872

RÉSUMÉ

Respiratory syncytial virus is the major cause of respiratory viral infections, particularly in infants, immunocompromised populations, and the elderly (over 65 years old), the prevention of RSV infection has become a priority. In this study, we generated a chimeric influenza virus, termed LAIV/RSV/HA-3F, using reverse genetics technology which contained three repeats of the RSV fusion protein neutralizing epitope site II to the N terminal in the background of the hemagglutinin (HA) gene of cold adapted influenza vaccine A/California/7/2009 ca. LAIV/RSV/HA-3F exhibited cold-adapted (ca) and attenuated (att) phenotype. BALB/c mice immunized intranasally with LAIV/RSV/HA-3F showed robust immunogenicity, inducing viral-specific antibody responses against both influenza and RSV, eliciting RSV-specific humoral, cellular and mucosal immune responses. LAIV/RSV/HA-3F also conferred protection as indicated by reduced viral titers and improved lung histopathological alterations against live RSV virus challenge. Mechanismly, single-cell RNA sequencing (scRNA-seq) and single-cell T cell antigen receptor (TCR) sequencing were employed to characterize the immune responses triggered by chimeric RSV vaccine, displaying that LAIV/RSV/HA-3F provided protection mainly via interferon-γ (IFN-γ). Moreover, we found that LAIV/RSV/HA-3F significantly inhibited viral replication in the challenged lung and protected against subsequent RSV challenge in cotton rats without causing lung disease. Taken together, our findings demonstrated that LAIV/RSV/HA-3F has potential as a promising bivalent vaccine with dual purpose candidate for the prevention of influenza and RSV, and preclinical and clinical studies warrant further investigations.


Sujet(s)
Anticorps antiviraux , Épitopes , Vaccins antigrippaux , Souris de lignée BALB C , Infections à virus respiratoire syncytial , Sigmodontinae , Protéines de fusion virale , Animaux , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/immunologie , Vaccins antigrippaux/immunologie , Vaccins antigrippaux/administration et posologie , Vaccins antigrippaux/génétique , Souris , Protéines de fusion virale/immunologie , Protéines de fusion virale/génétique , Épitopes/immunologie , Épitopes/génétique , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Femelle , Virus respiratoires syncytiaux/immunologie , Virus respiratoires syncytiaux/génétique , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Poumon/virologie , Poumon/immunologie , Poumon/anatomopathologie , Humains , Basse température , Anticorps neutralisants/immunologie , Anticorps neutralisants/sang , Virus respiratoire syncytial humain/immunologie , Virus respiratoire syncytial humain/génétique , Infections à Orthomyxoviridae/prévention et contrôle , Infections à Orthomyxoviridae/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE