Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.983
Filter
1.
J Exp Med ; 221(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38949639

ABSTRACT

Children resist COVID-19, and previous studies reported increased innate immunity in their upper airways. A new paper by Watkins et al. (https://doi.org/10.1084/jem.20230911) shows that the nasal mucosa of children is characterized by often asymptomatic viral and/or bacterial infections that dynamically regulate distinct innate immune programs.


Subject(s)
COVID-19 , Immunity, Innate , Nasal Mucosa , SARS-CoV-2 , Humans , COVID-19/immunology , Child , Immunity, Innate/immunology , SARS-CoV-2/immunology , Nasal Mucosa/virology , Nasal Mucosa/immunology
3.
Sci Adv ; 10(27): eadg3747, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38959314

ABSTRACT

Vaccination can help prevent infection and can also be used to treat cancer, allergy, and potentially even drug overdose. Adjuvants enhance vaccine responses, but currently, the path to their advancement and development is incremental. We used a phenotypic small-molecule screen using THP-1 cells to identify nuclear factor-κB (NF-κB)-activating molecules followed by counterscreening lead target libraries with a quantitative tumor necrosis factor immunoassay using primary human peripheral blood mononuclear cells. Screening on primary cells identified an imidazopyrimidine, dubbed PVP-037. Moreover, while PVP-037 did not overtly activate THP-1 cells, it demonstrated broad innate immune activation, including NF-κB and cytokine induction from primary human leukocytes in vitro as well as enhancement of influenza and SARS-CoV-2 antigen-specific humoral responses in mice. Several de novo synthesis structural enhancements iteratively improved PVP-037's in vitro efficacy, potency, species-specific activity, and in vivo adjuvanticity. Overall, we identified imidazopyrimidine Toll-like receptor-7/8 adjuvants that act in synergy with oil-in-water emulsion to enhance immune responses.


Subject(s)
Adjuvants, Immunologic , Pyrimidines , Toll-Like Receptor 7 , Toll-Like Receptor 8 , Humans , Toll-Like Receptor 8/agonists , Toll-Like Receptor 8/metabolism , Animals , Mice , Adjuvants, Immunologic/pharmacology , Toll-Like Receptor 7/agonists , Pyrimidines/pharmacology , Pyrimidines/chemistry , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , Imidazoles/pharmacology , Imidazoles/chemistry , THP-1 Cells , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/immunology , COVID-19/virology , COVID-19/immunology , NF-kappa B/metabolism , Female , Drug Discovery/methods , Immunity, Innate/drug effects
4.
J Med Virol ; 96(7): e29778, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38965882

ABSTRACT

Active and passive immunization is used in high-risk patients to prevent severe courses of COVID-19, but the impact of prophylactic neutralizing antibodies on the immune reaction to the mRNA vaccines has remained enigmatic. Here we show that CD4 T and B cell responses to Spikevax booster immunization are suppressed by the therapeutic antibodies Casirivimab and Imdevimab. B cell and T cell responses were significantly induced in controls but not in antibody-treated patients. The data indicates that humoral immunity, i. e. high levels of antibodies, negatively impacts reactive immunity, resulting in blunted cellular responses upon boosting. This argues for temporal separation of vaccination efforts; with active vaccination preferably applied before prophylactic therapeutic antibody treatment.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , B-Lymphocytes , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , COVID-19/prevention & control , COVID-19/immunology , B-Lymphocytes/immunology , Spike Glycoprotein, Coronavirus/immunology , Antibodies, Viral/immunology , Antibodies, Viral/blood , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , COVID-19 Vaccines/immunology , SARS-CoV-2/immunology , Middle Aged , Male , Female , Vaccination , Adult , Aged , CD4-Positive T-Lymphocytes/immunology , T-Lymphocytes/immunology , Immunization, Secondary , Immunity, Humoral , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use
5.
Front Public Health ; 12: 1353415, 2024.
Article in English | MEDLINE | ID: mdl-38966699

ABSTRACT

Background: The protective effectiveness provided by naturally acquired immunity against SARS-CoV-2 reinfection remain controversial. Objective: To systematically evaluate the protective effect of natural immunity against subsequent SARS-CoV-2 infection with different variants. Methods: We searched for related studies published in seven databases before March 5, 2023. Eligible studies included in the analysis reported the risk of subsequent infection for groups with or without a prior SARS-CoV-2 infection. The primary outcome was the overall pooled incidence rate ratio (IRR) of SARS-CoV-2 reinfection/infection between the two groups. We also focused on the protective effectiveness of natural immunity against reinfection/infection with different SARS-CoV-2 variants. We used a random-effects model to pool the data, and obtained the bias-adjusted results using the trim-and-fill method. Meta-regression and subgroup analyses were conducted to explore the sources of heterogeneity. Sensitivity analysis was performed by excluding included studies one by one to evaluate the stability of the results. Results: We identified 40 eligible articles including more than 20 million individuals without the history of SARS-CoV-2 vaccination. The bias-adjusted efficacy of naturally acquired antibodies against reinfection was estimated at 65% (pooled IRR = 0.35, 95% CI = 0.26-0.47), with higher efficacy against symptomatic COVID-19 cases (pooled IRR = 0.15, 95% CI = 0.08-0.26) than asymptomatic infection (pooled IRR = 0.40, 95% CI = 0.29-0.54). Meta-regression revealed that SARS-CoV-2 variant was a statistically significant effect modifier, which explaining 46.40% of the variation in IRRs. For different SARS-CoV-2 variant, the pooled IRRs for the Alpha (pooled IRR = 0.11, 95% CI = 0.06-0.19), Delta (pooled IRR = 0.19, 95% CI = 0.15-0.24) and Omicron (pooled IRR = 0.61, 95% CI = 0.42-0.87) variant were higher and higher. In other subgroup analyses, the pooled IRRs of SARS-CoV-2 infection were statistically various in different countries, publication year and the inclusion end time of population, with a significant difference (p = 0.02, p < 0.010 and p < 0.010), respectively. The risk of subsequent infection in the seropositive population appeared to increase slowly over time. Despite the heterogeneity in included studies, sensitivity analyses showed stable results. Conclusion: Previous SARS-CoV-2 infection provides protection against pre-omicron reinfection, but less against omicron. Ongoing viral mutation requires attention and prevention strategies, such as vaccine catch-up, in conjunction with multiple factors.


Subject(s)
COVID-19 , Reinfection , SARS-CoV-2 , Humans , COVID-19/prevention & control , COVID-19/epidemiology , COVID-19/immunology , SARS-CoV-2/immunology , Immunity, Innate
6.
Sci Rep ; 14(1): 15515, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38969667

ABSTRACT

Vaccine hesitancy is an inevitable risk for societies as it contributes to outbreaks of diseases. Prior research suggests that vaccination decisions of individuals tend to spread within social networks, resulting in a tendency to vaccination homophily. The clustering of individuals resistant to vaccination can substantially make the threshold necessary to achieve herd immunity harder to reach. In this study, we examined the extent of vaccination homophily among social contacts and its association with vaccine uptake during the COVID-19 pandemic in Hungary using a contact diary approach in two cross-sectional surveys. The results indicate strong clustering among both vaccinated and unvaccinated groups. The most powerful predictor of vaccine uptake was the perceived vaccination rate within the egos' social contact network. Vaccination homophily and the role of the interpersonal contact network in vaccine uptake were particularly pronounced in the networks of close relationships, including family, kinship, and strong social ties of the ego. Our findings have important implications for understanding COVID-19 spread dynamics by showing that the strong clustering of unvaccinated individuals posed a great risk in preventing the spread of the disease.


Subject(s)
COVID-19 , Vaccination , Humans , COVID-19/prevention & control , COVID-19/epidemiology , COVID-19/psychology , Female , Male , Adult , Cross-Sectional Studies , Hungary/epidemiology , Middle Aged , Vaccination/psychology , COVID-19 Vaccines/administration & dosage , SARS-CoV-2/immunology , Social Networking , Vaccination Hesitancy/psychology , Vaccination Hesitancy/statistics & numerical data , Pandemics/prevention & control , Ego , Young Adult , Aged , Adolescent
7.
Eur J Med Res ; 29(1): 356, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38970130

ABSTRACT

BACKGROUND: To date, multiple cases of adverse reactions to COVID-19 vaccines have been reported worldwide. Alopecia areata (AA) is an uncommon type of adverse reaction reported in some articles and has a significant social and psychological impact on patients. Our study aimed to review the AA and COVID-19 vaccine literature. METHODS: This systematic review was conducted by searching for articles on AA following COVID-19 vaccines in international databases such as Embase, MEDLINE, PubMed, Web of Knowledge, and Ovid from December 2019 to December 30, 2023. We included studies that provided data for AA patients following COVID-19 vaccination with at least one dose. Data on sex, age, country/region of origin, vaccine type, days between vaccination and symptom presentation, manifestations of AA, trichoscopy and histopathological findings, treatment, and outcomes were included. RESULTS: In total, 579 explored studies were identified and assessed, and 25 articles with a total of 51 patients were included in the review. Twenty-seven (52.9%) patients developed new-onset AA following receiving the COVID-19 vaccine, and AA recurrence or exacerbation occurred after receiving the COVID-19 vaccine in 24 (47.1%) patients with preexisting disease. Five vaccines were reported to cause AA in all cases. The Pfizer vaccine (45.1%) was the most frequently reported, followed by the ChAdOx1 nCoV-19 vaccine (27.5%), Moderna mRNA-1273 (19.6%), Sinopharm (3.9%) and SinoVac (3.9%). AA occurred most frequently within one month after the 1st dose, and then, the incidence decreased gradually with time. Topical or systemic corticosteroids were used in 38 patients. Eleven patients were treated with a Janus Kinase inhibitor (jakinib) inhibitor, eight with tofacitinib, and three with an unspecified jakinib. However, 3 of the 11 patients experienced exacerbations after treatment. CONCLUSION: AA after COVID-19 vaccination is rare, and physicians should be aware of this phenomenon to improve early diagnosis and appropriate treatment.


Subject(s)
Alopecia Areata , COVID-19 Vaccines , COVID-19 , Humans , Alopecia Areata/chemically induced , COVID-19 Vaccines/adverse effects , COVID-19/prevention & control , COVID-19/complications , COVID-19/epidemiology , SARS-CoV-2/immunology , Male , Female
9.
Immun Inflamm Dis ; 12(7): e1270, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38967360

ABSTRACT

BACKGROUND: The emergence of new SARS-CoV-2 variants and the global COVID-19 pandemic spurred urgent vaccine development. While common vaccine side effects are well-documented, rare adverse events necessitate post-marketing surveillance. Recent research linked messenger RNA vaccines to thrombotic microangiopathy (TMA), a group of syndromes characterized by microvascular hemolytic anemia and thrombocytopenia. This report describes a new-onset atypical hemolytic-uremic syndrome (aHUS) occurring after COVID-19 vaccination and complements recent literature. CASE PRESENTATION: A previously healthy 25-year-old woman developed malaise, nausea, edema, and renal dysfunction 60 days postvaccination. Laboratory findings confirmed TMA diagnosis. Genetic testing for complement system mutations was negative. Kidney biopsy supported the diagnosis, and the patient required hemodialysis. CONCLUSION: This case illustrates the rare occurrence of aHUS following COVID-19 vaccination, with unique characteristics compared to previous reports. Despite the critical role of vaccination in pandemic control, emerging adverse events, such as vaccine-related TMA, must be recognized and investigated. Additional clinical trials are imperative to comprehend the clinical features and pathophysiological mechanisms underlying TMA associated with COVID-19 vaccination.


Subject(s)
Atypical Hemolytic Uremic Syndrome , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Humans , Female , Adult , Atypical Hemolytic Uremic Syndrome/diagnosis , COVID-19/prevention & control , COVID-19/immunology , SARS-CoV-2/immunology , COVID-19 Vaccines/adverse effects , Renal Dialysis , Vaccination/adverse effects
10.
PLoS One ; 19(7): e0302332, 2024.
Article in English | MEDLINE | ID: mdl-38968176

ABSTRACT

Vaccination against COVID-19 can prevent severe illness and reduce hospitalizations and deaths. Understanding and addressing determinants contributing to vaccine uptake among high-risk groups, such as Latinos, are pivotal in ensuring equitable vaccine distribution, promoting health equity, and fostering community engagement to bridge the gap in vaccine acceptance and ultimately enhance public health. This study aimed to examine factors influencing vaccine uptake among Latinos. We conducted a cross-sectional study using an online platform (n = 242). The survey was administered using a multimodal approach. Strategies for recruitment included community outreach, social media, and targeting community networks serving Latinos. Descriptive statistics, chi-square, and multivariable analysis were performed. Overall, 81.4% of respondents had received at least one dose of the COVID-19 vaccine, with 77.0% recommending it and 70.6% believing it to be safe, 66.7% believing in its efficacy, 62.3% able to find trustful information in Spanish or Portuguese, and almost 40% who relied on health organizations as their primary resource for COVID-19 vaccine information. Factors significantly associated with vaccine uptake included higher education level (p<0.001), English level (p = 0.023), living in an urban area (p = 0.048), having insurance (p<0.001), and having a healthcare provider (p = 0.007). Furthermore, belief in vaccine safety and efficacy, trust in public health authorities, concerns about COVID-19, the ability to determine true/false vaccine information during the pandemic, and the availability of trustworthy information in Spanish/Portuguese had statistically significant associations (p<0.05) with COVID-19 vaccine uptake. COVID-19 vaccine uptake differed based on sociodemographic and other modifiable factors. Our findings emphasize the importance of implementing targeted interventions and culturally sensitive communication strategies to improve vaccination uptake among the Latino community in the United States.


Subject(s)
COVID-19 Vaccines , COVID-19 , Hispanic or Latino , Humans , Hispanic or Latino/statistics & numerical data , COVID-19 Vaccines/administration & dosage , Male , Cross-Sectional Studies , Female , Adult , COVID-19/prevention & control , COVID-19/epidemiology , Middle Aged , Vaccination/statistics & numerical data , Young Adult , SARS-CoV-2/immunology , Surveys and Questionnaires , Adolescent , Patient Acceptance of Health Care , Health Knowledge, Attitudes, Practice , Aged , Vaccination Hesitancy/statistics & numerical data , Vaccination Hesitancy/psychology
11.
PLoS One ; 19(7): e0292143, 2024.
Article in English | MEDLINE | ID: mdl-38968187

ABSTRACT

OBJECTIVE: This study aimed to map the existing literature to identify predictors of COVID-19 vaccine acceptability among refugees, immigrants, and other migrant populations. METHODS: A systematic search of Medline, Embase, Scopus, APA PsycInfo and Cumulative Index of Nursing and Allied Health Literature (CINAHL) was conducted up to 31 January 2023 to identify the relevant English peer-reviewed observational studies. Two independent reviewers screened abstracts, selected studies, and extracted data. RESULTS: We identified 34 cross-sectional studies, primarily conducted in high income countries (76%). Lower vaccine acceptance was associated with mistrust in the host countries' government and healthcare system, concerns about the safety and effectiveness of COVID-19 vaccines, limited knowledge of COVID-19 infection and vaccines, lower COVID-19 risk perception, and lower integration level in the host country. Female gender, younger age, lower education level, and being single were associated with lower vaccine acceptance in most studies. Additionally, sources of information about COVID-19 and vaccines and previous history of COVID-19 infection, also influence vaccine acceptance. Vaccine acceptability towards COVID-19 booster doses and various vaccine brands were not adequately studied. CONCLUSIONS: Vaccine hesitancy and a lack of trust in COVID-19 vaccines have become significant public health concerns within migrant populations. These findings may help in providing information for current and future vaccine outreach strategies among migrant populations.


Subject(s)
COVID-19 Vaccines , COVID-19 , Refugees , Transients and Migrants , Vaccination Hesitancy , Humans , Refugees/psychology , COVID-19 Vaccines/administration & dosage , COVID-19/prevention & control , COVID-19/epidemiology , Transients and Migrants/psychology , Vaccination Hesitancy/psychology , Vaccination Hesitancy/statistics & numerical data , Patient Acceptance of Health Care , SARS-CoV-2/immunology , Female , Male , Vaccination/psychology , Vaccination/statistics & numerical data
12.
Medicine (Baltimore) ; 103(27): e38809, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38968522

ABSTRACT

In kidney transplant recipients (KTRs), viral infection can lead to antibody and/or T-cell mediated rejection, resulting in kidney transplant dysfunction. Therefore, it is critical to prevent infections. However, KTRs exhibit suboptimal responses to SARS-CoV-2 and/or influenza vaccines, partly due to immunosuppressant therapy. Inter- and intra-individual differences in the biological responses to vaccines may also affect patients' antibody production ability. This study included KTRs who received an messenger RNA SARS-CoV-2 vaccine (3 doses), and an inactivated quadrivalent influenza vaccine (1 or 2 doses). We measured the patients' total antibody titers against SARS-CoV-2 spike antigen, and hemagglutination inhibition (HI) titers against influenza A/H1N1, A/H3N2, B/Yamagata, and B/Victoria. Five patients were eligible for this study. Of these 5 KTRs, two produced anti-SARS-CoV-2 spike antibody titers to a seroprotective level, and also produced HI titers against A/H1N1 to a seroprotective level. Another 2 KTRs did not produce seroprotective anti-SARS-CoV-2 antibody titers, but produced seroprotective HI titers against A/H1N1. The remaining KTR produced a seroprotective anti-SARS-CoV-2 antibody titer, but did not produce a seroprotective HI titer against A/H1N1. The 2 KTRs who did not produce seroprotective anti-SARS-CoV-2 antibody titers following vaccination, later developed COVID-19, and this infection increased their titers over the seroprotective level. This study demonstrated that inter- and intra-individual differences in biological responses to vaccines should be considered in pediatric KTRs, in addition to immunosuppressant effects. Personalized regimens, such as augmented or booster doses of vaccines, could potentially improve the vaccination efficacy against SARS-CoV-2 and influenza.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Influenza Vaccines , Influenza, Human , Kidney Transplantation , SARS-CoV-2 , Humans , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Male , Female , COVID-19/prevention & control , COVID-19/immunology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/immunology , Influenza, Human/prevention & control , Influenza, Human/immunology , SARS-CoV-2/immunology , Antibodies, Viral/blood , Child , Adolescent , Transplant Recipients , Influenza A Virus, H1N1 Subtype/immunology , Vaccination/methods
13.
Front Immunol ; 15: 1424374, 2024.
Article in English | MEDLINE | ID: mdl-38966641

ABSTRACT

At the beginning of the COVID-19 pandemic those with underlying chronic lung conditions, including tuberculosis (TB), were hypothesized to be at higher risk of severe COVID-19 disease. However, there is inconclusive clinical and preclinical data to confirm the specific risk SARS-CoV-2 poses for the millions of individuals infected with Mycobacterium tuberculosis (M.tb). We and others have found that compared to singly infected mice, mice co-infected with M.tb and SARS-CoV-2 leads to reduced SARS-CoV-2 severity compared to mice infected with SARS-CoV-2 alone. Consequently, there is a large interest in identifying the molecular mechanisms responsible for the reduced SARS-CoV-2 infection severity observed in M.tb and SARS-CoV-2 co-infection. To address this, we conducted a comprehensive characterization of a co-infection model and performed mechanistic in vitro modeling to dynamically assess how the innate immune response induced by M.tb restricts viral replication. Our study has successfully identified several cytokines that induce the upregulation of anti-viral genes in lung epithelial cells, thereby providing protection prior to challenge with SARS-CoV-2. In conclusion, our study offers a comprehensive understanding of the key pathways induced by an existing bacterial infection that effectively restricts SARS-CoV-2 activity and identifies candidate therapeutic targets for SARS-CoV-2 infection.


Subject(s)
COVID-19 , Coinfection , Immunity, Innate , Mycobacterium tuberculosis , SARS-CoV-2 , COVID-19/immunology , Animals , Mycobacterium tuberculosis/immunology , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Mice , Coinfection/immunology , Humans , Tuberculosis/immunology , Tuberculosis/microbiology , Cytokines/metabolism , Cytokines/immunology , Disease Models, Animal , Severity of Illness Index , Lung/immunology , Lung/virology , Lung/microbiology , Lung/pathology , Virus Replication , Mice, Inbred C57BL , Female
14.
Commun Biol ; 7(1): 805, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38961245

ABSTRACT

Precise epitope determination of therapeutic antibodies is of great value as it allows for further comprehension of mechanism of action, therapeutic responsiveness prediction, avoidance of unwanted cross reactivity, and vaccine design. The golden standard for discontinuous epitope determination is the laborious X-ray crystallography method. Here, we present a combinatorial method for rapid mapping of discontinuous epitopes by mammalian antigen display, eliminating the need for protein expression and purification. The method is facilitated by automated workflows and tailored software for antigen analysis and oligonucleotide design. These oligos are used in automated mutagenesis to generate an antigen receptor library displayed on mammalian cells for direct binding analysis by flow cytometry. Through automated analysis of 33930 primers an optimized single condition cloning reaction was defined allowing for mutation of all surface-exposed residues of the receptor binding domain of SARS-CoV-2. All variants were functionally expressed, and two reference binders validated the method. Furthermore, epitopes of three novel therapeutic antibodies were successfully determined followed by evaluation of binding also towards SARS-CoV-2 Omicron BA.2. We find the method to be highly relevant for rapid construction of antigen libraries and determination of antibody epitopes, especially for the development of therapeutic interventions against novel pathogens.


Subject(s)
COVID-19 , Epitope Mapping , Epitopes , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Humans , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Epitope Mapping/methods , Epitopes/immunology , Epitopes/genetics , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/metabolism , COVID-19/immunology , COVID-19/virology , Peptide Library , Antibodies, Viral/immunology , Animals , HEK293 Cells , Cell Surface Display Techniques/methods , Gene Library
15.
J Exp Med ; 221(9)2024 Sep 02.
Article in English | MEDLINE | ID: mdl-38949638

ABSTRACT

Studies during the COVID-19 pandemic showed that children had heightened nasal innate immune responses compared with adults. To evaluate the role of nasal viruses and bacteria in driving these responses, we performed cytokine profiling and comprehensive, symptom-agnostic testing for respiratory viruses and bacterial pathobionts in nasopharyngeal samples from children tested for SARS-CoV-2 in 2021-22 (n = 467). Respiratory viruses and/or pathobionts were highly prevalent (82% of symptomatic and 30% asymptomatic children; 90 and 49% for children <5 years). Virus detection and load correlated with the nasal interferon response biomarker CXCL10, and the previously reported discrepancy between SARS-CoV-2 viral load and nasal interferon response was explained by viral coinfections. Bacterial pathobionts correlated with a distinct proinflammatory response with elevated IL-1ß and TNF but not CXCL10. Furthermore, paired samples from healthy 1-year-olds collected 1-2 wk apart revealed frequent respiratory virus acquisition or clearance, with mucosal immunophenotype changing in parallel. These findings reveal that frequent, dynamic host-pathogen interactions drive nasal innate immune activation in children.


Subject(s)
COVID-19 , Immunity, Innate , SARS-CoV-2 , Humans , Immunity, Innate/immunology , Child, Preschool , Infant , COVID-19/immunology , COVID-19/virology , Child , SARS-CoV-2/immunology , Female , Male , Nasopharynx/immunology , Nasopharynx/virology , Nasopharynx/microbiology , Viral Load , Nasal Mucosa/immunology , Nasal Mucosa/virology , Nasal Mucosa/microbiology , Cytokines/metabolism , Cytokines/immunology , Host-Pathogen Interactions/immunology , Adolescent , Nose/immunology , Nose/virology , Nose/microbiology , Coinfection/immunology , Coinfection/virology
17.
JAMA Netw Open ; 7(7): e2419258, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38949812

ABSTRACT

Importance: In the context of emerging SARS-CoV-2 variants or lineages and new vaccines, it is key to accurately monitor COVID-19 vaccine effectiveness (CVE) to inform vaccination campaigns. Objective: To estimate the effectiveness of COVID-19 vaccines administered in autumn and winter 2022 to 2023 against symptomatic SARS-CoV-2 infection (with all circulating viruses and XBB lineage in particular) among people aged 60 years or older in Europe, and to compare different CVE approaches across the exposed and reference groups used. Design, Setting, and Participants: This case-control study obtained data from VEBIS (Vaccine Effectiveness, Burden and Impact Studies), a multicenter study that collects COVID-19 and influenza data from 11 European sites: Croatia; France; Germany; Hungary; Ireland; Portugal; the Netherlands; Romania; Spain, national; Spain, Navarre region; and Sweden. Participants were primary care patients aged 60 years or older with acute respiratory infection symptoms who were recruited at the 11 sites after the start of the COVID-19 vaccination campaign from September 2022 to August 2023. Cases and controls were defined as patients with positive and negative, respectively, reverse transcription-polymerase chain reaction (RT-PCR) test results. Exposures: The exposure was COVID-19 vaccination. The exposure group consisted of patients who received a COVID-19 vaccine during the autumn and winter 2022 to 2023 vaccination campaign and 14 days or more before symptom onset. Reference group included patients who were not vaccinated during or in the 6 months before the 2022 to 2023 campaign (seasonal CVE), those who were never vaccinated (absolute CVE), and those who were vaccinated with at least the primary series 6 months or more before the campaign (relative CVE). For relative CVE of second boosters, patients receiving their second booster during the campaign were compared with those receiving 1 booster 6 months or more before the campaign. Main Outcomes and Measures: The outcome was RT-PCR-confirmed, medically attended, symptomatic SARS-CoV-2 infection. Four CVE estimates were generated: seasonal, absolute, relative, and relative of second boosters. CVE was estimated using logistic regression, adjusting for study site, symptom onset date, age, chronic condition, and sex. Results: A total of 9308 primary care patients were included, with 1687 cases (1035 females; median [IQR] age, 71 [65-79] years) and 7621 controls (4619 females [61%]; median [IQR] age, 71 [65-78] years). Within 14 to 89 days after vaccination, seasonal CVE was 29% (95% CI, 14%-42%), absolute CVE was 39% (95% CI, 6%-60%), relative CVE was 31% (95% CI, 15% to 44%), and relative CVE of second boosters was 34% (95% CI, 18%-47%) against all SARS-CoV-2 variants. In the same interval, seasonal CVE was 44% (95% CI, -10% to 75%), absolute CVE was 52% (95% CI, -23% to 82%), relative CVE was 47% (95% CI, -8% to 77%), and relative CVE of second boosters was 46% (95% CI, -13% to 77%) during a period of high XBB circulation. Estimates decreased with time since vaccination, with no protection from 180 days after vaccination. Conclusions and Relevance: In this case-control study among older Europeans, all CVE approaches suggested that COVID-19 vaccines administered in autumn and winter 2022 to 2023 offered at least 3 months of protection against symptomatic, medically attended, laboratory-confirmed SARS-CoV-2 infection. The effectiveness of new COVID-19 vaccines against emerging SARS-CoV-2 variants should be continually monitored using CVE seasonal approaches.


Subject(s)
COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Seasons , Vaccine Efficacy , Humans , Aged , COVID-19/prevention & control , COVID-19/epidemiology , COVID-19 Vaccines/administration & dosage , COVID-19 Vaccines/therapeutic use , Female , Europe/epidemiology , Male , SARS-CoV-2/immunology , Middle Aged , Case-Control Studies , Aged, 80 and over , Vaccination/statistics & numerical data , European People
18.
BMC Infect Dis ; 24(1): 654, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951848

ABSTRACT

Vaccination against COVID-19 was integral to controlling the pandemic that persisted with the continuous emergence of SARS-CoV-2 variants. Using a mathematical model describing SARS-CoV-2 within-host infection dynamics, we estimate differences in virus and immunity due to factors of infecting variant, age, and vaccination history (vaccination brand, number of doses and time since vaccination). We fit our model in a Bayesian framework to upper respiratory tract viral load measurements obtained from cases of Delta and Omicron infections in Singapore, of whom the majority only had one nasopharyngeal swab measurement. With this dataset, we are able to recreate similar trends in URT virus dynamics observed in past within-host modelling studies fitted to longitudinal patient data.We found that Omicron had higher R0,within values than Delta, indicating greater initial cell-to-cell spread of infection within the host. Moreover, heterogeneities in infection dynamics across patient subgroups could be recreated by fitting immunity-related parameters as vaccination history-specific, with or without age modification. Our model results are consistent with the notion of immunosenescence in SARS-CoV-2 infection in elderly individuals, and the issue of waning immunity with increased time since last vaccination. Lastly, vaccination was not found to subdue virus dynamics in Omicron infections as well as it had for Delta infections.This study provides insight into the influence of vaccine-elicited immunity on SARS-CoV-2 within-host dynamics, and the interplay between age and vaccination history. Furthermore, it demonstrates the need to disentangle host factors and changes in pathogen to discern factors influencing virus dynamics. Finally, this work demonstrates a way forward in the study of within-host virus dynamics, by use of viral load datasets including a large number of patients without repeated measurements.


Subject(s)
COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Vaccination , Humans , COVID-19/immunology , COVID-19/prevention & control , COVID-19/virology , COVID-19/epidemiology , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Middle Aged , Aged , Adult , Singapore/epidemiology , Age Factors , Viral Load , Young Adult , Bayes Theorem , Models, Theoretical , Male , Aged, 80 and over , Female , Adolescent
19.
Front Immunol ; 15: 1379570, 2024.
Article in English | MEDLINE | ID: mdl-38957465

ABSTRACT

There is a reciprocal relationship between extracellular matrix (ECM) remodelling and inflammation that could be operating in the progression of severe COVID-19. To explore the immune-driven ECM remodelling in COVID-19, we in this explorative study analysed these interactions in hospitalised COVID-19 patients. RNA sequencing and flow analysis were performed on peripheral blood mononuclear cells. Inflammatory mediators in plasma were measured by ELISA and MSD, and clinical information from hospitalised COVID-19 patients (N=15) at admission was included in the analysis. Further, we reanalysed two publicly available datasets: (1) lung tissue RNA-sequencing dataset (N=5) and (2) proteomics dataset from PBCM. ECM remodelling pathways were enriched in PBMC from COVID-19 patients compared to healthy controls. Patients treated at the intensive care unit (ICU) expressed distinct ECM remodelling gene profiles compared to patients in the hospital ward. Several markers were strongly correlated to immune cell subsets, and the dysregulation in the ICU patients was positively associated with plasma levels of inflammatory cytokines and negatively associated with B-cell activating factors. Finally, our analysis of publicly accessible datasets revealed (i) an augmented ECM remodelling signature in inflamed lung tissue compared to non-inflamed tissue and (ii) proteomics analysis of PBMC from severe COVID-19 patients demonstrated an up-regulation in an ECM remodelling pathway. Our results may suggest the presence of an interaction between ECM remodelling, inflammation, and immune cells, potentially initiating or perpetuating pulmonary pathology in severe COVID-19.


Subject(s)
COVID-19 , Extracellular Matrix , Leukocytes, Mononuclear , SARS-CoV-2 , Humans , COVID-19/immunology , COVID-19/blood , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Extracellular Matrix/metabolism , Male , Female , Middle Aged , SARS-CoV-2/physiology , SARS-CoV-2/immunology , Aged , Cytokines/blood , Proteomics/methods , Lung/immunology , Lung/pathology , Adult
20.
FASEB J ; 38(13): e23814, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38959046

ABSTRACT

As we enter a new era of mRNA-based therapeutics, evidence on genetic or environmental factors that might predispose to unknown off-target side effects, gains in importance. Among these factors, exercise appears likely to have influenced otherwise cryptic cases of early-onset postvaccination myocarditis. And the existence of a distinct late-onset myocarditis is now being recognized. Here, three case-history reports suggest crypticity (the author's own case), unless provoked by a preexisting cardiac morbidity (one case), or by immune checkpoint blockade to enhance anticancer autoimmunity (several cases). These reports are supported by noninvasive fluorodeoxyglucose-based cardiac scan comparisons of multiple vaccinated and unvaccinated subjects. In pre-pandemic decades, applications for funds by the leading innovator in mRNA-based therapeutics seldom gained peer-review approval. Thus, at the start of the pandemic, the meager data on such side effects could justify only emergency approval. We must do better.


Subject(s)
COVID-19 , Myocarditis , Vaccination , Myocarditis/etiology , Humans , Male , COVID-19/prevention & control , COVID-19/immunology , Vaccination/adverse effects , Female , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/immunology , Middle Aged , SARS-CoV-2/immunology , Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...