Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 385
Filter
1.
Signal Transduct Target Ther ; 9(1): 129, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740763

ABSTRACT

The safety and efficacy of COVID-19 vaccines in the elderly, a high-risk group for severe COVID-19 infection, have not been fully understood. To clarify these issues, this prospective study followed up 157 elderly and 73 young participants for 16 months and compared the safety, immunogenicity, and efficacy of two doses of the inactivated vaccine BBIBP-CorV followed by a booster dose of the recombinant protein vaccine ZF2001. The results showed that this vaccination protocol was safe and tolerable in the elderly. After administering two doses of the BBIBP-CorV, the positivity rates and titers of neutralizing and anti-RBD antibodies in the elderly were significantly lower than those in the young individuals. After the ZF2001 booster dose, the antibody-positive rates in the elderly were comparable to those in the young; however, the antibody titers remained lower. Gender, age, and underlying diseases were independently associated with vaccine immunogenicity in elderly individuals. The pseudovirus neutralization assay showed that, compared with those after receiving two doses of BBIBP-CorV priming, some participants obtained immunological protection against BA.5 and BF.7 after receiving the ZF2001 booster. Breakthrough infection symptoms last longer in the infected elderly and pre-infection antibody titers were negatively associated with the severity of post-infection symptoms. The antibody levels in the elderly increased significantly after breakthrough infection but were still lower than those in the young. Our data suggest that multiple booster vaccinations at short intervals to maintain high antibody levels may be an effective strategy for protecting the elderly against COVID-19.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Vaccines, Inactivated , Humans , COVID-19/prevention & control , COVID-19/immunology , Female , Male , Aged , COVID-19 Vaccines/immunology , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/administration & dosage , SARS-CoV-2/immunology , Prospective Studies , Antibodies, Viral/immunology , Antibodies, Viral/blood , Vaccines, Inactivated/immunology , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/administration & dosage , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Aged, 80 and over , Adult , Vaccination , Longitudinal Studies , Middle Aged , Vaccines, Synthetic/immunology , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/administration & dosage , Immunogenicity, Vaccine/immunology , Immunization, Secondary
2.
Nat Med ; 30(5): 1363-1372, 2024 May.
Article in English | MEDLINE | ID: mdl-38637636

ABSTRACT

Here we conducted a multicenter open-label, randomized phase 2 and 3 study to assess the safety and immunogenicity of a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) Omicron-specific (BA.1/B.1.1.529), monovalent, thermostable, self-amplifying mRNA vaccine, GEMCOVAC-OM, when administered intradermally as a booster in healthy adults who had received two doses of BBV152 or ChAdOx1 nCoV-19. GEMCOVAC-OM was well tolerated with no related serious adverse events in both phase 2 and phase 3. In phase 2, the safety and immunogenicity of GEMCOVAC-OM was compared with our prototype mRNA vaccine GEMCOVAC-19 (D614G variant-specific) in 140 participants. At day 29 after vaccination, there was a significant rise in anti-spike (BA.1) IgG antibodies with GEMCOVAC-OM (P < 0.0001) and GEMCOVAC-19 (P < 0.0001). However, the IgG titers (primary endpoint) and seroconversion were higher with GEMCOVAC-OM (P < 0.0001). In phase 3, GEMCOVAC-OM was compared with ChAdOx1 nCoV-19 in 3,140 participants (safety cohort), which included an immunogenicity cohort of 420 participants. At day 29, neutralizing antibody titers against the BA.1 variant of SARS-CoV-2 were significantly higher than baseline in the GEMCOVAC-OM arm (P < 0.0001), but not in the ChAdOx1 nCoV-19 arm (P = 0.1490). GEMCOVAC-OM was noninferior (primary endpoint) and superior to ChAdOx1 nCoV-19 in terms of neutralizing antibody titers and seroconversion rate (lower bound 95% confidence interval of least square geometric mean ratio >1 and difference in seroconversion >0% for superiority). At day 29, anti-spike IgG antibodies and seroconversion (secondary endpoints) were significantly higher with GEMCOVAC-OM (P < 0.0001). These results demonstrate that GEMCOVAC-OM is safe and boosts immune responses against the B.1.1.529 variant. Clinical Trial Registry India identifier: CTRI/2022/10/046475 .


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization, Secondary , SARS-CoV-2 , Humans , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/adverse effects , SARS-CoV-2/immunology , Male , Female , Adult , COVID-19/prevention & control , COVID-19/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Middle Aged , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Immunoglobulin G/immunology , Immunoglobulin G/blood , Young Adult , mRNA Vaccines/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Immunogenicity, Vaccine/immunology , ChAdOx1 nCoV-19/immunology
3.
Pediatrics ; 153(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38548700

ABSTRACT

BACKGROUND AND OBJECTIVES: The messenger RNA (mRNA)-based coronavirus disease 2019 vaccines approved for use in children <5 years of age have different antigen doses and administration schedules that could affect vaccine immunogenicity and effectiveness. We sought to compare the strength and breadth of serum binding and neutralizing antibodies to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) elicited by monovalent mRNA-based coronavirus disease 2019 vaccines in young children. METHODS: We conducted a prospective cohort study of children 6 months to 4 years of age who completed primary series vaccination with monovalent mRNA-1273 or BNT162b2 vaccines. Serum was collected 1 month after primary vaccine series completion for the measurement of SARS-CoV-2-specific humoral immune responses, including antibody binding responses to Spike proteins from an ancestral strain (D614G) and major variants of SARS-CoV-2 and antibody neutralizing activity against D614G and Omicron subvariants (BA.1, BA.4/5). RESULTS: Of 75 participants, 40 (53%) received mRNA-1273 and 35 (47%) received BNT162b2. Children receiving either primary vaccine series developed robust and broad SARS-CoV-2-specific binding and neutralizing antibodies, including to Omicron subvariants. Children with a previous history of SARS-CoV-2 infection developed significantly higher antibody binding responses and neutralization titers to Omicron subvariants, which is consistent with the occurrence of identified infections during the circulation of Omicron subvariants in the region. CONCLUSIONS: Monovalent mRNA-1273 and BNT162b2 elicited similar antibody responses 1 month after vaccination in young children. In addition, previous infection significantly enhanced the strength of antibody responses to Omicron subvariants. The authors of future studies should evaluate incorporation of these vaccines into the standard childhood immunization schedule.


Subject(s)
2019-nCoV Vaccine mRNA-1273 , Antibodies, Neutralizing , Antibodies, Viral , BNT162 Vaccine , COVID-19 , Immunogenicity, Vaccine , Humans , BNT162 Vaccine/immunology , BNT162 Vaccine/administration & dosage , Infant , 2019-nCoV Vaccine mRNA-1273/immunology , Child, Preschool , Male , Antibodies, Neutralizing/blood , Prospective Studies , Female , Immunogenicity, Vaccine/immunology , COVID-19/prevention & control , COVID-19/immunology , Antibodies, Viral/blood , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Cohort Studies , Spike Glycoprotein, Coronavirus/immunology
4.
Immunohorizons ; 7(10): 635-651, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37819998

ABSTRACT

Spike-encoding mRNA vaccines in early 2021 effectively reduced SARS-CoV-2-associated morbidity and mortality. New booster regimens were introduced due to successive waves of distinct viral variants. Therefore, people now have a diverse immune memory resulting from multiple SARS-CoV-2 Ag exposures, from infection to following vaccination. This level of community-wide immunity can induce immunological protection from SARS-CoV-2; however, questions about the trajectory of the adaptive immune responses and long-term immunity with respect to priming and repeated Ag exposure remain poorly explored. In this study, we examined the trajectory of adaptive immune responses following three doses of monovalent Pfizer BNT162b2 mRNA vaccination in immunologically naive and SARS-CoV-2 preimmune individuals without the occurrence of breakthrough infection. The IgG, B cell, and T cell Spike-specific responses were assessed in human blood samples collected at six time points between a moment before vaccination and up to 6 mo after the third immunization. Overall, the impact of repeated Spike exposures had a lower improvement on T cell frequency and longevity compared with IgG responses. Natural infection shaped the responses following the initial vaccination by significantly increasing neutralizing Abs and specific CD4+ T cell subsets (circulating T follicular helper, effector memory, and Th1-producing cells), but it had a small benefit at long-term immunity. At the end of the three-dose vaccination regimen, both SARS-CoV-2-naive and preimmune individuals had similar immune memory quality and quantity. This study provides insights into the durability of mRNA vaccine-induced immunological memory and the effects of preimmunity on long-term responses.


Subject(s)
BNT162 Vaccine , COVID-19 , mRNA Vaccines , Humans , BNT162 Vaccine/immunology , BNT162 Vaccine/therapeutic use , COVID-19/immunology , COVID-19/prevention & control , Immunoglobulin G/immunology , mRNA Vaccines/immunology , SARS-CoV-2 , Vaccines, Synthetic/immunology , Immunogenicity, Vaccine/immunology , Vaccine Efficacy , Immunization, Secondary , Lymphocyte Subsets/immunology
5.
Sci Rep ; 13(1): 11798, 2023 07 21.
Article in English | MEDLINE | ID: mdl-37479776

ABSTRACT

The ChAdOx1 nCoV-19 vaccine (AZD1222) was used in Thailand during the early outbreak of coronavirus disease 2019 (COVID-19). A previous study showed a low immune response in diabetes patients after the first dose of the AZD1222 vaccine. Furthermore, humoral immune responses after the second vaccination were inconsistent. This study evaluated the immunogenicity following the first and second doses of the AZD1222 vaccine in people with type 2 diabetes (T2D) compared with the general population of Thailand. This was a prospective, single-center cohort study. 59 adults with T2D and 118 age- and sex-matched healthcare personnel were eligible. The participants received two doses of AZD1222 12 weeks apart. Antibodies against the receptor-binding domain (anti-RBD) of the SARS-CoV-2 spike protein, using an automated electrochemiluminesence immunoassay (ECLIA), were measured at baseline, 8 and 12 weeks after the first dose of vaccine, and 4 weeks after the second dose of vaccine. The anti-RBD levels were reported as the geometric mean concentration (GMC) and compared between groups using the geometric mean ratio (GMR). A total of 177 participants were included: The average age of 59 T2D patients was 60.1 years (SD: 11.4), and 31 (52.5%) of them were female. The GMC of anti-RBD 8 and 12 weeks after the first vaccination were significantly lower in T2D (week 8 60; 17.05 BAU/mL, 95% confidence interval [CI] 11.1-26.19, P = 0.035, week 12; 24.68 BAU/mL, 95% CI 16.4-37.0, P = 0.002) than in those without diabetes (week 8; 29.79 BAU/mL, 95% CI 22.07-40.42, week 12; 50.67 BAU/mL, 95% CI 40.62-63.20). However, there was no difference in the GMC of anti-RBD 4 weeks after the second vaccination among groups (T2D; 687.95 BAU/mL, 95% CI 462.7-1022.7, Normal; 697.95 BAU/mL, 95% CI 583.7-834.5, P = 0.947). In both groups, the GMC of anti-RBD was persistently high without decline 12 weeks after the first vaccination. Albuminuria was a major factor related to low humoral immune responses in T2D patients after the second dose of AZD122 vaccine (the GMR was 0.29, 95% CI 0.08-0.98, P = 0.047) whereas the HbA1C level and age were not. Immunogenicity in T2D cases was lower than in the normal population after the first dose of the AZD1222 vaccine. The two doses of AZD122 vaccine induced immunity in T2D equal to that of normal individuals in Thailand. People with diabetes should be boosted as soon as possible to induce adequate immunity to prevent COVID-19 infection.


Subject(s)
COVID-19 , ChAdOx1 nCoV-19 , Diabetes Mellitus, Type 2 , Immunogenicity, Vaccine , Female , Humans , Male , Middle Aged , ChAdOx1 nCoV-19/administration & dosage , ChAdOx1 nCoV-19/immunology , ChAdOx1 nCoV-19/therapeutic use , Cohort Studies , COVID-19/complications , COVID-19/immunology , COVID-19/prevention & control , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/immunology , Prospective Studies , SARS-CoV-2 , Southeast Asian People , Thailand/epidemiology , Immunogenicity, Vaccine/immunology , Aged
6.
MMWR recomm. rep ; 79(29): 1-6, 20230721. tab
Article in English | BIGG - GRADE guidelines | ID: biblio-1512317

ABSTRACT

A Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) review of the evidence for benefits and harms for Pfizer Respiratory Syncytial Virus (RSV) preF vaccine was presented to the Advisory Committee on Immunization Practices (ACIP) on June 21, 2023. GRADE evidence type indicates the certainty in estimates from the available body of evidence. Evidence certainty ranges from high certainty to very low certainty [1]. The policy questions were, "Should vaccination with Pfizer bivalent RSVpreF vaccine (120µg antigen, 1 dose administered intramuscularly [IM]), rather than no vaccine, be recommended in persons aged ≥65 years?" and "Should vaccination with Pfizer bivalent RSVpreF vaccine (120µg antigen, 1 dose IM), rather than no vaccine, be recommended in persons aged 60­64 years?" The benefits chosen by the ACIP RSV Vaccines Work Group (Work Group) as critical or important to policy decisions were prevention of RSV lower respiratory tract illness/disease (LRTI/LRTD) (critical), medically attended RSV LRTI/LRTD (critical), hospitalization for RSV respiratory illness (important), severe RSV respiratory illness requiring supplemental oxygen (O2) or other respiratory support (important), and death due to RSV respiratory illness (important). The harms chosen by the Work Group as critical or important to policy decisions were serious adverse events (critical), inflammatory neurologic events* (important) and reactogenicity grade ≥3 (important). A systematic review of evidence on the efficacy and safety of Pfizer RSVpreF vaccine among persons aged 60 years and older was conducted. The quality of evidence from one Phase 3 randomized controlled trial (RCT) and one Phase 1/2 RCT were assessed using the GRADE approach [2-4]. A lower risk of RSV LRTI† was observed with vaccination compared to placebo (incidence rate ratio [IRR] 0.156, 95% confidence interval [CI]: 0.048, 0.404, evidence certainty: moderate), corresponding to a vaccine efficacy of 84.4% (95% CI: 59.1%, 95.2%)§. This was observed over one complete RSV season (Season 1) and one partial second RSV season (Season 2). A lower risk of medically attended RSV LRTI¶ was also observed (IRR 0.191; 95% CI: 0.047, 0.563; evidence certainty: moderate), corresponding to a vaccine efficacy of 81.0% (95% CI: 43.5%, 95.2%)§. The trial was not powered to detect a lower risk of hospitalization for RSV respiratory illness (IRR 0.333; 95% CI: 0.006, 4.143; evidence certainty: very low) nor a lower risk for severe RSV respiratory illness requiring supplemental oxygen or other respiratory support** (IRR 0.000; 95% CI: 0.013, 78.33; evidence certainty: very low), corresponding to a vaccine efficacy of 66.7% (95% CI: -315%, 99.4%) and 0% (-7750%, 98.7%) for the outcomes, respectively. No deaths due to RSV respiratory illness were identified among vaccine recipients or placebo recipients. In terms of harms, the pooled available data from the Phase 3 and Phase 1/2 RCTs indicated that serious adverse events (SAEs)†† were balanced between the vaccine and placebo arms (risk ratio [RR] 1.041; 95% CI: 0.944, 1.148; evidence certainty: high). Reactogenicity grade ≥3§§ was similar between the vaccine and placebo arms of the trials (RR 1.43; 95% CI: 0.852, 2.385; evidence certainty: moderate), with 1.0% of vaccine recipients and 0.7% of placebo recipients reporting any grade ≥3 local or systemic reactions following injection. Three participants in the intervention group of the Phase 3 trial were reported to have inflammatory neurologic events within 42 days after vaccination (one case of Guillain-Barré syndrome [GBS], one case of Miller Fisher syndrome [a GBS variant], and one case of undifferentiated motor-sensory axonal polyneuropathy with worsening of pre-existing symptoms), compared with zero participants in the placebo group. No inflammatory neurologic events were reported in the phase 1/2 RCT.


Subject(s)
Humans , Middle Aged , Aged , Pneumococcal Infections/prevention & control , Influenza Vaccines/therapeutic use , Respiratory Syncytial Virus, Human/immunology , Immunogenicity, Vaccine/immunology , COVID-19/prevention & control
7.
Nat Commun ; 14(1): 1138, 2023 03 06.
Article in English | MEDLINE | ID: mdl-36878897

ABSTRACT

Adjuvant-containing subunit vaccines represent a promising approach for protection against tuberculosis (TB), but current candidates require refrigerated storage. Here we present results from a randomized, double-blinded Phase 1 clinical trial (NCT03722472) evaluating the safety, tolerability, and immunogenicity of a thermostable lyophilized single-vial presentation of the ID93 + GLA-SE vaccine candidate compared to the non-thermostable two-vial vaccine presentation in healthy adults. Participants were monitored for primary, secondary, and exploratory endpoints following intramuscular administration of two vaccine doses 56 days apart. Primary endpoints included local and systemic reactogenicity and adverse events. Secondary endpoints included antigen-specific antibody (IgG) and cellular immune responses (cytokine-producing peripheral blood mononuclear cells and T cells). Both vaccine presentations are safe and well tolerated and elicit robust antigen-specific serum antibody and Th1-type cellular immune responses. Compared to the non-thermostable presentation, the thermostable vaccine formulation generates greater serum antibody responses (p < 0.05) and more antibody-secreting cells (p < 0.05). In this work, we show the thermostable ID93 + GLA-SE vaccine candidate is safe and immunogenic in healthy adults.


Subject(s)
Immunogenicity, Vaccine , Tuberculosis Vaccines , Vaccines, Subunit , Adult , Humans , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/pharmacology , Adjuvants, Immunologic/therapeutic use , Antibodies/immunology , Antibody-Producing Cells/immunology , Leukocytes, Mononuclear/immunology , Tuberculosis Vaccines/adverse effects , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/pharmacology , Tuberculosis Vaccines/therapeutic use , Immunogenicity, Vaccine/immunology , Treatment Outcome , Healthy Volunteers , Temperature , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/adverse effects , Vaccines, Subunit/immunology , Vaccines, Subunit/pharmacology , Vaccines, Subunit/therapeutic use , Double-Blind Method
8.
J Virol Methods ; 316: 114716, 2023 06.
Article in English | MEDLINE | ID: mdl-36965633

ABSTRACT

Cervical cancer, the second leading cause of cancer-related deaths among women, is caused by human papillomavirus (HPV), a sexually transmitted virus. Vaccination is an effective preventive measure against viral infections and subsequent development of cervical cancer. Enzyme-linked immunosorbent assay (ELISA) is commonly used to measure specific binding antibody titers and assess the immunogenicity of test vaccines in preclinical models or clinical volunteers. Two methods of deriving titers, the endpoint titer (ET) and the effective dilution producing a median maximal effective fold of dilution (ED50) with a cut-off value, are widely used. For HPV, a pseudovirion-based neutralization assay (PBNA) is used to measure functional antibody titers. The ELISA binding titers and functional PBNA titers were found to be well-correlated for all nine HPV types tested in the vaccine, consistent with previous studies on HPV 16/18. Comparing the PBNA results with the two titration methods, the ED50 method showed higher precision and a closer correlation with PBNA results, both for individual types and pooled data analysis for all nine types. When comparing the titration results of the ET method based on a cut-off value with the ED50 method using all the data points across the dilution series, the ED50 method demonstrated better precision and a stronger correlation with PBNA results.


Subject(s)
Correlation of Data , Enzyme-Linked Immunosorbent Assay , Immunogenicity, Vaccine , Neutralization Tests , Papillomavirus Vaccines , Papillomavirus Vaccines/classification , Papillomavirus Vaccines/immunology , Enzyme-Linked Immunosorbent Assay/methods , Neutralization Tests/methods , Humans , Male , Female , Young Adult , Adult , Antibodies, Neutralizing/immunology , Reproducibility of Results , Immunogenicity, Vaccine/immunology
9.
Viruses ; 15(3)2023 03 21.
Article in English | MEDLINE | ID: mdl-36992507

ABSTRACT

Severe fever with thrombocytopenia syndrome virus (SFTSV) is an emerging tick-borne pathogen for which approved therapeutic drugs or vaccines are not available. We previously developed a recombinant vesicular stomatitis virus-based vaccine candidate (rVSV-SFTSV) by replacing the original glycoprotein with Gn/Gc from SFTSV, which conferred complete protection in a mouse model. Here, we found that two spontaneous mutations, M749T/C617R, emerged in the Gc glycoprotein during passaging that could significantly increase the titer of rVSV-SFTSV. M749T/C617R enhanced the genetic stability of rVSV-SFTSV, and no further mutations appeared after 10 passages. Using immunofluorescence analysis, we found that M749T/C617R could increase glycoprotein traffic to the plasma membrane, thus facilitating virus assembly. Remarkably, the broad-spectrum immunogenicity of rVSV-SFTSV was not affected by the M749T/C617R mutations. Overall, M749T/C617R could enhance the further development of rVSV-SFTSV into an effective vaccine in the future.


Subject(s)
Glycoproteins , Point Mutation , Vesiculovirus , Viral Envelope Proteins , Viral Vaccines , Animals , Glycoproteins/genetics , Glycoproteins/metabolism , Phlebovirus/genetics , Vesiculovirus/genetics , Viral Envelope Proteins/genetics , Viral Vaccines/genetics , Viral Vaccines/immunology , Immunogenicity, Vaccine/genetics , Immunogenicity, Vaccine/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vero Cells , Chlorocebus aethiops
10.
Viruses ; 15(2)2023 01 23.
Article in English | MEDLINE | ID: mdl-36851527

ABSTRACT

Virus-like particles (VLPs), composed of the small hepatitis B virus surface antigen (HBsAgS), are the antigenic components of the hepatitis B virus (HBV) vaccine and represent the backbones for a chimeric anti-malaria vaccine and various vaccine candidates. Biological vectors have to face pre-existing anti-vector immune responses due to previous immune exposure. Vector recognition after natural infections or vaccinations can result in unwarranted outcomes, with compromising effects on clinical outcomes. In order to evaluate the impact of a pre-existing anti-HBsAgS immune response, we developed mutant VLPs composed of subunits with reduced HBsAgS-specific antigenicity. The insertion of a Plasmodium falciparum circumsporozoite protein (CSP)-derived epitope as a read-out allowed the assessment of wild type (wt) and mutant VLPs in the context of a pre-existing immune response. Mutant and wt VLP platforms with a CSP-epitope insert are immunogenic and have the ability to generate anti-CSP antibody responses in both naïve BALB/c mice and mice with a pre-existing anti-HBsAgS immune response, but with superior anti-CSP responses in mice with a pre-existing immunity. The data indicate that previous HBsAgS exposure facilitates enhanced antibody responses against foreign epitopes delivered by the HBsAgS platform, and, in this context, the state of immune sensitization alters the outcome of subsequent vaccinations.


Subject(s)
Hepatitis B Surface Antigens , Immunogenicity, Vaccine , Malaria Vaccines , Plasmodium falciparum , Vaccines, Virus-Like Particle , Animals , Mice , Epitopes/genetics , Epitopes/immunology , Hepatitis B Surface Antigens/genetics , Hepatitis B Surface Antigens/immunology , Immunogenicity, Vaccine/genetics , Immunogenicity, Vaccine/immunology , Malaria/prevention & control , Malaria Vaccines/genetics , Malaria Vaccines/immunology , Mice, Inbred BALB C , Models, Animal , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Vaccination , Vaccines, Virus-Like Particle/genetics , Vaccines, Virus-Like Particle/immunology
12.
N Engl J Med ; 388(7): 609-620, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36791161

ABSTRACT

BACKGROUND: Respiratory syncytial virus (RSV) can cause serious lower respiratory tract disease in older adults, but no licensed RSV vaccine currently exists. An adenovirus serotype 26 RSV vector encoding a prefusion F (preF) protein (Ad26.RSV.preF) in combination with RSV preF protein was previously shown to elicit humoral and cellular immunogenicity. METHODS: We conducted a randomized, double-blind, placebo-controlled, phase 2b, proof-of-concept trial to evaluate the efficacy, immunogenicity, and safety of an Ad26.RSV.preF-RSV preF protein vaccine. Adults who were 65 years of age or older were randomly assigned in a 1:1 ratio to receive vaccine or placebo. The primary end point was the first occurrence of RSV-mediated lower respiratory tract disease that met one of three case definitions: three or more symptoms of lower respiratory tract infection (definition 1), two or more symptoms of lower respiratory tract infection (definition 2), and either two or more symptoms of lower respiratory tract infection or one or more symptoms of lower respiratory tract infection plus at least one systemic symptom (definition 3). RESULTS: Overall, 5782 participants were enrolled and received an injection. RSV-mediated lower respiratory tract disease meeting case definitions 1, 2, and 3 occurred in 6, 10, and 13 vaccine recipients and in 30, 40, and 43 placebo recipients, respectively. Vaccine efficacy was 80.0% (94.2% confidence interval [CI], 52.2 to 92.9), 75.0% (94.2% CI, 50.1 to 88.5), and 69.8% (94.2% CI, 43.7 to 84.7) for case definitions 1, 2, and 3, respectively. After vaccination, RSV A2 neutralizing antibody titers increased by a factor of 12.1 from baseline to day 15, a finding consistent with other immunogenicity measures. Percentages of participants with solicited local and systemic adverse events were higher in the vaccine group than in the placebo group (local, 37.9% vs. 8.4%; systemic, 41.4% vs. 16.4%); most adverse events were mild to moderate in severity. The frequency of serious adverse events was similar in the vaccine group and the placebo group (4.6% and 4.7%, respectively). CONCLUSIONS: In adults 65 years of age or older, Ad26.RSV.preF-RSV preF protein vaccine was immunogenic and prevented RSV-mediated lower respiratory tract disease. (Funded by Janssen Vaccines and Prevention; CYPRESS ClinicalTrials.gov number, NCT03982199.).


Subject(s)
Antibodies, Neutralizing , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Respiratory Tract Infections , Aged , Humans , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Double-Blind Method , Respiratory Syncytial Virus Infections/blood , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Vaccines/adverse effects , Respiratory Syncytial Virus Vaccines/therapeutic use , Respiratory Syncytial Virus, Human/immunology , Respiratory Tract Infections/blood , Respiratory Tract Infections/immunology , Respiratory Tract Infections/prevention & control , Vaccine Efficacy , Immunogenicity, Vaccine/immunology , Treatment Outcome
16.
N Engl J Med ; 387(18): 1673-1687, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36260859

ABSTRACT

BACKGROUND: The safety, reactogenicity, immunogenicity, and efficacy of the mRNA-1273 coronavirus disease 2019 (Covid-19) vaccine in young children are unknown. METHODS: Part 1 of this ongoing phase 2-3 trial was open label for dose selection; part 2 was an observer-blinded, placebo-controlled evaluation of the selected dose. In part 2, we randomly assigned young children (6 months to 5 years of age) in a 3:1 ratio to receive two 25-µg injections of mRNA-1273 or placebo, administered 28 days apart. The primary objectives were to evaluate the safety and reactogenicity of the vaccine and to determine whether the immune response in these children was noninferior to that in young adults (18 to 25 years of age) in a related phase 3 trial. Secondary objectives were to determine the incidences of Covid-19 and severe acute respiratory syndrome coronavirus 2 infection after administration of mRNA-1273 or placebo. RESULTS: On the basis of safety and immunogenicity results in part 1 of the trial, the 25-µg dose was evaluated in part 2. In part 2, 3040 children 2 to 5 years of age and 1762 children 6 to 23 months of age were randomly assigned to receive two 25-µg injections of mRNA-1273; 1008 children 2 to 5 years of age and 593 children 6 to 23 months of age were randomly assigned to receive placebo. The median duration of follow-up after the second injection was 71 days in the 2-to-5-year-old cohort and 68 days in the 6-to-23-month-old cohort. Adverse events were mainly low-grade and transient, and no new safety concerns were identified. At day 57, neutralizing antibody geometric mean concentrations were 1410 (95% confidence interval [CI], 1272 to 1563) among 2-to-5-year-olds and 1781 (95% CI, 1616 to 1962) among 6-to-23-month-olds, as compared with 1391 (95% CI, 1263 to 1531) among young adults, who had received 100-µg injections of mRNA-1273, findings that met the noninferiority criteria for immune responses for both age cohorts. The estimated vaccine efficacy against Covid-19 was 36.8% (95% CI, 12.5 to 54.0) among 2-to-5-year-olds and 50.6% (95% CI, 21.4 to 68.6) among 6-to-23-month-olds, at a time when B.1.1.529 (omicron) was the predominant circulating variant. CONCLUSIONS: Two 25-µg doses of the mRNA-1273 vaccine were found to be safe in children 6 months to 5 years of age and elicited immune responses that were noninferior to those in young adults. (Funded by the Biomedical Advanced Research and Development Authority and National Institute of Allergy and Infectious Diseases; KidCOVE ClinicalTrials.gov number, NCT04796896.).


Subject(s)
2019-nCoV Vaccine mRNA-1273 , COVID-19 , Immunogenicity, Vaccine , Child , Child, Preschool , Humans , Infant , Young Adult , 2019-nCoV Vaccine mRNA-1273/immunology , 2019-nCoV Vaccine mRNA-1273/therapeutic use , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/epidemiology , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , Double-Blind Method , Immunogenicity, Vaccine/immunology , Vaccine Efficacy , Treatment Outcome , Adolescent , Adult
17.
N Engl J Med ; 387(14): 1279-1291, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36112399

ABSTRACT

BACKGROUND: The safety and immunogenicity of the bivalent omicron-containing mRNA-1273.214 booster vaccine are not known. METHODS: In this ongoing, phase 2-3 study, we compared the 50-µg bivalent vaccine mRNA-1273.214 (25 µg each of ancestral Wuhan-Hu-1 and omicron B.1.1.529 [BA.1] spike messenger RNAs) with the previously authorized 50-µg mRNA-1273 booster. We administered mRNA-1273.214 or mRNA-1273 as a second booster in adults who had previously received a two-dose (100-µg) primary series and first booster (50-µg) dose of mRNA-1273 (≥3 months earlier). The primary objectives were to assess the safety, reactogenicity, and immunogenicity of mRNA-1273.214 at 28 days after the booster dose. RESULTS: Interim results are presented. Sequential groups of participants received 50 µg of mRNA-1273.214 (437 participants) or mRNA-1273 (377 participants) as a second booster dose. The median time between the first and second boosters was similar for mRNA-1273.214 (136 days) and mRNA-1273 (134 days). In participants with no previous severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, the geometric mean titers of neutralizing antibodies against the omicron BA.1 variant were 2372.4 (95% confidence interval [CI], 2070.6 to 2718.2) after receipt of the mRNA-1273.214 booster and 1473.5 (95% CI, 1270.8 to 1708.4) after receipt of the mRNA-1273 booster. In addition, 50-µg mRNA-1273.214 and 50-µg mRNA-1273 elicited geometric mean titers of 727.4 (95% CI, 632.8 to 836.1) and 492.1 (95% CI, 431.1 to 561.9), respectively, against omicron BA.4 and BA.5 (BA.4/5), and the mRNA-1273.214 booster also elicited higher binding antibody responses against multiple other variants (alpha, beta, gamma, and delta) than the mRNA-1273 booster. Safety and reactogenicity were similar with the two booster vaccines. Vaccine effectiveness was not assessed in this study; in an exploratory analysis, SARS-CoV-2 infection occurred in 11 participants after the mRNA-1273.214 booster and in 9 participants after the mRNA-1273 booster. CONCLUSIONS: The bivalent omicron-containing vaccine mRNA-1273.214 elicited neutralizing antibody responses against omicron that were superior to those with mRNA-1273, without evident safety concerns. (Funded by Moderna; ClinicalTrials.gov number, NCT04927065.).


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunization, Secondary , Vaccines, Combined , mRNA Vaccines , 2019-nCoV Vaccine mRNA-1273/immunology , 2019-nCoV Vaccine mRNA-1273/therapeutic use , Adult , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/immunology , COVID-19 Vaccines/therapeutic use , Humans , Immunogenicity, Vaccine/immunology , SARS-CoV-2 , Vaccines, Combined/immunology , Vaccines, Combined/therapeutic use , mRNA Vaccines/immunology , mRNA Vaccines/therapeutic use
18.
Front Immunol ; 13: 832924, 2022.
Article in English | MEDLINE | ID: mdl-35935974

ABSTRACT

Vaccination against COVID-19 in patients with end-stage renal disease (ESRD) on replacement therapy and kidney transplant recipients (KTRs) is particularly important due to the high mortality rate. Here, we tested the local and systemic immunity to the novel Pfizer BioNTech (BNT162b2) messenger RNA (mRNA) in ESRD, KTR patients, and healthy individuals (150 subjects). The ESRD group was divided into: hemodialysis (HD) and peritoneal dialysis (PD). We investigated the local and systemic immunity based on anti-N (nucleoprotein) and anti-S (spike1/2) Immunoglobulin A (IgA) and Immunoglobulin G (IgG) antibodies, respectively. Additionally, we performed an Interferon gamma (IFN-γ) release test Interferon-gamma release assay (IGRA) to monitor the cellular component of vaccine response. The control group had the highest level of anti-S IgG antibodies (153/2,080 binding antibody units (BAU)/ml) among all analyzed patients after the 1st and 2nd dose, respectively. The HD group (48/926 BAU/ml) had a diminished antibody level compared to PD (93/1,607 BAU/ml). Moreover, the seroconversion rate after the 1st dose was lower in HD than PD (56% vs. 86%). KTRs had extremely low seroconversion (33%). IgA-mediated immunity was the most effective in the control group, while other patients had diminished IgA production. We observed a lower percentage of vaccine responders based on the IFN-γ level in all research participants (100% vs. 85% in control, 100% vs. 80% in PD, 97% vs. 64% in HD). 63% of seropositive KTRs had a positive IGRA, while 28% of seronegative patients produced IFN-γ. Collectively, PD patients had the strongest response among ESRD patients. Two doses of the Pfizer vaccine are ineffective, especially in HD and KTRs. A closer investigation of ESRD and KTRs is required to set the COVID-19 vaccine clinical guidance. Clinical Trial Registration Number: www.ClinicalTrials.gov, identifier: NCT04 905 862.


Subject(s)
BNT162 Vaccine , COVID-19 , Immunogenicity, Vaccine , Kidney Failure, Chronic , Kidney Transplantation , Peritoneal Dialysis , BNT162 Vaccine/administration & dosage , BNT162 Vaccine/adverse effects , BNT162 Vaccine/immunology , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Immunogenicity, Vaccine/immunology , Immunoglobulin A , Immunoglobulin G , Kidney Failure, Chronic/therapy , Peritoneal Dialysis/adverse effects , Renal Dialysis , SARS-CoV-2
19.
Nat Commun ; 13(1): 1237, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35246560

ABSTRACT

The BNT162b2 COVID-19 vaccine has been shown to reduce viral load of breakthrough infections (BTIs), an important factor affecting infectiousness. This viral-load protective effect has been waning with time post the second vaccine and later restored with a booster shot. It is currently unclear though for how long this regained effectiveness lasts. Analyzing Ct values of SARS-CoV-2 qRT-PCR tests of over 22,000 infections during a Delta-variant-dominant period in Israel, we find that this viral-load reduction effectiveness significantly declines within months post the booster dose. Adjusting for age, sex and calendric date, Ct values of RdRp gene initially increases by 2.7 [CI: 2.3-3.0] relative to unvaccinated in the first month post the booster dose, yet then decays to a difference of 1.3 [CI: 0.7-1.9] in the second month and becomes small and insignificant in the third to fourth months. The rate and magnitude of this post-booster decline in viral-load reduction effectiveness mirror those observed post the second vaccine. These results suggest rapid waning of the booster's effectiveness in reducing infectiousness, possibly affecting community-level spread of the virus.


Subject(s)
BNT162 Vaccine/immunology , COVID-19 Vaccines/immunology , COVID-19/immunology , Immunization, Secondary/methods , SARS-CoV-2/immunology , Viral Load/immunology , Adult , Algorithms , BNT162 Vaccine/administration & dosage , COVID-19/prevention & control , COVID-19/virology , COVID-19 Vaccines/administration & dosage , Female , Humans , Immunization, Secondary/statistics & numerical data , Immunogenicity, Vaccine/immunology , Linear Models , Male , Middle Aged , SARS-CoV-2/genetics , SARS-CoV-2/physiology , Time Factors , Treatment Outcome , Vaccination/methods , Vaccination/statistics & numerical data
20.
J Virol ; 96(7): e0220121, 2022 04 13.
Article in English | MEDLINE | ID: mdl-35266806

ABSTRACT

Respiratory syncytial virus (RSV) is a leading cause of severe lower respiratory tract disease of children, the elderly, and immunocompromised individuals. Currently, there are no FDA-approved RSV vaccines. The RSV G glycoprotein is used for viral attachment to host cells and impairment of host immunity by interacting with the human chemokine receptor CX3CR1. Antibodies that disrupt this interaction are protective against infection and disease. Nevertheless, development of an RSV G vaccine antigen has been hindered by its low immunogenicity and safety concerns. A previous study described three engineered RSV G proteins containing single-point mutations that induce higher levels of IgG antibodies and have improved safety profiles compared to wild-type RSV G (H. C. Bergeron, J. Murray, A. M. Nuñez Castrejon, et al., Viruses 13:352, 2021, https://doi.org/10.3390/v13020352). However, it is unclear if the mutations affect RSV G protein folding and display of its conformational epitopes. In this study, we show that the RSV G S177Q protein retains high-affinity binding to protective human and mouse monoclonal antibodies and has equal reactivity as wild-type RSV G protein to human reference immunoglobulin to RSV. Additionally, we determined the high-resolution crystal structure of RSV G S177Q protein in complex with the anti-RSV G antibody 3G12, further validating its antigenic structure. These studies show for the first time that an engineered RSV G protein with increased immunogenicity and safety retains conformational epitopes to high-affinity protective antibodies, supporting its further development as an RSV vaccine immunogen. IMPORTANCE Respiratory syncytial virus (RSV) causes severe lower respiratory diseases of children, the elderly, and immunocompromised populations. There currently are no FDA-approved RSV vaccines. Most vaccine development efforts have focused on the RSV F protein, and the field has generally overlooked the receptor-binding antigen RSV G due to its poor immunogenicity and safety concerns. However, single-point mutant RSV G proteins have been previously identified that have increased immunogenicity and safety. In this study, we investigate the antibody reactivities of three known RSV G mutant proteins. We show that one mutant RSV G protein retains high-affinity binding to protective monoclonal antibodies, is equally recognized by anti-RSV antibodies in human sera, and forms the same three-dimensional structure as the wild-type RSV G protein. Our study validates the structure-guided design of the RSV G protein as an RSV vaccine antigen.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Respiratory Syncytial Virus, Human , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Neutralizing/metabolism , Antigens, Viral/genetics , Antigens, Viral/immunology , Epitopes/genetics , Epitopes/immunology , Immunogenicity, Vaccine/genetics , Immunogenicity, Vaccine/immunology , Mice , Mutation , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus, Human/genetics , Respiratory Syncytial Virus, Human/immunology , Viral Fusion Proteins/genetics , Viral Fusion Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...