Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 203
Filter
2.
JAMA ; 331(11): 938-950, 2024 03 19.
Article in English | MEDLINE | ID: mdl-38502075

ABSTRACT

Importance: In January 2023, the US Centers for Disease Control and Prevention and the US Food and Drug Administration noted a safety concern for ischemic stroke among adults aged 65 years or older who received the Pfizer-BioNTech BNT162b2; WT/OMI BA.4/BA.5 COVID-19 bivalent vaccine. Objective: To evaluate stroke risk after administration of (1) either brand of the COVID-19 bivalent vaccine, (2) either brand of the COVID-19 bivalent plus a high-dose or adjuvanted influenza vaccine on the same day (concomitant administration), and (3) a high-dose or adjuvanted influenza vaccine. Design, Setting, and Participants: Self-controlled case series including 11 001 Medicare beneficiaries aged 65 years or older who experienced stroke after receiving either brand of the COVID-19 bivalent vaccine (among 5 397 278 vaccinated individuals). The study period was August 31, 2022, through February 4, 2023. Exposures: Receipt of (1) either brand of the COVID-19 bivalent vaccine (primary) or (2) a high-dose or adjuvanted influenza vaccine (secondary). Main Outcomes and Measures: Stroke risk (nonhemorrhagic stroke, transient ischemic attack, combined outcome of nonhemorrhagic stroke or transient ischemic attack, or hemorrhagic stroke) during the 1- to 21-day or 22- to 42-day risk window after vaccination vs the 43- to 90-day control window. Results: There were 5 397 278 Medicare beneficiaries who received either brand of the COVID-19 bivalent vaccine (median age, 74 years [IQR, 70-80 years]; 56% were women). Among the 11 001 beneficiaries who experienced stroke after receiving either brand of the COVID-19 bivalent vaccine, there were no statistically significant associations between either brand of the COVID-19 bivalent vaccine and the outcomes of nonhemorrhagic stroke, transient ischemic attack, nonhemorrhagic stroke or transient ischemic attack, or hemorrhagic stroke during the 1- to 21-day or 22- to 42-day risk window vs the 43- to 90-day control window (incidence rate ratio [IRR] range, 0.72-1.12). Among the 4596 beneficiaries who experienced stroke after concomitant administration of either brand of the COVID-19 bivalent vaccine plus a high-dose or adjuvanted influenza vaccine, there was a statistically significant association between vaccination and nonhemorrhagic stroke during the 22- to 42-day risk window for the Pfizer-BioNTech BNT162b2; WT/OMI BA.4/BA.5 COVID-19 bivalent vaccine (IRR, 1.20 [95% CI, 1.01-1.42]; risk difference/100 000 doses, 3.13 [95% CI, 0.05-6.22]) and a statistically significant association between vaccination and transient ischemic attack during the 1- to 21-day risk window for the Moderna mRNA-1273.222 COVID-19 bivalent vaccine (IRR, 1.35 [95% CI, 1.06-1.74]; risk difference/100 000 doses, 3.33 [95% CI, 0.46-6.20]). Among the 21 345 beneficiaries who experienced stroke after administration of a high-dose or adjuvanted influenza vaccine, there was a statistically significant association between vaccination and nonhemorrhagic stroke during the 22- to 42-day risk window (IRR, 1.09 [95% CI, 1.02-1.17]; risk difference/100 000 doses, 1.65 [95% CI, 0.43-2.87]). Conclusions and Relevance: Among Medicare beneficiaries aged 65 years or older who experienced stroke after receiving either brand of the COVID-19 bivalent vaccine, there was no evidence of a significantly elevated risk for stroke during the days immediately after vaccination.


Subject(s)
COVID-19 , Influenza Vaccines , Influenza, Human , Ischemic Attack, Transient , Ischemic Stroke , Stroke , Aged , Female , Humans , Male , 2019-nCoV Vaccine mRNA-1273/adverse effects , 2019-nCoV Vaccine mRNA-1273/therapeutic use , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/therapeutic use , BNT162 Vaccine/adverse effects , BNT162 Vaccine/therapeutic use , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/therapeutic use , Hemorrhagic Stroke/chemically induced , Hemorrhagic Stroke/epidemiology , Hemorrhagic Stroke/etiology , Influenza Vaccines/adverse effects , Influenza Vaccines/therapeutic use , Ischemic Attack, Transient/chemically induced , Ischemic Attack, Transient/epidemiology , Ischemic Attack, Transient/etiology , Medicare , Stroke/epidemiology , Stroke/etiology , Stroke/prevention & control , United States/epidemiology , Vaccination/adverse effects , Vaccination/methods , Vaccines, Combined/adverse effects , Vaccines, Combined/therapeutic use , Centers for Disease Control and Prevention, U.S./statistics & numerical data , United States Food and Drug Administration/statistics & numerical data , Ischemic Stroke/chemically induced , Ischemic Stroke/epidemiology , Ischemic Stroke/etiology , Influenza, Human/prevention & control , Aged, 80 and over
3.
JAMA ; 331(5): 408-416, 2024 02 06.
Article in English | MEDLINE | ID: mdl-38319331

ABSTRACT

Importance: Bivalent mRNA COVID-19 vaccines were recommended in the US for children and adolescents aged 12 years or older on September 1, 2022, and for children aged 5 to 11 years on October 12, 2022; however, data demonstrating the effectiveness of bivalent COVID-19 vaccines are limited. Objective: To assess the effectiveness of bivalent COVID-19 vaccines against SARS-CoV-2 infection and symptomatic COVID-19 among children and adolescents. Design, Setting, and Participants: Data for the period September 4, 2022, to January 31, 2023, were combined from 3 prospective US cohort studies (6 sites total) and used to estimate COVID-19 vaccine effectiveness among children and adolescents aged 5 to 17 years. A total of 2959 participants completed periodic surveys (demographics, household characteristics, chronic medical conditions, and COVID-19 symptoms) and submitted weekly self-collected nasal swabs (irrespective of symptoms); participants submitted additional nasal swabs at the onset of any symptoms. Exposure: Vaccination status was captured from the periodic surveys and supplemented with data from state immunization information systems and electronic medical records. Main Outcome and Measures: Respiratory swabs were tested for the presence of the SARS-CoV-2 virus using reverse transcriptase-polymerase chain reaction. SARS-CoV-2 infection was defined as a positive test regardless of symptoms. Symptomatic COVID-19 was defined as a positive test and 2 or more COVID-19 symptoms within 7 days of specimen collection. Cox proportional hazards models were used to estimate hazard ratios for SARS-CoV-2 infection and symptomatic COVID-19 among participants who received a bivalent COVID-19 vaccine dose vs participants who received no vaccine or monovalent vaccine doses only. Models were adjusted for age, sex, race, ethnicity, underlying health conditions, prior SARS-CoV-2 infection status, geographic site, proportion of circulating variants by site, and local virus prevalence. Results: Of the 2959 participants (47.8% were female; median age, 10.6 years [IQR, 8.0-13.2 years]; 64.6% were non-Hispanic White) included in this analysis, 25.4% received a bivalent COVID-19 vaccine dose. During the study period, 426 participants (14.4%) had laboratory-confirmed SARS-CoV-2 infection. Among these 426 participants, 184 (43.2%) had symptomatic COVID-19, 383 (89.9%) were not vaccinated or had received only monovalent COVID-19 vaccine doses (1.38 SARS-CoV-2 infections per 1000 person-days), and 43 (10.1%) had received a bivalent COVID-19 vaccine dose (0.84 SARS-CoV-2 infections per 1000 person-days). Bivalent vaccine effectiveness against SARS-CoV-2 infection was 54.0% (95% CI, 36.6%-69.1%) and vaccine effectiveness against symptomatic COVID-19 was 49.4% (95% CI, 22.2%-70.7%). The median observation time after vaccination was 276 days (IQR, 142-350 days) for participants who received only monovalent COVID-19 vaccine doses vs 50 days (IQR, 27-74 days) for those who received a bivalent COVID-19 vaccine dose. Conclusion and Relevance: The bivalent COVID-19 vaccines protected children and adolescents against SARS-CoV-2 infection and symptomatic COVID-19. These data demonstrate the benefit of COVID-19 vaccine in children and adolescents. All eligible children and adolescents should remain up to date with recommended COVID-19 vaccinations.


Subject(s)
COVID-19 Vaccines , COVID-19 , Adolescent , Child , Female , Humans , Male , COVID-19/diagnosis , COVID-19/prevention & control , COVID-19 Vaccines/therapeutic use , Prospective Studies , SARS-CoV-2 , mRNA Vaccines/therapeutic use , Vaccines, Combined/therapeutic use , Child, Preschool , Vaccine Efficacy , United States
4.
N Engl J Med ; 389(24): 2245-2255, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38091531

ABSTRACT

BACKGROUND: Quadrivalent recombinant influenza vaccines contain three times the amount of hemagglutinin protein as standard-dose egg-based vaccines, and the recombinant formulation is not susceptible to antigenic drift during manufacturing. Data are needed on the relative effectiveness of recombinant vaccines as compared with standard-dose vaccines against influenza-related outcomes in adults under the age of 65 years. METHODS: In this cluster-randomized observational study, Kaiser Permanente Northern California facilities routinely administered either a high-dose recombinant influenza vaccine (Flublok Quadrivalent) or one of two standard-dose influenza vaccines during the 2018-2019 and 2019-2020 influenza seasons to adults 50 to 64 years of age (primary age group) and 18 to 49 years of age. Each facility alternated weekly between the two vaccine formulations. The primary outcome was influenza (A or B) confirmed by polymerase-chain-reaction (PCR) testing. Secondary outcomes included influenza A, influenza B, and influenza-related hospitalization outcomes. We used Cox regression analysis to estimate the hazard ratio of the recombinant vaccine as compared with the standard-dose vaccines against each outcome. We calculated the relative vaccine effectiveness as 1 minus the hazard ratio. RESULTS: The study population included 1,630,328 vaccinees between the ages of 18 and 64 years (632,962 in the recombinant-vaccine group and 997,366 in the standard-dose group). During this study period, 1386 cases of PCR-confirmed influenza were diagnosed in the recombinant-vaccine group and 2435 cases in the standard-dose group. Among the participants who were 50 to 64 years of age, 559 participants (2.00 cases per 1000) tested positive for influenza in the recombinant-vaccine group as compared with 925 participants (2.34 cases per 1000) in the standard-dose group (relative vaccine effectiveness, 15.3%; 95% confidence interval [CI], 5.9 to 23.8; P = 0.002). In the same age group, the relative vaccine effectiveness against influenza A was 15.7% (95% CI, 6.0 to 24.5; P = 0.002). The recombinant vaccine was not significantly more protective against influenza-related hospitalization than were the standard-dose vaccines. CONCLUSIONS: The high-dose recombinant vaccine conferred more protection against PCR-confirmed influenza than an egg-based standard-dose vaccine among adults between the ages of 50 and 64 years. (Funded by Sanofi; ClinicalTrials.gov number, NCT03694392.).


Subject(s)
Influenza Vaccines , Influenza, Human , Vaccines, Combined , Vaccines, Synthetic , Adolescent , Adult , Humans , Middle Aged , Young Adult , Hospitalization , Influenza Vaccines/administration & dosage , Influenza Vaccines/therapeutic use , Influenza, Human/prevention & control , Influenza, Human/epidemiology , Proportional Hazards Models , Vaccines, Combined/administration & dosage , Vaccines, Combined/therapeutic use , Vaccines, Inactivated , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/therapeutic use
6.
Vaccine ; 41(42): 6359-6365, 2023 10 06.
Article in English | MEDLINE | ID: mdl-37696717

ABSTRACT

BACKGROUND: Influenza vaccines prevent influenza-related morbidity and mortality; however, suboptimal vaccine effectiveness (VE) of non-adjuvanted trivalent inactivated influenza vaccine (naTIV) or quadrivalent formulations in older adults prompted the use of enhanced products such as adjuvanted TIV (aTIV). Here, the VE of aTIV is compared to naTIV for preventing influenza-associated hospitalization among older adults. METHODS: A test-negative design study was used with pooled data from the 2012 to 2015 influenza seasons. An inverse probability of treatment (IPT)-weighted logistic regression estimated the Odds Ratio (OR) for laboratory-confirmed influenza-associated hospitalization. VE was calculated as (1-OR)*100% with accompanying 95% confidence intervals (CI). RESULTS: Of 7,101 adults aged ≥ 65, 3,364 received naTIV and 526 received aTIV. The overall VE against influenza hospitalization was 45.9% (95% CI: 40.2%-51.1%) for naTIV and 53.5% (42.8%-62.3%) for aTIV. No statistically significant differences in VE were found between aTIV and naTIV by age group or influenza season, though a trend favoring aTIV over naTIV was noted. Frailty may have impacted VE in aTIV recipients compared to those receiving naTIV, according to an exploratory analysis; VE adjusted by frailty was 59.1% (49.6%-66.8%) for aTIV and 44.8% (39.1%-50.0%) for naTIV. The overall relative VE of aTIV to naTIV against laboratory-confirmed influenza hospital admission was 25% (OR 0.75; 0.61-0.92), demonstrating statistically significant benefit favoring aTIV. CONCLUSIONS: Adjusting for frailty, aTIV showed statistically significantly better protection than naTIV against influenza-associated hospitalizations in older adults. In future studies, it is important to consider frailty as a significant confounder of VE.


Subject(s)
Adjuvants, Immunologic , Frailty , Influenza Vaccines , Influenza, Human , Vaccine Efficacy , Aged , Humans , Canada/epidemiology , Hospitalization , Immunization , Influenza Vaccines/therapeutic use , Influenza, Human/prevention & control , Influenza, Human/epidemiology , Seasons , Vaccines, Inactivated , Vaccines, Combined/therapeutic use
7.
Hum Vaccin Immunother ; 19(2): 2235926, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37549923

ABSTRACT

Streptococcus pneumoniae causes a considerable disease burden among children in China. Many isolates exhibit antimicrobial resistance but are often serotypes covered by the 13-valent pneumococcal conjugate vaccine (PCV13). Because the approved infant immunization schedule in China allows PCV13 vaccination only for those 6 weeks to 15 months of age, this phase 3 study was conducted to evaluate PCV13 immunogenicity and safety in unvaccinated older infants and children. Eligible participants were stratified by age into four cohorts: Cohort 1 (n = 125), 6 weeks-2 months; Cohort 2 (n = 354), 7-<12 months; Cohort 3 (n = 250), 1 -<2 years; Cohort 4 (n = 207), 2-<6 years. Cohort 1 received PCV13 at ages 2, 4, and 6 months; older cohorts were randomized 2:1 to PCV13 or Haemophilus influenzae type b (Hib) vaccine using age-appropriate schedules. Within-group immune responses were assessed by immunoglobulin G (IgG) concentrations and opsonophagocytic activity (OPA) titers. Safety evaluations included solicited reactogenicity events and adverse events (AEs). IgG geometric mean concentrations and OPA geometric mean titers for all 13 PCV13 serotypes increased for all participants vaccinated with PCV13, but not those vaccinated with Hib. Immune responses in Cohorts 2-4 were generally comparable with those in Cohort 1 (the infant series) for most serotypes. PCV13 was well tolerated across cohorts, with reported AEs consistent with expectations in these age groups; no new safety signals were identified. These results suggest that PCV13 administered as a catch-up regimen to infants and children 7 months-<6 years of age in China will effectively reduce vaccine-type pneumococcal disease in this population. NCT03574389.


Subject(s)
East Asian People , Immunogenicity, Vaccine , Pneumococcal Infections , Pneumococcal Vaccines , Child , Child, Preschool , Humans , Infant , Antibodies, Bacterial , Immunoglobulin G , Pneumococcal Infections/prevention & control , Pneumococcal Vaccines/immunology , Pneumococcal Vaccines/therapeutic use , Streptococcus pneumoniae , Vaccines, Conjugate/immunology , Vaccines, Conjugate/therapeutic use , Treatment Outcome , Vaccines, Combined/immunology , Vaccines, Combined/therapeutic use
8.
N Engl J Med ; 389(3): 215-227, 2023 Jul 20.
Article in English | MEDLINE | ID: mdl-37467497

ABSTRACT

BACKGROUND: Natural history studies have correlated serotype-specific anti-capsular polysaccharide (CPS) IgG in newborns with a reduced risk of group B streptococcal disease. A hexavalent CPS-cross-reactive material 197 glycoconjugate vaccine (GBS6) is being developed as a maternal vaccine to prevent invasive group B streptococcus in young infants. METHODS: In an ongoing phase 2, placebo-controlled trial involving pregnant women, we assessed the safety and immunogenicity of a single dose of various GBS6 formulations and analyzed maternally transferred anti-CPS antibodies. In a parallel seroepidemiologic study that was conducted in the same population, we assessed serotype-specific anti-CPS IgG concentrations that were associated with a reduced risk of invasive disease among newborns through 89 days of age to define putative protective thresholds. RESULTS: Naturally acquired anti-CPS IgG concentrations were associated with a reduced risk of disease among infants in the seroepidemiologic study. IgG thresholds that were determined to be associated with 75 to 95% reductions in the risk of disease were 0.184 to 0.827 µg per milliliter. No GBS6-associated safety signals were observed among the mothers or infants. The incidence of adverse events and of serious adverse events were similar across the trial groups for both mothers and infants; more local reactions were observed in the groups that received GBS6 containing aluminum phosphate. Among the infants, the most common serious adverse events were minor congenital anomalies (umbilical hernia and congenital dermal melanocytosis). GBS6 induced maternal antibody responses to all serotypes, with maternal-to-infant antibody ratios of approximately 0.4 to 1.3, depending on the dose. The percentage of infants with anti-CPS IgG concentrations above 0.184 µg per milliliter varied according to serotype and formulation, with 57 to 97% of the infants having a seroresponse to the most immunogenic formulation. CONCLUSIONS: GBS6 elicited anti-CPS antibodies against group B streptococcus in pregnant women that were transferred to infants at levels associated with a reduced risk of invasive group B streptococcal disease. (Funded by Pfizer and the Bill and Melinda Gates Foundation; C1091002 ClinicalTrials.gov number, NCT03765073.).


Subject(s)
Streptococcal Infections , Streptococcal Vaccines , Streptococcus agalactiae , Female , Humans , Infant , Infant, Newborn , Pregnancy , Antibodies, Bacterial , Immunoglobulin G , Seroepidemiologic Studies , Streptococcal Infections/epidemiology , Streptococcal Infections/immunology , Streptococcal Infections/prevention & control , Vaccines, Combined/administration & dosage , Vaccines, Combined/adverse effects , Vaccines, Combined/immunology , Vaccines, Combined/therapeutic use , Vaccines, Conjugate/administration & dosage , Vaccines, Conjugate/adverse effects , Vaccines, Conjugate/immunology , Vaccines, Conjugate/therapeutic use , Streptococcal Vaccines/administration & dosage , Streptococcal Vaccines/adverse effects , Streptococcal Vaccines/immunology , Streptococcal Vaccines/therapeutic use , Immunity, Maternally-Acquired/immunology
10.
N Engl J Med ; 388(16): 1465-1477, 2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37018468

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) infection causes considerable illness in older adults. The efficacy and safety of an investigational bivalent RSV prefusion F protein-based (RSVpreF) vaccine in this population are unknown. METHODS: In this ongoing, phase 3 trial, we randomly assigned, in a 1:1 ratio, adults (≥60 years of age) to receive a single intramuscular injection of RSVpreF vaccine at a dose of 120 µg (RSV subgroups A and B, 60 µg each) or placebo. The two primary end points were vaccine efficacy against seasonal RSV-associated lower respiratory tract illness with at least two or at least three signs or symptoms. The secondary end point was vaccine efficacy against RSV-associated acute respiratory illness. RESULTS: At the interim analysis (data-cutoff date, July 14, 2022), 34,284 participants had received RSVpreF vaccine (17,215 participants) or placebo (17,069 participants). RSV-associated lower respiratory tract illness with at least two signs or symptoms occurred in 11 participants in the vaccine group (1.19 cases per 1000 person-years of observation) and 33 participants in the placebo group (3.58 cases per 1000 person-years of observation) (vaccine efficacy, 66.7%; 96.66% confidence interval [CI], 28.8 to 85.8); 2 cases (0.22 cases per 1000 person-years of observation) and 14 cases (1.52 cases per 1000 person-years of observation), respectively, occurred with at least three signs or symptoms (vaccine efficacy, 85.7%; 96.66% CI, 32.0 to 98.7). RSV-associated acute respiratory illness occurred in 22 participants in the vaccine group (2.38 cases per 1000 person-years of observation) and 58 participants in the placebo group (6.30 cases per 1000 person-years of observation) (vaccine efficacy, 62.1%; 95% CI, 37.1 to 77.9). The incidence of local reactions was higher with vaccine (12%) than with placebo (7%); the incidences of systemic events were similar (27% and 26%, respectively). Similar rates of adverse events through 1 month after injection were reported (vaccine, 9.0%; placebo, 8.5%), with 1.4% and 1.0%, respectively, considered by the investigators to be injection-related. Severe or life-threatening adverse events were reported in 0.5% of vaccine recipients and 0.4% of placebo recipients. Serious adverse events were reported in 2.3% of participants in each group through the data-cutoff date. CONCLUSIONS: RSVpreF vaccine prevented RSV-associated lower respiratory tract illness and RSV-associated acute respiratory illness in adults (≥60 years of age), without evident safety concerns. (Funded by Pfizer; RENOIR ClinicalTrials.gov number, NCT05035212; EudraCT number, 2021-003693-31.).


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Tract Infections , Aged , Humans , Antibodies, Viral , Double-Blind Method , Respiratory Syncytial Virus Infections/diagnosis , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/therapeutic use , Vaccines, Combined/administration & dosage , Vaccines, Combined/adverse effects , Vaccines, Combined/therapeutic use , Vaccine Efficacy , Treatment Outcome , Middle Aged , Injections, Intramuscular , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/prevention & control
11.
N Engl J Med ; 388(16): 1451-1464, 2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37018474

ABSTRACT

BACKGROUND: Whether vaccination during pregnancy could reduce the burden of respiratory syncytial virus (RSV)-associated lower respiratory tract illness in newborns and infants is uncertain. METHODS: In this phase 3, double-blind trial conducted in 18 countries, we randomly assigned, in a 1:1 ratio, pregnant women at 24 through 36 weeks' gestation to receive a single intramuscular injection of 120 µg of a bivalent RSV prefusion F protein-based (RSVpreF) vaccine or placebo. The two primary efficacy end points were medically attended severe RSV-associated lower respiratory tract illness and medically attended RSV-associated lower respiratory tract illness in infants within 90, 120, 150, and 180 days after birth. A lower boundary of the confidence interval for vaccine efficacy (99.5% confidence interval [CI] at 90 days; 97.58% CI at later intervals) greater than 20% was considered to meet the success criterion for vaccine efficacy with respect to the primary end points. RESULTS: At this prespecified interim analysis, the success criterion for vaccine efficacy was met with respect to one primary end point. Overall, 3682 maternal participants received vaccine and 3676 received placebo; 3570 and 3558 infants, respectively, were evaluated. Medically attended severe lower respiratory tract illness occurred within 90 days after birth in 6 infants of women in the vaccine group and 33 infants of women in the placebo group (vaccine efficacy, 81.8%; 99.5% CI, 40.6 to 96.3); 19 cases and 62 cases, respectively, occurred within 180 days after birth (vaccine efficacy, 69.4%; 97.58% CI, 44.3 to 84.1). Medically attended RSV-associated lower respiratory tract illness occurred within 90 days after birth in 24 infants of women in the vaccine group and 56 infants of women in the placebo group (vaccine efficacy, 57.1%; 99.5% CI, 14.7 to 79.8); these results did not meet the statistical success criterion. No safety signals were detected in maternal participants or in infants and toddlers up to 24 months of age. The incidences of adverse events reported within 1 month after injection or within 1 month after birth were similar in the vaccine group (13.8% of women and 37.1% of infants) and the placebo group (13.1% and 34.5%, respectively). CONCLUSIONS: RSVpreF vaccine administered during pregnancy was effective against medically attended severe RSV-associated lower respiratory tract illness in infants, and no safety concerns were identified. (Funded by Pfizer; MATISSE ClinicalTrials.gov number, NCT04424316.).


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Tract Infections , Female , Humans , Infant , Infant, Newborn , Pregnancy , Antibodies, Viral , Communicable Diseases/therapy , Double-Blind Method , Injections, Intramuscular , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/therapeutic use , Respiratory Syncytial Viruses , Treatment Outcome , Vaccination/adverse effects , Vaccination/methods , Vaccine Efficacy , Vaccines, Combined/administration & dosage , Vaccines, Combined/adverse effects , Vaccines, Combined/therapeutic use , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/prevention & control
15.
N Engl J Med ; 388(3): 214-227, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36652353

ABSTRACT

BACKGROUND: The emergence of immune-escape variants of severe acute respiratory syndrome coronavirus 2 warrants the use of sequence-adapted vaccines to provide protection against coronavirus disease 2019. METHODS: In an ongoing phase 3 trial, adults older than 55 years who had previously received three 30-µg doses of the BNT162b2 vaccine were randomly assigned to receive 30 µg or 60 µg of BNT162b2, 30 µg or 60 µg of monovalent B.1.1.529 (omicron) BA.1-adapted BNT162b2 (monovalent BA.1), or 30 µg (15 µg of BNT162b2 + 15 µg of monovalent BA.1) or 60 µg (30 µg of BNT162b2 + 30 µg of monovalent BA.1) of BA.1-adapted BNT162b2 (bivalent BA.1). Primary objectives were to determine superiority (with respect to 50% neutralizing titer [NT50] against BA.1) and noninferiority (with respect to seroresponse) of the BA.1-adapted vaccines to BNT162b2 (30 µg). A secondary objective was to determine noninferiority of bivalent BA.1 to BNT162b2 (30 µg) with respect to neutralizing activity against the ancestral strain. Exploratory analyses assessed immune responses against omicron BA.4, BA.5, and BA.2.75 subvariants. RESULTS: A total of 1846 participants underwent randomization. At 1 month after vaccination, bivalent BA.1 (30 µg and 60 µg) and monovalent BA.1 (60 µg) showed neutralizing activity against BA.1 superior to that of BNT162b2 (30 µg), with NT50 geometric mean ratios (GMRs) of 1.56 (95% confidence interval [CI], 1.17 to 2.08), 1.97 (95% CI, 1.45 to 2.68), and 3.15 (95% CI, 2.38 to 4.16), respectively. Bivalent BA.1 (both doses) and monovalent BA.1 (60 µg) were also noninferior to BNT162b2 (30 µg) with respect to seroresponse against BA.1; between-group differences ranged from 10.9 to 29.1 percentage points. Bivalent BA.1 (either dose) was noninferior to BNT162b2 (30 µg) with respect to neutralizing activity against the ancestral strain, with NT50 GMRs of 0.99 (95% CI, 0.82 to 1.20) and 1.30 (95% CI, 1.07 to 1.58), respectively. BA.4-BA.5 and BA.2.75 neutralizing titers were numerically higher with 30-µg bivalent BA.1 than with 30-µg BNT162b2. The safety profile of either dose of monovalent or bivalent BA.1 was similar to that of BNT162b2 (30 µg). Adverse events were more common in the 30-µg monovalent-BA.1 (8.5%) and 60-µg bivalent-BA.1 (10.4%) groups than in the other groups (3.6 to 6.6%). CONCLUSIONS: The candidate monovalent or bivalent omicron BA.1-adapted vaccines had a safety profile similar to that of BNT162b2 (30 µg), induced substantial neutralizing responses against ancestral and omicron BA.1 strains, and, to a lesser extent, neutralized BA.4, BA.5, and BA.2.75 strains. (Funded by BioNTech and Pfizer; ClinicalTrials.gov number, NCT04955626.).


Subject(s)
BNT162 Vaccine , COVID-19 , SARS-CoV-2 , Vaccines, Combined , Humans , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , BNT162 Vaccine/adverse effects , BNT162 Vaccine/immunology , BNT162 Vaccine/therapeutic use , COVID-19/genetics , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , SARS-CoV-2/drug effects , SARS-CoV-2/genetics , SARS-CoV-2/immunology , Vaccination , Vaccines, Combined/therapeutic use , Middle Aged
19.
Article in English | LILACS, CUMED | ID: biblio-1442245

ABSTRACT

This study was conducted to prepare and evaluate the potency of different inactivated vaccine formulations that protect chickens against Salmonella Enteritidis and Newcastle disease virus using Montanide as adjuvant. Protection and the humoral immune response of prepared vaccines against Salmonella Enteritidis and Newcastle disease virus was evaluated and compared to imported vaccine. In this study, different formulae of Salmonella Enteritidis and Newcastle disease vaccines were prepared and compared with the imported one by measuring the antibody titer against Newcastle disease virus by hemagglutination inhibition test and the antibody titer against Salmonella Enteritidis using Enzyme Linked Immunosorbent Assay. On the other hand, the protection percentages against Newcastle disease and Salmonella Enteritidis were recorded to determine the best effective formula. The highest hemagglutination inhibition antibody level against NDV at first week was recorded for the prepared combined Newcastle disease and Salmonella Enteritidis vaccine (4.2 log2) followed by the prepared monovalent Newcastle disease (3.4 log2); the lowest antibody level (3.1 log2) was obtained with the imported vaccine. A gradual increase was observed in all groups to 7.1 log2, 6.8 log2 and 6.4 log2 at fourth week post vaccination, respectively. The antibody titer against Salmonella Enteritidis was 552 for the prepared combined Salmonella Enteritidis and Newcastle disease, followed by the prepared monovalent Salmonella Enteritidis (477) at first week post vaccination; the antibody titer obtained for the imported vaccine was 477. There was a gradual increase to 1456, 1406 and 1130 at fourth week post vaccination, respectively. Prepared combined vaccines gave the highest protection percentage, followed by prepared monovalent types and finally imported vaccines. Vaccination by the prepared combined Salmonella Enteritidis and Newcastle disease vaccine may be a way to increase the resistance of birds to Salmonella and Newcastle and to decrease the shedding rate(AU)


Este estudio se llevó a cabo para preparar y evaluar la potencia de diferentes formulaciones de vacunas inactivadas que protegen a los pollos contra Salmonella Enteritidis y el virus de la enfermedad de Newcastle utilizando Montanide como adyuvante. Se evaluó la protección y la respuesta inmune humoral de las vacunas preparadas contra Salmonella Enteritidis y el virus de la enfermedad de Newcastle y se comparó con la vacuna importada. En este estudio se prepararon diferentes fórmulas de vacunas contra Salmonella Enteritidis y la enfermedad de Newcastle y se compararon con la importada midiendo el título de anticuerpos contra el virus de la enfermedad de Newcastle mediante la prueba de inhibición de la hemaglutinación y el título de anticuerpos contra Salmonella Enteritidis mediante ELISA. Por otra parte, se registraron los porcentajes de protección contra la enfermedad de Newcastle y Salmonella Enteritidis para determinar la fórmula más eficaz. El mayor nivel de anticuerpos inhibidores de la hemaglutinación contra el virus de la enfermedad de Newcastle, en la primera semana, se registró con la vacuna combinada preparada contra la enfermedad de Newcastle y Salmonella Enteritidis (4,2 log2), seguida de la vacuna monovalente preparada contra la enfermedad de Newcastle (3,4 log2); el menor nivel de anticuerpos (3,1 log2) se obtuvo con la vacuna importada. Se observó un aumento gradual en todos los grupos hasta alcanzar 7,1 log2, 6,8 log2 y 6,4 log2 en la cuarta semana tras la vacunación, respectivamente. El título de anticuerpos contra Salmonella Enteritidis fue de 552 para la vacuna combinada preparada contra la Salmonella Enteritidis y enfermedad de Newcastle, seguida por la vacuna monovalente preparada contra Salmonella Enteritidis (477) en la primera semana después de la vacunación; el título de anticuerpos obtenido con la vacuna importada fue de 477. Hubo un aumento gradual hasta 1456, 1406 y 1130 en la cuarta semana después de la vacunación, respectivamente. Las vacunas combinadas preparadas dieron el mayor porcentaje de protección, seguidas por los tipos monovalentes preparados y, por último, por las vacunas importadas. La vacunación con la vacuna combinada preparada contra la Salmonella Enteritidis y la enfermedad de Newcastle puede ser una forma de aumentar la resistencia de las aves a la Salmonella y Newcastle y de disminuir la tasa de excreción(AU)


Subject(s)
Humans , Salmonella enteritidis , Newcastle disease virus , Enzyme-Linked Immunosorbent Assay/methods , Hemagglutination Inhibition Tests/methods , Vaccines, Combined/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL