Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 314
Filtrer
1.
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
2.
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
4.
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
5.
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
6.
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
8.
Viruses ; 16(6)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38932136

RÉSUMÉ

Respiratory syncytial virus (RSV) is a major cause of severe respiratory tract disease worldwide, and a pediatric vaccine is not available. We generated a filamentous RSV-based virus-like particle (VLP) that presents the central conserved region of the attachment protein G. This was achieved by co-expressing the matrix protein, phosphoprotein, nucleoprotein, and a hybrid fusion protein in which the F ectodomain was replaced with the G central region (GCR). The latter is relatively conserved and contains a receptor binding site and hence is a logical vaccine target. The immunogenicity and efficacy of the resulting VLP, termed VLP-GCR, were examined in mice using intranasal application without adjuvant. VLP-GCR induced substantial anti-N antibody levels but very low anti-G antibody levels, even after three vaccinations. In contrast, a VLP presenting prefusion-stabilized fusion (preF) protein instead of GCR induced both high anti-F and anti-nucleoprotein antibody levels, suggesting that our GCR antigen was poorly immunogenic. Challenge of VLP-GCR-vaccinated mice caused increased weight loss and lung pathology, and both VLPs induced mucus in the lungs. Thus, neither VLP is suitable as a vaccine for RSV-naive individuals. However, VLP-preF enhanced the proportion of preF antibodies and could serve as a multi-antigen mucosal booster vaccine in the RSV-experienced population.


Sujet(s)
Anticorps antiviraux , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Vaccins à pseudo-particules virales , Animaux , Femelle , Humains , Souris , Administration par voie nasale , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Poumon/virologie , Poumon/anatomopathologie , Poumon/immunologie , Souris de lignée BALB C , 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 , Virus respiratoire syncytial humain/immunologie , Virus respiratoire syncytial humain/génétique , Vaccination , Vaccins à pseudo-particules virales/immunologie , Vaccins à pseudo-particules virales/administration et posologie , Vaccins à pseudo-particules virales/génétique , Protéines de l'enveloppe virale/immunologie , Protéines de l'enveloppe virale/génétique , Protéines de l'enveloppe virale/administration et posologie , Protéines de fusion virale/immunologie , Protéines de fusion virale/génétique , Perte de poids
9.
J Biosci Bioeng ; 138(2): 127-136, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38851988

RÉSUMÉ

Respiratory syncytial virus (RSV) infection is an acute respiratory infection caused by RSV. It occurs worldwide, and for over 50 years, several attempts have been made to research and develop vaccines to prevent RSV infection; effective preventive vaccines are eagerly awaited. The RSV fusion (F) protein, which has gained attention as a vaccine antigen, causes a dynamic structural change from the preF to postF state. Therefore, the structural changes in proteins must be regulated to produce a vaccine antigen that can efficiently induce antibodies with high virus-neutralizing activity. We successfully discovered several mutations that stabilized the antigen site Ø in the preF state, trimerized it, and improved the level of protein expression through observation and computational analysis of the RSV-F protein structure and amino acid mutation analysis of RSV strains. The four RSV-F protein mutants that resulted from the combination of these effective mutations stably conserved a wide range of preF- and trimeric preF-specific epitopes with high virus-neutralizing activity. Absorption assay using human serum revealed that mutants constructed bound to antibodies with virus-neutralizing activity that were induced by natural RSV infection, whereas they hardly bound to anti-postF antibodies without virus-neutralizing activity. Furthermore, mouse immunization demonstrated that our constructed mutants induced a high percentage of antibodies that bind to the preF-specific antigen site. These characteristics suggest that the mutants constructed can be superior vaccine antigens from the viewpoint of RSV infection prevention effect and safety.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Antigènes viraux , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Protéines de fusion virale , Animaux , Humains , Protéines de fusion virale/immunologie , Protéines de fusion virale/génétique , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/génétique , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Souris , Anticorps antiviraux/immunologie , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/immunologie , Anticorps neutralisants/immunologie , Antigènes viraux/immunologie , Antigènes viraux/génétique , Mutation , Épitopes/immunologie , Épitopes/génétique , Virus respiratoire syncytial humain/immunologie , Virus respiratoire syncytial humain/génétique , Souris de lignée BALB C , Virus respiratoires syncytiaux/immunologie , Virus respiratoires syncytiaux/génétique
10.
Hum Vaccin Immunother ; 20(1): 2358566, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38847198

RÉSUMÉ

A maternal vaccine and long-acting monoclonal antibody (mAb) were recently approved to protect infants against respiratory syncytial virus (RSV). We identified subgroups of pregnant people with different preferences for RSV preventives and respondent characteristics associated with subgroup membership. An online survey, including a discrete choice experiment (DCE), was conducted among US pregnant people. RSV preventive attributes included effectiveness, duration of protection during RSV season, injection recipient/timing, preventive type (vaccine or mAb), and type of visit required to receive injection. In DCE choice tasks, pregnant people selected between two hypothetical preventive profiles with varying attribute-levels and a no-preventive option. Logistic regression, including latent class analysis (LCA), was used to analyze the data. Of 992 pregnant people (mean age: 30.0 years), 60.3% were expecting their second/later birth. LCA identified three preference subgroups: 'Effectiveness' (preventive choice mostly driven by increases in effectiveness; 51.4% class membership probability), 'Season' (preventive choice mostly driven by improvement in duration of protection during the RSV season; 39.2% class membership probability), and 'No Preventive' (frequently chose no-preventive option; 9.4% class membership probability). 'Effectiveness' and 'Season' preferred maternal vaccine over mAb; mAb was preferred by 'No Preventive.' Perceiving RSV as serious for infants, higher health literacy, and lower household income were associated with 'Effectiveness.' Perceiving RSV as serious for pregnant people was associated with 'Season.' Perceiving RSV to not be serious for pregnant people and not being employed were associated with 'No Preventive.' Subgroups of pregnant people vary in preferences for RSV preventives. Most pregnant people preferred a maternal vaccine, although some may be more willing to accept alternative preventive options.


Sujet(s)
Analyse de structure latente , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Virus respiratoire syncytial humain , Humains , Femelle , Grossesse , Infections à virus respiratoire syncytial/prévention et contrôle , États-Unis , Adulte , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Jeune adulte , Virus respiratoire syncytial humain/immunologie , Nourrisson , Enquêtes et questionnaires , Préférence des patients/statistiques et données numériques , Vaccination/statistiques et données numériques , Femmes enceintes/psychologie , Anticorps monoclonaux/usage thérapeutique , Adolescent
11.
J Med Econ ; 27(1): 887-896, 2024.
Article de Anglais | MEDLINE | ID: mdl-38896438

RÉSUMÉ

AIM: The current study compared preparation time, errors, satisfaction, and preference for a prefilled syringe (PFS) versus two RSV vaccines requiring reconstitution (VRR1 and VRR2) in a randomized, single-blinded time and motion study. METHODS: Pharmacists, nurses, and pharmacy technicians were randomized to a preparation sequence of the three vaccines. Participants read instructions, then consecutively prepared the three vaccines with a 3-5-min washout period in between. Preparations were video recorded and reviewed by a trained pharmacist for preparation time and errors using predefined, vaccine-specific checklists. Participant demographics, satisfaction with vaccine preparation, and vaccine preference were recorded. Within-subjects analysis of variance was used to compare preparation time. Mixed-effects Poisson and ordered logistic regression models were used to compare the number of preparation errors and satisfaction scores, respectively. RESULTS: Sixty-three pharmacists (60%), nurses (35%), and pharmacy technicians (5%) participated at four sites in the United States. The least squares mean preparation time per dose for PFS was 141.8 s (95% CI = 156.8-126.7; p <.0001) faster than for VRR1, 103.6 s (95% CI = 118.7-88.5; p <.0001) faster than for VRR2, and 122.7 s (95% CI = 134.2-111.2; p <.0001) faster than the pooled VRRs. Overall satisfaction (combined "Very" and "Extremely") was 87.3% for PFS, 28.6% for VRR1, and 47.6% for VRR2. Most participants (81.0%) preferred the PFS vaccine. LIMITATIONS: The study is limited by the inability to completely blind observers. To minimize the effects of order, we utilized a 3-sequence block design; however, the order in which the vaccines were prepared may have affected outcomes. Participants were assessed once, whereas if repeated preparations were performed there may have been trained efficiencies gained for each vaccine. CONCLUSION: PFS vaccines can greatly simplify the vaccine preparation process, allowing administrators to prepare almost four times more doses per hour than with vial and syringe systems.


Sujet(s)
Seringues , Études ergonomiques , Humains , Femelle , Mâle , Adulte , Adulte d'âge moyen , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Méthode en simple aveugle , Facteurs temps , Pharmaciens , Techniciens en pharmacie , Préparation de médicament , Infirmières et infirmiers , États-Unis
13.
Vaccine ; 42(20): 126094, 2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-38944576

RÉSUMÉ

Maternal immunisation is a powerful tool to protect both pregnant women and their children. A new maternal RSV vaccine holds promise to protect newborns from RSV-associated illness in the first few months of life, but no official recommendation has been made in Germany. Since RSV causes a significant burden of paediatric hospital admissions, we consider it a pertinent opportunity to review barriers to maternal vaccination in Germany, which might also apply to other settings. Access to vaccination for pregnant women in Germany is shaped by an interplay of legal, regulatory, institutional, and sociocultural factors, with a less permissive clinical research environment, delays in recommendation and roll-out, and lower acceptance by healthcare professionals and the population. Actionable recommendations to improve availability and uptake include coordination with other national regulatory bodies to reduce delays, awareness and literacy campaigns for health professionals and the general public, and capacity building for vaccine clinical research.


Sujet(s)
Femmes enceintes , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Vaccination , Humains , Femelle , Grossesse , Allemagne , Infections à virus respiratoire syncytial/prévention et contrôle , Vaccination/statistiques et données numériques , Vaccination/méthodes , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Complications infectieuses de la grossesse/prévention et contrôle , Nouveau-né , Développement de vaccin
14.
Hum Vaccin Immunother ; 20(1): 2341505, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38723786

RÉSUMÉ

Respiratory Syncytial Virus poses a significant global public health threat, particularly affecting infants aged less than one year of age. Recently, two forms of passive immunization against infant RSV have been developed and brought to market; nirsevimab a long-acting monoclonal antibody (mAb) and RSV-PreF, a maternal RSV vaccine. The acceptability and uptake of these products will play a pivotal role in determining the success of any national immunization strategy aimed at safeguarding infants from RSV. It is crucial at this time to reflect on the factors that influence parental decisions surrounding immunization to facilitate more informed discussions, enhance healthcare communication, and contribute to the design of effective RSV prevention strategies that resonate with the concerns and aspirations of parents worldwide.


Sujet(s)
Anticorps monoclonaux , Vaccins contre les virus respiratoires syncytiaux , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Vaccins contre les virus respiratoires syncytiaux/effets indésirables , Vaccins contre les virus respiratoires syncytiaux/immunologie , Anticorps monoclonaux/administration et posologie , Anticorps monoclonaux/immunologie , Humains , Femelle , Grossesse , Nouveau-né , Nourrisson , Guides de bonnes pratiques cliniques comme sujet
16.
Cochrane Database Syst Rev ; 5: CD015134, 2024 05 02.
Article de Anglais | MEDLINE | ID: mdl-38695784

RÉSUMÉ

BACKGROUND: Respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections (LRTIs) in infants. Maternal RSV vaccination is a preventive strategy of great interest, as it could have a substantial impact on infant RSV disease burden. In recent years, the clinical development of maternal RSV vaccines has advanced rapidly. OBJECTIVES: To assess the efficacy and safety of maternal respiratory syncytial virus (RSV) vaccination for preventing RSV disease in infants. SEARCH METHODS: We searched Cochrane Pregnancy and Childbirth's Trials Register and two other trials registries on 21 October 2022. We updated the search on 27 July 2023, when we searched MEDLINE, Embase, CENTRAL, CINAHL, and two trials registries. Additionally, we searched the reference lists of retrieved studies and conference proceedings. There were no language restrictions on our searches. SELECTION CRITERIA: We included randomised controlled trials (RCTs) comparing maternal RSV vaccination with placebo or no intervention in pregnant women of any age. The primary outcomes were hospitalisation with clinically confirmed or laboratory-confirmed RSV disease in infants. The secondary outcomes covered adverse pregnancy outcomes (intrauterine growth restriction, stillbirth, and maternal death) and adverse infant outcomes (preterm birth, congenital abnormalities, and infant death). DATA COLLECTION AND ANALYSIS: We used standard Cochrane methods and assessed the certainty of evidence using the GRADE approach. MAIN RESULTS: We included six RCTs (25 study reports) involving 17,991 pregnant women. The intervention was an RSV pre-F protein vaccine in four studies, and an RSV F protein nanoparticle vaccine in two studies. In all studies, the comparator was a placebo (saline, formulation buffer, or sterile water). We judged four studies at overall low risk of bias and two studies at overall high risk (mainly due to selection bias). All studies were funded by pharmaceutical companies. Maternal RSV vaccination compared with placebo reduces infant hospitalisation with laboratory-confirmed RSV disease (risk ratio (RR) 0.50, 95% confidence interval (CI) 0.31 to 0.82; 4 RCTs, 12,216 infants; high-certainty evidence). Based on an absolute risk with placebo of 22 hospitalisations per 1000 infants, our results represent 11 fewer hospitalisations per 1000 infants from vaccinated pregnant women (15 fewer to 4 fewer). No studies reported infant hospitalisation with clinically confirmed RSV disease. Maternal RSV vaccination compared with placebo has little or no effect on the risk of congenital abnormalities (RR 0.96, 95% CI 0.88 to 1.04; 140 per 1000 with placebo, 5 fewer per 1000 with RSV vaccination (17 fewer to 6 more); 4 RCTs, 12,304 infants; high-certainty evidence). Maternal RSV vaccination likely has little or no effect on the risk of intrauterine growth restriction (RR 1.32, 95% CI 0.75 to 2.33; 3 per 1000 with placebo, 1 more per 1000 with RSV vaccination (1 fewer to 4 more); 4 RCTs, 12,545 pregnant women; moderate-certainty evidence). Maternal RSV vaccination may have little or no effect on the risk of stillbirth (RR 0.81, 95% CI 0.38 to 1.72; 3 per 1000 with placebo, no difference with RSV vaccination (2 fewer to 3 more); 5 RCTs, 12,652 pregnant women). There may be a safety signal warranting further investigation related to preterm birth. This outcome may be more likely with maternal RSV vaccination, although the 95% CI includes no effect, and the evidence is very uncertain (RR 1.16, 95% CI 0.99 to 1.36; 6 RCTs, 17,560 infants; very low-certainty evidence). Based on an absolute risk of 51 preterm births per 1000 infants from pregnant women who received placebo, there may be 8 more per 1000 infants from pregnant women with RSV vaccination (1 fewer to 18 more). There was one maternal death in the RSV vaccination group and none in the placebo group. Our meta-analysis suggests that RSV vaccination compared with placebo may have little or no effect on the risk of maternal death (RR 3.00, 95% CI 0.12 to 73.50; 3 RCTs, 7977 pregnant women; low-certainty evidence). The effect of maternal RSV vaccination on the risk of infant death is very uncertain (RR 0.81, 95% CI 0.36 to 1.81; 6 RCTs, 17,589 infants; very low-certainty evidence). AUTHORS' CONCLUSIONS: The findings of this review suggest that maternal RSV vaccination reduces laboratory-confirmed RSV hospitalisations in infants. There are no safety concerns about intrauterine growth restriction and congenital abnormalities. We must be careful in drawing conclusions about other safety outcomes owing to the low and very low certainty of the evidence. The evidence available to date suggests RSV vaccination may have little or no effect on stillbirth, maternal death, and infant death (although the evidence for infant death is very uncertain). However, there may be a safety signal warranting further investigation related to preterm birth. This is driven by data from one trial, which is not fully published yet. The evidence base would be much improved by more RCTs with substantial sample sizes and well-designed observational studies with long-term follow-up for assessment of safety outcomes. Future studies should aim to use standard outcome measures, collect data on concomitant vaccines, and stratify data by timing of vaccination, gestational age at birth, race, and geographical setting.


Sujet(s)
Essais contrôlés randomisés comme sujet , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Mortinatalité , Humains , Grossesse , Femelle , Infections à virus respiratoire syncytial/prévention et contrôle , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Vaccins contre les virus respiratoires syncytiaux/usage thérapeutique , Vaccins contre les virus respiratoires syncytiaux/effets indésirables , Nourrisson , Nouveau-né , Mortinatalité/épidémiologie , Naissance prématurée/prévention et contrôle , Naissance prématurée/épidémiologie , Complications infectieuses de la grossesse/prévention et contrôle , Hospitalisation/statistiques et données numériques , Retard de croissance intra-utérin/prévention et contrôle , Issue de la grossesse , Vaccination , Malformations/prévention et contrôle , Biais (épidémiologie) , Mort infantile/prévention et contrôle
17.
Front Immunol ; 15: 1377374, 2024.
Article de Anglais | MEDLINE | ID: mdl-38745662

RÉSUMÉ

Respiratory syncytial virus (RSV) is the primary cause of bronchiolitis-related hospitalizations among children under 5 years of age, with reinfection being common throughout life. Maternal vaccination has emerged as a promising strategy, delivering elevated antibody levels to newborns for immediate protection. However, limited research has explored the protective efficacy of maternal antibodies (matAbs) against secondary RSV infections in offspring. To address this gap, we employed a mouse model of maternal RSV vaccination and secondary infection of offspring to evaluate lung pathology following RSV reinfection in mice with varying levels of maternal antibody (matAb). Additionally, we aimed to investigate the potential causes of exacerbated lung inflammation in offspring with high matAb levels following secondary RSV exposure. Our findings revealed that offspring with elevated levels of maternal pre-F antibody demonstrated effective protection against lung pathology following the initial RSV infection. However, this protection was compromised upon reinfection, manifesting as heightened weight loss, exacerbated lung pathology, increased expression of RSV-A N genes, eosinophilia, enhanced IL-5, IL-13, MUC5AC, and eosinophils Major Basic Protein (MBP) production in lung tissue compared to offspring lacking matAbs. Importantly, these unexpected outcomes were not attributed to antibody-dependent enhancement (ADE) resulting from declining matAb levels over time. Notably, our findings showed a decline in secretory IgA (sIgA), mucosal IgA, and mucosal IgG levels in offspring with high matAb levels post-primary RSV challenge. We propose that this decline may be a critical factor contributing to the ineffective protection observed during secondary RSV exposure. Overall, these findings offer valuable insights into maternal vaccination against RSV, contributing to a comprehensive understanding and mitigation of potential risks associated with maternal RSV vaccination.


Sujet(s)
Anticorps antiviraux , Pneumopathie infectieuse , Infections à virus respiratoire syncytial , Animaux , Infections à virus respiratoire syncytial/immunologie , Souris , Femelle , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Pneumopathie infectieuse/immunologie , Immunité acquise d'origine maternelle , Poumon/immunologie , Poumon/virologie , Poumon/anatomopathologie , Grossesse , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/effets indésirables , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Modèles animaux de maladie humaine , Virus respiratoires syncytiaux/immunologie , Souris de lignée BALB C
19.
Immun Inflamm Dis ; 12(4): e1257, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38661110

RÉSUMÉ

BACKGROUND: Respiratory syncytial virus (RSV) is the world's leading cause of viral acute lower respiratory infections (ALRI) in infants. WHO has identified maternal RSV vaccination a priority and candidate vaccines are in development; however, vaccine hesitancy remains an impediment to successful implementation of maternal immunization. This study, the largest antenatal survey conducted to-date, aimed to examine maternal RSV awareness, likely acceptance of RSV vaccination in pregnancy, and attitudes to maternal vaccination. METHODS: Pregnant women of all gestations attending antenatal clinic of a university maternity hospital in Ireland were invited to participate. An information leaflet provided, consent obtained, and survey administered examining RSV awareness, willingness to avail of antenatal RSV vaccination, factors influencing acceptability and preferred sources of assistance. Research Ethics Committee (REC) approval obtained, and general data protection regulation (GDPR) guidelines followed. RESULTS: 528 women completed the survey. A large proportion (75.6%) had never heard of RSV, yet 48.5% would still avail of a vaccine, 45.8% were undecided and only 5.3% would not. The main factor making vaccination acceptable to women (76.4%) was that it protects their infant from illness (p < .001, CV 0.336 for association with acceptance) and general practitioner (GP) was the preferred guidance source in decision-making (57.7%). CONCLUSIONS: Despite low levels of maternal awareness of RSV, pregnant women in Ireland are open to availing of antenatal vaccination. Maternal immunization strategies need to focus on infant's protection from RSV-associated ALRI along with vaccine safety, and build on an interdisciplinary collaboration of maternal, neonatal, primary care and public health services.


Sujet(s)
Connaissances, attitudes et pratiques en santé , Acceptation des soins par les patients , Infections à virus respiratoire syncytial , Vaccins contre les virus respiratoires syncytiaux , Vaccination , Humains , Femelle , Irlande/épidémiologie , Grossesse , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/psychologie , Infections à virus respiratoire syncytial/immunologie , Adulte , Vaccins contre les virus respiratoires syncytiaux/immunologie , Vaccins contre les virus respiratoires syncytiaux/administration et posologie , Vaccination/psychologie , Acceptation des soins par les patients/psychologie , Acceptation des soins par les patients/statistiques et données numériques , Complications infectieuses de la grossesse/prévention et contrôle , Complications infectieuses de la grossesse/psychologie , Complications infectieuses de la grossesse/immunologie , Enquêtes et questionnaires , Jeune adulte , Réticence à l'égard de la vaccination/psychologie , Réticence à l'égard de la vaccination/statistiques et données numériques , Femmes enceintes/psychologie , Virus respiratoire syncytial humain/immunologie , Adolescent
20.
Vaccine ; 42(13): 3172-3179, 2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38616438

RÉSUMÉ

BACKGROUND: Bivalent RSV prefusion F subunit vaccine (RSVpreF), comprised of equal quantities of stabilized prefusion F antigens from the major circulating subgroups (RSV A, RSV B), is licensed for prevention of RSV-associated lower respiratory tract illness (LRTI) in older adults and for maternal vaccination for prevention of RSV-associated LRTI in infants. To support licensure and large-scale manufacturing, this lot consistency study was conducted to demonstrate equivalence in immunogenicity across 3 RSVpreF lots. METHODS: This phase 3, multicenter, parallel-group, placebo-controlled, randomized (1:1:1:1), double-blind study evaluated immunogenicity, safety, and tolerability of RSVpreF in healthy 18-49-year-old adults. Participants received a single 120-µg injection of 1 of 3RSVpreF lots or placebo. Geometric mean ratio (GMR) of RSV serum 50 % neutralizing geometric mean titers obtained 1 month after vaccination were compared between each vaccine lot for RSV A and RSV B, separately. Equivalence between lots was defined using a 1.5-fold criterion (GMR 95 % CIs for every lot pair within the 0.667-1.5 interval). Safety and tolerability were assessed. RESULTS: Of 992participants vaccinated, 948 were included in the evaluable immunogenicity population. All 3 RSVpreF lots elicited strong immune responses, meeting the 1.5-fold equivalence criterion for all between-lot comparisons for both RSV A and RSV B. Across the 3 lots, RSV A and RSV B 50 % neutralizing geometric mean titers substantially increased from baseline (RSV A, 1671-1795; RSV B 1358-1429) to 1 month after RSVpreF vaccination (RSV A, 24,131-25,238; RSV B, 19,238-21,702), corresponding to ≥14-fold increases in 50 % neutralizing titers for both RSV A and RSV B from before to 1 month after vaccination. Single doses of RSVpreF were safe and well tolerated, with similar safety profiles across the 3 RSVpreF lots. CONCLUSIONS: These findings support the reproducibility of RSVpreF vaccine manufacturing with similar safety and reactogenicity profiles (NCT05096208).


Sujet(s)
Anticorps antiviraux , Immunogénicité des vaccins , 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 , Femelle , Mâle , Adulte , Méthode en double aveugle , Jeune adulte , Adolescent , Anticorps antiviraux/sang , Adulte d'âge moyen , Infections à virus respiratoire syncytial/prévention et contrôle , Infections à virus respiratoire syncytial/immunologie , Virus respiratoire syncytial humain/immunologie , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Vaccins sous-unitaires/immunologie , Vaccins sous-unitaires/administration et posologie , Vaccins sous-unitaires/effets indésirables , Volontaires sains , Vaccination/méthodes , Protéines de fusion virale/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE