Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.496
Filter
1.
Euro Surveill ; 29(31)2024 Aug.
Article in English | MEDLINE | ID: mdl-39092531

ABSTRACT

BackgroundHealthcare personnel (HCP) are at high risk for respiratory infections through occupational exposure to respiratory viruses.AimWe used data from a prospective influenza vaccine effectiveness study in HCP to quantify the incidence of acute respiratory infections (ARI) and their associated presenteeism and absenteeism.MethodsAt the start and end of each season, HCP at two Israeli hospitals provided serum to screen for antibodies to influenza virus using the haemagglutination inhibition assay. During the season, active monitoring for the development of ARI symptoms was conducted twice a week by RT-PCR testing of nasal swabs for influenza and respiratory syncytial virus (RSV). Workplace presenteeism and absenteeism were documented. We calculated incidences of influenza- and RSV-associated ARI and applied sampling weights to make estimates representative of the source population.ResultsThe median age of 2,505 participating HCP was 41 years, and 70% were female. Incidence was 9.1 per 100 person-seasons (95% CI: 5.8-14.2) for RT-PCR-confirmed influenza and 2.5 per 100 person-seasons (95% CI: 0.9-7.1) for RSV illness. Each season, 18-23% of unvaccinated and influenza-negative HCP seroconverted. The incidence of seroconversion or RT-PCR-confirmed influenza was 27.5 per 100 person-seasons (95% CI: 17.8-42.5). Work during illness occurred in 92% (95% CI: 91-93) of ARI episodes, absence from work in 38% (95% CI: 36-40).ConclusionInfluenza virus and RSV infections and associated presenteeism and absenteeism were common among HCP. Improving vaccination uptake among HCP, infection control, and encouraging sick HCP to stay home are important strategies to reduce ARI incidence and decrease the risk of in-hospital transmission.


Subject(s)
Absenteeism , Health Personnel , Influenza, Human , Presenteeism , Respiratory Syncytial Virus Infections , Seasons , Humans , Respiratory Syncytial Virus Infections/epidemiology , Respiratory Syncytial Virus Infections/diagnosis , Respiratory Syncytial Virus Infections/virology , Influenza, Human/epidemiology , Influenza, Human/virology , Influenza, Human/diagnosis , Influenza, Human/prevention & control , Female , Incidence , Male , Health Personnel/statistics & numerical data , Israel/epidemiology , Adult , Presenteeism/statistics & numerical data , Middle Aged , Prospective Studies , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/virology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Respiratory Syncytial Viruses/isolation & purification , Respiratory Syncytial Virus, Human/isolation & purification , Respiratory Syncytial Virus, Human/genetics , Occupational Exposure/statistics & numerical data , Hemagglutination Inhibition Tests
2.
Clin Epigenetics ; 16(1): 114, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169387

ABSTRACT

BACKGROUND: The effect of vaccination on the epigenome remains poorly characterized. In previous research, we identified an association between seroprotection against influenza and DNA methylation at sites associated with the RIG-1 signaling pathway, which recognizes viral double-stranded RNA and leads to a type I interferon response. However, these studies did not fully account for confounding factors including age, gender, and BMI, along with changes in cell-type composition. RESULTS: Here, we studied the influenza vaccine response in a longitudinal cohort vaccinated over two consecutive years (2019-2020 and 2020-2021), using peripheral blood mononuclear cells and a targeted DNA methylation approach. To address the effects of multiple factors on the epigenome, we designed a multivariate multiple regression model that included seroprotection levels as quantified by the hemagglutination-inhibition (HAI) assay test. CONCLUSIONS: Our findings indicate that 179 methylation sites can be combined as potential signatures to predict seroprotection. These sites were not only enriched for genes involved in the regulation of the RIG-I signaling pathway, as found previously, but also enriched for other genes associated with innate immunity to viruses and the transcription factor binding sites of BRD4, which is known to impact T cell memory. We propose a model to suggest that the RIG-I pathway and BRD4 could potentially be modulated to improve immunization strategies.


Subject(s)
DNA Methylation , Immunity, Innate , Influenza Vaccines , Influenza, Human , Humans , DNA Methylation/genetics , DNA Methylation/drug effects , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Immunity, Innate/genetics , Female , Male , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza, Human/genetics , Middle Aged , Adult , Signal Transduction , T-Lymphocytes/immunology , Longitudinal Studies , Epigenesis, Genetic , Vaccination , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism
3.
Nat Commun ; 15(1): 6603, 2024 Aug 04.
Article in English | MEDLINE | ID: mdl-39097574

ABSTRACT

Vaccine responsiveness is often reduced in older adults. Yet, our lack of understanding of low vaccine responsiveness hampers the development of effective vaccination strategies to reduce the impact of infectious diseases in the ageing population. Young-adult (25-49 y), middle-aged (50-64 y) and older-adult ( ≥ 65 y) participants of the VITAL clinical trials (n = 315, age-range: 28-98 y), were vaccinated with an annual (2019-2020) quadrivalent influenza (QIV) booster vaccine, followed by a primary 13-valent pneumococcal-conjugate (PCV13) vaccine (summer/autumn 2020) and a primary series of two SARS-CoV-2 mRNA-1273 vaccines (spring 2021). This unique setup allowed investigation of humoral responsiveness towards multiple vaccines within the same individuals over the adult age-range. Booster QIV vaccination induced comparable H3N2 hemagglutination inhibition (HI) titers in all age groups, whereas primary PCV13 and mRNA-1273 vaccination induced lower antibody concentrations in older as compared to younger adults (primary endpoint). The persistence of humoral responses, towards the 6 months timepoint, was shorter in older adults for all vaccines (secondary endpoint). Interestingly, highly variable vaccine responder profiles overarching multiple vaccines were observed. Yet, approximately 10% of participants, mainly comprising of older male adults, were classified as low responders to multiple vaccines. This study aids the identification of risk groups for low vaccine responsiveness and hence supports targeted vaccination strategies. Trial number: NL69701.041.19, EudraCT: 2019-000836-24.


Subject(s)
2019-nCoV Vaccine mRNA-1273 , Antibodies, Viral , COVID-19 , Immunity, Humoral , Immunization, Secondary , Influenza Vaccines , Influenza, Human , Pneumococcal Vaccines , SARS-CoV-2 , Humans , Middle Aged , Adult , Aged , Male , Female , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Antibodies, Viral/immunology , Antibodies, Viral/blood , Immunity, Humoral/immunology , Pneumococcal Vaccines/immunology , Pneumococcal Vaccines/administration & dosage , COVID-19/prevention & control , COVID-19/immunology , SARS-CoV-2/immunology , Aged, 80 and over , 2019-nCoV Vaccine mRNA-1273/immunology , Influenza, Human/prevention & control , Influenza, Human/immunology , Age Factors , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Influenza A Virus, H3N2 Subtype/immunology , Vaccination , Hemagglutination Inhibition Tests
4.
Nat Commun ; 15(1): 6802, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122688

ABSTRACT

Influenza virus infection remains a major global health problem and requires a universal vaccine with broad protection against different subtypes as well as a rapid-response vaccine to provide immediate protection in the event of an epidemic outbreak. Here, we show that intranasal administration of probiotic Escherichia coli Nissle 1917 activates innate immunity in the respiratory tract and provides immediate protection against influenza virus infection within 1 day. Based on this vehicle, a recombinant strain is engineered to express and secret five tandem repeats of the extracellular domain of matrix protein 2 from different influenza virus subtypes. Intranasal vaccination with this strain induces durable humoral and mucosal responses in the respiratory tract, and provides broad protection against the lethal challenge of divergent influenza viruses in female BALB/c mice. Our findings highlight a promising delivery platform for developing mucosal vaccines that provide immediate and sustained protection against respiratory pathogens.


Subject(s)
Administration, Intranasal , Escherichia coli , Influenza Vaccines , Mice, Inbred BALB C , Orthomyxoviridae Infections , Probiotics , Animals , Escherichia coli/genetics , Probiotics/administration & dosage , Female , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Mice , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Influenza A virus/immunology , Influenza A virus/genetics , Viral Matrix Proteins/genetics , Viral Matrix Proteins/immunology , Immunity, Innate , Immunity, Mucosal , Humans , Antibodies, Viral/immunology , Viroporin Proteins
5.
Int J Mol Sci ; 25(15)2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39126117

ABSTRACT

Avian influenza virus has been long considered the main threat for a future pandemic. Among the possible avian influenza virus subtypes, A(H5N1) clade 2.3.4.4b is becoming enzootic in mammals, representing an alarming step towards a pandemic. In particular, genotype B3.13 has recently caused an outbreak in US dairy cattle. Since pandemic preparedness is largely based on the availability of prepandemic candidate vaccine viruses, in this review we will summarize the current status of the enzootics, and challenges for H5 vaccine manufacturing and delivery.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza Vaccines , Influenza in Birds , Animals , Influenza Vaccines/immunology , Humans , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/genetics , Influenza in Birds/prevention & control , Influenza in Birds/virology , Influenza in Birds/epidemiology , Influenza, Human/prevention & control , Influenza, Human/epidemiology , Influenza, Human/immunology , Influenza, Human/virology , Birds/virology , Pandemics/prevention & control
7.
PLoS Pathog ; 20(8): e1012393, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39116029

ABSTRACT

Live-attenuated influenza vaccines (LAIV) offer advantages over the commonly used inactivated split influenza vaccines. However, finding the optimal balance between sufficient attenuation and immunogenicity has remained a challenge. We recently developed an alternative LAIV based on the 2009 pandemic H1N1 virus with a truncated NS1 protein and lacking PA-X protein expression (NS1(1-126)-ΔPAX). This virus showed a blunted replication and elicited a strong innate immune response. In the present study, we evaluated the efficacy of this vaccine candidate in the porcine animal model as a pertinent in vivo system. Immunization of pigs via the nasal route with the novel NS1(1-126)-ΔPAX LAIV did not cause disease and elicited a strong mucosal immune response that completely blocked replication of the homologous challenge virus in the respiratory tract. However, we observed prolonged shedding of our vaccine candidate from the upper respiratory tract. To improve LAIV safety, we developed a novel prime/boost vaccination strategy combining primary intramuscular immunization with a haemagglutinin-encoding propagation-defective vesicular stomatitis virus (VSV) replicon, followed by a secondary immunization with the NS1(1-126)-ΔPAX LAIV via the nasal route. This two-step immunization procedure significantly reduced LAIV shedding, increased the production of specific serum IgG, neutralizing antibodies, and Th1 memory cells, and resulted in sterilizing immunity against homologous virus challenge. In conclusion, our novel intramuscular prime/intranasal boost regimen interferes with virus shedding and transmission, a feature that will help combat influenza epidemics and pandemics.


Subject(s)
Administration, Intranasal , Influenza Vaccines , Orthomyxoviridae Infections , Animals , Swine , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Injections, Intramuscular , Vaccines, Attenuated/immunology , Vaccines, Attenuated/administration & dosage , Influenza A Virus, H1N1 Subtype/immunology , Disease Models, Animal , Antibodies, Viral/immunology , Immunization, Secondary/methods , Vaccination/methods , Influenza, Human/prevention & control , Influenza, Human/immunology
8.
Hum Vaccin Immunother ; 20(1): 2384180, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39106971

ABSTRACT

During the coronavirus disease 2019 (COVID-19) pandemic, scheduled vaccinations were postponed, mass vaccination programmes were suspended and opportunities for healthcare workers to administer vaccines ad hoc decreased. The aims of this systematic literature review were to determine the impact of the COVID-19 pandemic on vaccine confidence, intent and uptake in preexisting routine childhood or adult vaccination programmes, and to identify factors associated with changes in acceptance, intent and uptake of preexisting vaccines. Medline and Embase were searched for studies in Australia, Brazil, Canada, China, Japan, the USA, and European countries, published between 1 January 2021 and 4 August 2022. A complementary gray literature search was conducted between 11 and 13 October 2022, and supplemented with additional gray research in October 2023. In total, 54 citations were included in the review. Study design and geography were heterogeneous. The number of adults who received or intended to receive an influenza or pneumococcal vaccine was higher during the pandemic than in previous seasons (n = 28 studies). In addition, increased acceptance of adult vaccinations was observed during 2020-21 compared with 2019-20 (n = 12 studies). The rates of childhood vaccinations decreased during the COVID-19 pandemic across several countries (n = 11 studies). Factors associated with changes in intention to receive a vaccination, or uptake of influenza vaccine, included previous vaccination, older age, higher perceived risk of contracting COVID-19, anxiety regarding the pandemic and fear of contracting COVID-19. Acceptance and uptake of influenza and pneumococcal vaccines generally increased after onset of the COVID-19 pandemic.


Subject(s)
COVID-19 , Humans , COVID-19/prevention & control , COVID-19/epidemiology , Vaccination/psychology , Vaccination/statistics & numerical data , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Adult , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Immunization Programs , Child , SARS-CoV-2/immunology , Vaccination Hesitancy/statistics & numerical data , Vaccination Hesitancy/psychology , Pneumococcal Vaccines/administration & dosage , Pandemics/prevention & control , Patient Acceptance of Health Care/statistics & numerical data , Patient Acceptance of Health Care/psychology
9.
Clin Immunol ; 266: 110333, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39089348

ABSTRACT

Understanding the molecular mechanisms underpinning diverse vaccination responses is critical for developing efficient vaccines. Molecular subtyping can offer insights into heterogeneous nature of responses and aid in vaccine design. We analyzed multi-omic data from 62 haemagglutinin seasonal influenza vaccine recipients (2019-2020), including transcriptomics, proteomics, glycomics, and metabolomics data collected pre-vaccination. We performed a subtyping analysis on the integrated data revealing five subtypes with distinct molecular signatures. These subtypes differed in the expression of pre-existing adaptive or innate immunity signatures, which were linked to significant variation in baseline immunoglobulin A (IgA) and hemagglutination inhibition (HAI) titer levels. It is worth noting that these differences persisted through day 28 post-vaccination, indicating the effect of initial immune state on vaccination response. These findings highlight the significance of interpersonal variation in baseline immune status as a crucial factor in determining the effectiveness of seasonal vaccines. Ultimately, incorporating molecular profiling could enable personalized vaccine optimization.


Subject(s)
Antibodies, Viral , Influenza Vaccines , Influenza, Human , Multiomics , Vaccination , Humans , Adaptive Immunity/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Antibody Formation/immunology , Hemagglutination Inhibition Tests , Immunity, Innate/immunology , Immunoglobulin A/immunology , Immunoglobulin A/blood , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Influenza, Human/immunology , Influenza, Human/prevention & control , Proteomics/methods , Seasons
10.
Emerg Microbes Infect ; 13(1): 2389095, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39101691

ABSTRACT

Influenza virus infection poses a continual menace to public health. Here, we developed soluble trimeric HA ectodomain vaccines by establishing interprotomer disulfide bonds in the stem region, which effectively preserve the native antigenicity of stem epitopes. The stable trimeric H1 ectodomain proteins exhibited higher thermal stabilities in comparison with unmodified HAs and showed strong binding activities towards a panel of anti-stem cross-reactive antibodies that recognize either interprotomer or intraprotomer epitopes. Negative stain transmission electron microscopy (TEM) analysis revealed the stable trimer architecture of the interprotomer disulfide-stapled WA11#5, NC99#2, and FLD#1 proteins as well as the irregular aggregation of unmodified HA molecules. Immunizations of mice with those trimeric HA ectodomain vaccines formulated with incomplete Freund's adjuvant elicited significantly more potent cross-neutralizing antibody responses and offered broader immuno-protection against lethal infections with heterologous influenza strains compared to unmodified HA proteins. Additionally, the findings of our study indicate that elevated levels of HA stem-specific antibody responses correlate with strengthened cross-protections. Our design strategy has proven effective in trimerizing HA ectodomains derived from both influenza A and B viruses, thereby providing a valuable reference for designing future influenza HA immunogens.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , Disulfides , Hemagglutinin Glycoproteins, Influenza Virus , Influenza Vaccines , Mice, Inbred BALB C , Orthomyxoviridae Infections , Animals , Influenza Vaccines/immunology , Influenza Vaccines/genetics , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Antibodies, Viral/immunology , Mice , Disulfides/chemistry , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/virology , Antibodies, Neutralizing/immunology , Female , Cross Protection/immunology , Cross Reactions , Humans , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza, Human/virology , Epitopes/immunology , Epitopes/genetics , Epitopes/chemistry , Protein Multimerization , Influenza B virus/immunology , Influenza B virus/genetics , Influenza B virus/chemistry
11.
Vaccine ; 42(21): 126182, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39116486

ABSTRACT

OBJECTIVES: This study was to assess the lot-to-lot consistency, immunogenicity and safety of three manufacturing lots of a quadrivalent inactivated influenza vaccine (IIV4). METHODS: A randomized, double-blind, phase IV clinical trial was conducted in healthy children, adolescents and adults aged 9-59 years in Guizhou Province, China. Eligible participants were enrolled and randomized into three groups in a ratio of 1:1:1 to receive a single dose of one of three manufacturing lots of IIV4. Serum samples were collected before and 28 days after vaccination for hemagglutination inhibition (HI) antibody testing. Safety data were collected for up to 28 days after vaccination. The primary objective was to evaluate the lot-to-lot consistency of immune response as assessed by the geometric mean titer (GMT) of HI antibody at 28 days after vaccination. RESULTS: Between November 27, 2022 and December 18, 2022, 1260 eligible participants were enrolled, with similar participant demographics among groups. Immune responses after vaccination were comparable across groups, with the 95% confidence intervals (CIs) of GMT ratios for all 4 strains falling into the equivalence criterion of (0.67, 1.5). The seroconversion rates (SCRs) and seroprotection rates (SPRs) met the US Center or Biologics Evaluation and Research (CBER) criteria for all strains for each lot (lower limit of 95% CI of SCR ≥ 40% and SPR ≥ 70%). The incidences of solicited and unsolicited adverse reactions were similar among three groups, most of which (91.9%) were mild or moderate in severity. A total of 11 serious adverse events were reported during the study, and all were considered unrelated to vaccination. CONCLUSION: The three manufacturing lots of IIV4 demonstrated consistent immunogenicity. IIV4 can elicit satisfactory immune responses for all four strains and no safety concerns were identified. CLINICAL TRIAL REGISTRATION: Identifier No. NCT05512494.


Subject(s)
Antibodies, Viral , Hemagglutination Inhibition Tests , Immunogenicity, Vaccine , Influenza Vaccines , Influenza, Human , Vaccines, Inactivated , Humans , Influenza Vaccines/immunology , Influenza Vaccines/adverse effects , Influenza Vaccines/administration & dosage , Adolescent , Male , Female , Double-Blind Method , Child , Adult , Young Adult , Vaccines, Inactivated/immunology , Vaccines, Inactivated/adverse effects , Vaccines, Inactivated/administration & dosage , Antibodies, Viral/blood , Influenza, Human/prevention & control , Influenza, Human/immunology , Middle Aged , China , Healthy Volunteers , Vaccination/methods
12.
Proc Natl Acad Sci U S A ; 121(35): e2401058121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39163333

ABSTRACT

B cell receptors (BCRs) play a crucial role in recognizing and fighting foreign antigens. High-throughput sequencing enables in-depth sampling of the BCRs repertoire after immunization. However, only a minor fraction of BCRs actively participate in any given infection. To what extent can we accurately identify antigen-specific sequences directly from BCRs repertoires? We present a computational method grounded on sequence similarity, aimed at identifying statistically significant responsive BCRs. This method leverages well-known characteristics of affinity maturation and expected diversity. We validate its effectiveness using longitudinally sampled human immune repertoire data following influenza vaccination and SARS-CoV-2 infections. We show that different lineages converge to the same responding Complementarity Determining Region 3, demonstrating convergent selection within an individual. The outcomes of this method hold promise for application in vaccine development, personalized medicine, and antibody-derived therapeutics.


Subject(s)
COVID-19 , Receptors, Antigen, B-Cell , SARS-CoV-2 , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, B-Cell/genetics , Humans , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , SARS-CoV-2/immunology , Influenza Vaccines/immunology , Immunization/methods , Complementarity Determining Regions/genetics , Complementarity Determining Regions/immunology , B-Lymphocytes/immunology , Vaccination , Influenza, Human/immunology , Influenza, Human/prevention & control , Computational Biology/methods , High-Throughput Nucleotide Sequencing
13.
Ann Med ; 56(1): 2392882, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39155852

ABSTRACT

BACKGROUND: Systemic lupus erythematosus (SLE), an extensive autoimmune disorder, compromises viral resistance and alters immune responses post respiratory virus vaccines. This study aims to assess immune response levels and safety in SLE patients following respiratory virus vaccines. METHODS: Extensive searches, until 1 March 2024, were conducted using PubMed, EMBASE, and Cochrane Library. Outcomes, encompassing seroconversion rate (SCR), antibody and IgG titers, neutralizing antibodies, anti-spike antibodies, anti-receptor binding domain (RBD) IgG, and adverse events, were appraised. RESULTS: Sixteen articles, comprising 25 observational studies, were included. SLE patients exhibited lower SCR (OR = 0.42, 95%CI: 0.26 to 0.69), antibody titers (SMD=-2.84, 95%CI: -3.36 to -1.61), and neutralizing antibodies (OR = 0.27, 95%CI: 0.13 to 0.56) compared to the healthy population post respiratory virus vaccines. Notably, differences were statistically insignificant for anti-RBD IgG (OR = 1.75, 95%CI: 0.10 to 29.42), IgG titers (SMD=-2.54, 95%CI: -5.57 to -0.49), anti-spike antibodies (OR = 0.35, 95%CI: 0.08 to 1.53), injection site discomfort (OR = 1.03, 95%CI: 0.52 to 2.06), fatigue (OR = 1.23, 95%CI: 0.74 to 2.03), fever (OR = 1.02, 95%CI: 0.64 to 1.63), localized reactions (OR = 0.69, 95%CI: 0.37 to 1.30), systemic reactions (OR = 1.00, 95%CI: 0.59 to 1.69), allergic reactions (OR = 5.11, 95%CI: 0.24 to 107.10), self-reported vaccination-related adverse events (OR = 1.61, 95%CI: 0.56 to 4.63), and disease flares after vaccination (OR = 1.00, 95%CI: 0.14 to 7.28). CONCLUSION: Despite the reduced immune response and host protection in SLE patients post-Corona Virus Disease 2019 (COVID-19) and influenza vaccines compared to the healthy population, safety profiles are comparable. Therefore, it is recommended that SLE patients receive COVID-19 and influenza viral vaccines to fortify their resistance.


Subject(s)
Antibodies, Viral , Immunity, Humoral , Lupus Erythematosus, Systemic , Observational Studies as Topic , Humans , Lupus Erythematosus, Systemic/immunology , Immunity, Humoral/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Immunoglobulin G/blood , Immunoglobulin G/immunology , COVID-19/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , Female , Male , Influenza Vaccines/immunology , Influenza Vaccines/adverse effects , Influenza Vaccines/administration & dosage
14.
EBioMedicine ; 106: 105269, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39111250

ABSTRACT

BACKGROUND: Influenza viruses pose a persistent threat to global public health, necessitating the development of innovative and broadly effective vaccines. METHODS: This study focuses on a multiepitope vaccine (MEV) designed to provide broad-spectrum protection against different influenza viruses. The MEV, containing 19 B-cell linear epitopes, 7 CD4+ T cells, and 11 CD8+ T cells epitopes identified through enzyme-linked immunospot assay (ELISPOT) in influenza viruses infected mice, was administered through a regimen of two doses of DNA vaccine followed by one dose of a protein vaccine in C57BL/6 female mice. FINDINGS: Upon lethal challenge with both seasonal circulating strains (H1N1, H3N2, BV, and BY) and historical strains (H1N1-PR8 and H3N2-X31), MEV demonstrated substantial protection against different influenza seasonal strains, with partial efficacy against historical strains. Notably, the increased germinal centre B cells and antibody-secreting cells, along with robust T cell immune responses, highlighted the comprehensive immune defence elicited by MEV. Elevated hemagglutinin inhibition antibody was also observed against seasonal circulating and historical strains. Additionally, mice vaccinated with MEV exhibited significantly lower counts of inflammatory cells in the lungs compared to negative control groups. INTERPRETATION: Our results demonstrated the efficacy of a broad-spectrum MEV against influenza viruses in mice. Conducting long-term studies to evaluate the durability of MEV-induced immune responses and explore its potential application in diverse populations will offer valuable insights for the continued advancement of this promising vaccine. FUNDING: Funding bodies are described in the Acknowledgments section.


Subject(s)
Epitopes, B-Lymphocyte , Influenza B virus , Influenza Vaccines , Orthomyxoviridae Infections , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Mice , Influenza B virus/immunology , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae Infections/immunology , Female , Epitopes, B-Lymphocyte/immunology , Influenza A virus/immunology , Antibodies, Viral/immunology , Epitopes, T-Lymphocyte/immunology , Disease Models, Animal , Mice, Inbred C57BL , Vaccines, DNA/immunology , Vaccines, DNA/administration & dosage , Seasons , Influenza A Virus, H3N2 Subtype/immunology , Humans
15.
Dtsch Med Wochenschr ; 149(17): 1045-1049, 2024 Aug.
Article in German | MEDLINE | ID: mdl-39146753

ABSTRACT

SARS-COV-2 : During the COVID-19 pandemic, mRNA-based vaccines were approved for the first time. The mRNA encodes for the viral spike protein, leading to the development of specific antibodies and T-cells, providing effective protection against severe illness and death from COVID-19. New variants regularly emerge due to rapid viral evolution. Available COVID-19 vaccines can be adapted to circulating variants. For the upcoming winter season, adapted vaccines against the Omicron sublineage JN.1 have been recommended. SEASONAL INFLUENZA : Seasonal influenza viruses change significantly with regard to their antigenic properties each season, necessitating updated vaccines. The WHO predicts and recommends which genetic variants should be included in the vaccine. Quadrivalent vaccines were recommended previously, but for 2024/2025, trivalent vaccines are advised as the Influenza B/Yamagata lineage has not circulated since 2020. For all people over 60 years of age, a high-dose vaccine is recommended, showing slightly improved efficacy.


Subject(s)
COVID-19 Vaccines , COVID-19 , Influenza Vaccines , Influenza, Human , Humans , COVID-19/prevention & control , COVID-19/immunology , Influenza, Human/prevention & control , Influenza, Human/immunology , Influenza Vaccines/immunology , Influenza Vaccines/therapeutic use , COVID-19 Vaccines/immunology , SARS-CoV-2/immunology , Pandemics/prevention & control , Vaccination
17.
Viruses ; 16(7)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39066267

ABSTRACT

The aim of this study was to determine the level of anti-hemagglutinin antibodies in blood sera collected from patients during the 2022/2023 epidemic season in Poland. A total of 700 sera samples from patients across the country were tested. The samples were divided into seven groups according to the age of the patients, with 100 samples from each age group. The hemagglutination inhibition test (OZHA) was used to determine the level of anti-hemagglutinin antibodies. The test results have confirmed the presence of anti-hemagglutinin antibodies for antigens A/Victoria/2570/2019 (H1N1)pdm09, A/Darwin/9/2021 (H3N2), B/Austria/1359417/2021 (B/Yamagata lineage) and B/ Phuket/3073/2013 (B/Victoria lineage) present in the influenza vaccine recommended by the World Health Organization (WHO) for the 2022/2023 epidemic season. The highest geometric mean antibody titres (GMT) and protection rate values (%) were recorded for hemagglutinin A/H3N2. In Poland, in the 2022/2023 epidemic season, the percentage of the population vaccinated against influenza was 5.7%. Therefore, the test results can be interpreted as the response of the immune system in patients who have been previously infected with an influenza virus.


Subject(s)
Antibodies, Viral , Hemagglutination Inhibition Tests , Hemagglutinin Glycoproteins, Influenza Virus , Influenza A Virus, H3N2 Subtype , Influenza, Human , Humans , Poland/epidemiology , Influenza, Human/immunology , Influenza, Human/epidemiology , Influenza, Human/virology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Adult , Middle Aged , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Adolescent , Young Adult , Male , Influenza A Virus, H3N2 Subtype/immunology , Female , Child , Child, Preschool , Influenza Vaccines/immunology , Influenza A Virus, H1N1 Subtype/immunology , Aged , Epidemics , Seasons , Infant , Influenza B virus/immunology
18.
Front Immunol ; 15: 1376395, 2024.
Article in English | MEDLINE | ID: mdl-38975350

ABSTRACT

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.


Subject(s)
Antibodies, Viral , Immunity, Mucosal , Influenza A virus , Influenza Vaccines , Mice, Inbred BALB C , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus Vaccines , Animals , Mice , Respiratory Syncytial Virus Infections/prevention & control , Respiratory Syncytial Virus Infections/immunology , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Respiratory Syncytial Virus Vaccines/immunology , Respiratory Syncytial Virus Vaccines/administration & dosage , Antibodies, Viral/immunology , Influenza A virus/immunology , Female , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Respiratory Syncytial Viruses/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Vaccines, Combined/immunology , Vaccines, Combined/administration & dosage , Humans , Adenoviridae/immunology , Adenoviridae/genetics , Genetic Vectors
19.
Am J Reprod Immunol ; 92(1): e13896, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38994889

ABSTRACT

BACKGROUND: Recent data in nonpregnant individuals suggest a protective effect of influenza vaccination against SARS-CoV-2 infection and its severity. OBJECTIVES: Our primary objective was to evaluate whether influenza vaccination was associated with COVID-19 severity and pregnancy and neonatal outcomes among those infected with SARS-CoV-2. The secondary objective was to examine the association between influenza vaccination and SARS-CoV-2 infection. STUDY DESIGN: Secondary analysis of a multicenter retrospective cohort of pregnant people who tested positive for SARS-CoV-2 between March and August 2020, and a cohort of random deliveries during the same time period. The associations between 2019 influenza vaccination and the primary outcome of moderate-to-critical COVID-19 as well as maternal and perinatal outcomes were examined among all people who tested positive for SARS-CoV-2 between March and August 2020. The association between 2019 influenza vaccination and having a positive SARS-CoV-2 test was examined among a cohort of individuals who delivered on randomly selected dates between March and August 2020. Univariable and multivariable analyses were performed. RESULTS: Of 2325 people who tested positive for SARS-CoV-2, 1068 (45.9%) were vaccinated against influenza in 2019. Those who received the influenza vaccine were older, leaner, more likely to have private insurance, and identify as White or Hispanic. They were less likely to smoke tobacco and identify as Black. Overall, 419 (18.0%) had moderate, 193 (8.3%) severe, and 52 (2.2%) critical COVID-19. There was no association between influenza vaccination and moderate-to-critical COVID-19 (29.2% vs. 28.0%, adjusted OR 1.10, 95% CI 0.90-1.34) or adverse maternal and perinatal outcomes among those who tested positive. Of 8152 people who delivered in 2020, 4658 (57.1%) received the influenza vaccine. Prior vaccination was not associated with a difference in the odds of SARS-CoV-2 infection (3.8% vs. 4.2%, adjusted OR 0.94, 95% CI 0.74-1.19). CONCLUSION: Prior influenza vaccination was not associated with decreased severity of COVID-19 or lower odds of SARS-CoV-2 infection in pregnancy.


Subject(s)
COVID-19 , Influenza Vaccines , Influenza, Human , Pregnancy Complications, Infectious , SARS-CoV-2 , Vaccination , Humans , Female , Pregnancy , COVID-19/prevention & control , COVID-19/epidemiology , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , Adult , Retrospective Studies , SARS-CoV-2/immunology , Pregnancy Complications, Infectious/epidemiology , Pregnancy Complications, Infectious/prevention & control , Influenza, Human/prevention & control , Influenza, Human/epidemiology , Pregnancy Outcome , Infant, Newborn , Young Adult , Severity of Illness Index
20.
Nat Commun ; 15(1): 6007, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030218

ABSTRACT

An influenza vaccine approach that overcomes the problem of viral sequence diversity and provides long-lived heterosubtypic protection is urgently needed to protect against pandemic influenza viruses. Here, to determine if lung-resident effector memory T cells induced by cytomegalovirus (CMV)-vectored vaccines expressing conserved internal influenza antigens could protect against lethal influenza challenge, we immunize Mauritian cynomolgus macaques (MCM) with cynomolgus CMV (CyCMV) vaccines expressing H1N1 1918 influenza M1, NP, and PB1 antigens (CyCMV/Flu), and challenge with heterologous, aerosolized avian H5N1 influenza. All six unvaccinated MCM died by seven days post infection with acute respiratory distress, while 54.5% (6/11) CyCMV/Flu-vaccinated MCM survived. Survival correlates with the magnitude of lung-resident influenza-specific CD4 + T cells prior to challenge. These data demonstrate that CD4 + T cells targeting conserved internal influenza proteins can protect against highly pathogenic heterologous influenza challenge and support further exploration of effector memory T cell-based vaccines for universal influenza vaccine development.


Subject(s)
CD4-Positive T-Lymphocytes , Cytomegalovirus , Influenza A Virus, H1N1 Subtype , Influenza Vaccines , Macaca fascicularis , Animals , Influenza Vaccines/immunology , Influenza Vaccines/administration & dosage , CD4-Positive T-Lymphocytes/immunology , Influenza A Virus, H1N1 Subtype/immunology , Cytomegalovirus/immunology , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Influenza A Virus, H5N1 Subtype/immunology , Lung/immunology , Lung/virology , Lung/pathology , Genetic Vectors/genetics , Genetic Vectors/immunology , Male , Female , Memory T Cells/immunology , Immunologic Memory/immunology , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL