Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Clin Infect Dis ; 2022 Nov 04.
Article in English | MEDLINE | ID: covidwho-2281396

ABSTRACT

BACKGROUND: Understanding vaccine-dependent effects on protective and sustained humoral immune response are crucial to plan further vaccination strategies against COVID-19. Population-based data comparing different vaccination strategies are lacking. METHODS: This multicenter, population-based cohort study included 4,601 individuals ≥18 years of age after primary vaccination against COVID-19 at least four months ago (full immunization). We compared factors associated with residual antibody levels against SARS-CoV-2 receptor binding domain (RBD) across different vaccination strategies (BNT162b2, mRNA-1273, or ChAdOx1 nCoV-19 [ChAdOx1]). RESULTS: Our main model including 3,787 individuals (2xBNT162b2 n = 2,271, 2xmRNA-1273 n = 251, 2xChAdOx1 n = 1,265) predicted significantly lower levels of anti-RBD-antibodies after 6 months in individuals vaccinated with ChAdOx1 (392.7 BAU/ml) compared to those vaccinated with BNT162b2 (1179.5 BAU/ml) or mRNA-1273 (2098.2 BAU/ml). Vaccine-dependent association of antibody levels was found for age with a significant predicted difference in BAU/ml per year for BNT162b2 (-21.5 [95%CI -24.7 to -18.3]) and no significant association for mRNA-1273 (-4.0 [95%CI -20.0 to 12.1]) or ChAdOx1 (1.7 [95%CI 0.2 to 3.1]). The predicted decrease over time since full immunization was highest in mRNA-1273 (-23.4 [95%CI -31.4 to -15.4]) compared to BNT162b2 -5.9 [95%CI -7 to -4.8]). Higher antibody levels were observed for individuals with systemic adverse events upon vaccination and current smoking (BNT162b2), for days between first and second vaccination (BNT162b2 and ChAdOx1) and for absence of comorbidities (all). CONCLUSION: Our study revealed population-based evidence of vaccine-dependent effects of age and time since full immunization on humoral immune response. Findings underline the importance of an individualized vaccine selection, especially in elderly individuals.

2.
J Biol Rhythms ; : 7487304221132355, 2022 Nov 11.
Article in English | MEDLINE | ID: covidwho-2229775

ABSTRACT

Data from human and animal studies are highly suggestive of an influence of time of day of vaccine administration on host immune responses. In this population-based study, we aimed to investigate the effect of time of day of administration of a COVID-19 vector vaccine, ChAdOx1 nCoV-19 (AstraZeneca), on SARS-CoV-2 anti-spike S1 immunoglobulin (IgG) levels. Participants were 803 university employees who received their first vaccine dose in March 2021, had serology data at baseline and at 3 weeks, and were seronegative at baseline. Antibody levels were determined in binding antibody units (BAU/mL) using enzyme-linked immunosorbent assay (ELISA). Generalized additive models (GAM) and linear regression were used to evaluate the association of time of day of vaccination continuously and in hourly bins with antibody levels at 3 weeks. Participants had a mean age of 42 years (SD: 12; range: 21-74) and 60% were female. Time of day of vaccination was associated non-linearly ("reverse J-shape") with antibody levels. Morning vaccination was associated with the highest (9:00-10:00 h: mean 292.1 BAU/mL; SD: 262.1), early afternoon vaccination with the lowest (12:00-13:00 h: mean 217.3 BAU/mL; SD: 153.6), and late afternoon vaccination with intermediate (14:00-15:00 h: mean 280.7 BAU/mL; SD: 262.4) antibody levels. Antibody levels induced by 12:00-13:00 h vaccination (but not other time intervals) were significantly lower compared to 9:00-10:00 h vaccination after adjusting for potential confounders (beta coefficient = -75.8, 95% confidence interval [CI] = -131.3, -20.4). Our findings show that time of day of vaccination against SARS-CoV-2 has an impact on the magnitude of IgG antibody levels at 3 weeks. Whether this difference persists after booster vaccine doses and whether it influences the level of protection against COVID-19 needs further evaluation.

3.
Pharmaceutics ; 15(1)2022 Dec 29.
Article in English | MEDLINE | ID: covidwho-2216706

ABSTRACT

Nature offers a wide range of evolutionary optimized materials that combine unique properties with intrinsic biocompatibility and that can be exploited as biomimetic materials. The R5 and RRIL peptides employed here are derived from silaffin proteins that play a crucial role in the biomineralization of marine diatom silica shells and are also able to form silica materials in vitro. Here, we demonstrate the application of biomimetic silica particles as a vaccine delivery and adjuvant platform by linking the precipitating peptides R5 and the RRIL motif to a variety of peptide antigens. The resulting antigen-loaded silica particles combine the advantages of biomaterial-based vaccines with the proven intracellular uptake of silica particles. These particles induce NETosis in human neutrophils as well as IL-6 and TNF-α secretion in murine bone marrow-derived dendritic cells.

4.
Immunol Lett ; 250: 1-6, 2022 10.
Article in English | MEDLINE | ID: covidwho-2028118

ABSTRACT

Antibody testing after COVID-19 vaccination is generally not recommended. Here, we present the results of a retrospective study, in which we analyzed antibody levels before and after the first dose of the ChAdOx1 vector vaccine. We identified 5% non-responders (43.6 ± 10.6 years; females: 41%) and 3.4% low-responders (44.2 ± 10.1 years; females: 64%) after the first dose. Of these, 61 individuals received a timely second dose either with a homologous (ChAdOx1/ChAdOx1) or heterologous (ChAdOx1/mRNA-1273) schedule. All vaccinees achieved positive S1-specific IgG titers to the ancestral SARS-CoV-2 strain after the second dose, but antibody levels as well as neutralization titers against the ancestral SARS-CoV-2 strain were higher after the heterologous schedule. However, Omicron-specific neutralizing antibodies were not detectable after two doses in either group, indicating that a third vaccine dose is needed to enhance cross-reactive antibodies against currently circulating and emerging variants of concern.


Subject(s)
COVID-19 Vaccines , COVID-19 , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , Female , Humans , Immunoglobulin G , Retrospective Studies , SARS-CoV-2 , Seroconversion , Vaccination
5.
Nat Commun ; 13(1): 5362, 2022 09 12.
Article in English | MEDLINE | ID: covidwho-2016704

ABSTRACT

Impaired response to COVID-19 vaccination is of particular concern in immunosuppressed patients. To determine the best vaccination strategy for this vulnerable group we performed a single center, 1:1 randomized blinded clinical trial. Patients who failed to seroconvert upon two mRNA vaccinations (BNT162b2 or mRNA-1273) are randomized to receive either a third dose of the same mRNA or the vector vaccine ChAdOx1 nCoV-19. Primary endpoint is the difference in SARS-CoV-2 spike antibody seroconversion rate between vector and mRNA vaccinated patients four weeks after the third dose. Secondary outcomes include cellular immune responses. Seroconversion rates at week four are significantly higher in the mRNA (homologous vaccination, 15/24, 63%) as compared to the vector vaccine group (heterologous vaccination, 4/22, 18%). SARS-CoV-2-specific T-cell responses are reduced but could be increased after a third dose of either vector or mRNA vaccine. In a multivariable logistic regression analysis, patient age and vaccine type are associated with seroconversion. No serious adverse event is attributed to COVID-19 booster vaccination. Efficacy and safety data underline the importance of a booster vaccination and support the use of a homologous mRNA booster vaccination in immunosuppressed patients.Trial registration: EudraCT No.: 2021-002693-10.


Subject(s)
BNT162 Vaccine , COVID-19 Vaccines , COVID-19 , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines/adverse effects , ChAdOx1 nCoV-19 , Humans , Immunization, Secondary , RNA, Messenger , SARS-CoV-2/genetics , Vaccination , Vaccines, Synthetic , mRNA Vaccines
6.
Allergy ; 77(8): 2431-2445, 2022 08.
Article in English | MEDLINE | ID: covidwho-1985600

ABSTRACT

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is responsible for the ongoing global COVID-19 pandemic. One possibility to control the pandemic is to induce sterilizing immunity through the induction and maintenance of neutralizing antibodies preventing SARS-CoV-2 from entering human cells to replicate in. METHODS: We report the construction and in vitro and in vivo characterization of a SARS-CoV-2 subunit vaccine (PreS-RBD) based on a structurally folded recombinant fusion protein consisting of two SARS-CoV-2 Spike protein receptor-binding domains (RBD) fused to the N- and C-terminus of hepatitis B virus (HBV) surface antigen PreS to enable the two unrelated proteins serving as immunologic carriers for each other. RESULTS: PreS-RBD, but not RBD alone, induced a robust and uniform RBD-specific IgG response in rabbits. Currently available genetic SARS-CoV-2 vaccines induce mainly transient IgG1 responses in vaccinated subjects whereas the PreS-RBD vaccine induced RBD-specific IgG antibodies consisting of an early IgG1 and sustained IgG4 antibody response in a SARS-CoV-2 naive subject. PreS-RBD-specific IgG antibodies were detected in serum and mucosal secretions, reacted with SARS-CoV-2 variants, including the omicron variant of concern and the HBV receptor-binding sites on PreS of currently known HBV genotypes. PreS-RBD-specific antibodies of the immunized subject more potently inhibited the interaction of RBD with its human receptor ACE2 and their virus-neutralizing titers (VNTs) were higher than median VNTs in a random sample of healthy subjects fully immunized with registered SARS-CoV-2 vaccines or in COVID-19 convalescent subjects. CONCLUSION: The PreS-RBD vaccine has the potential to serve as a combination vaccine for inducing sterilizing immunity against SARS-CoV-2 and HBV by stopping viral replication through the inhibition of cellular virus entry.


Subject(s)
COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , Antibodies, Neutralizing , Antibodies, Viral , COVID-19/prevention & control , COVID-19 Vaccines/immunology , Humans , Immunoglobulin G , Pandemics/prevention & control , Rabbits , Spike Glycoprotein, Coronavirus/immunology
7.
Front Immunol ; 13: 889138, 2022.
Article in English | MEDLINE | ID: covidwho-1875415

ABSTRACT

Background: Individuals with secondary immunodeficiencies belong to the most vulnerable groups to succumb to COVID-19 and thus are prioritized for SARS-CoV-2 vaccination. However, knowledge about the persistence and anamnestic responses following SARS-CoV-2-mRNA vaccinations is limited in these patients. Methods: In a prospective, open-label, phase four trial we analyzed S1-specific IgG, neutralizing antibodies and cytokine responses in previously non-infected patients with cancer or autoimmune disease during primary mRNA vaccination and up to one month after booster. Results: 263 patients with solid tumors (SOT, n=63), multiple myeloma (MM, n=70), inflammatory bowel diseases (IBD, n=130) and 66 controls were analyzed. One month after the two-dose primary vaccination the highest non-responder rate was associated with lower CD19+ B-cell counts and was found in MM patients (17%). S1-specific IgG levels correlated with IL-2 and IFN-γ responses in controls and IBD patients, but not in cancer patients. Six months after the second dose, 18% of patients with MM, 10% with SOT and 4% with IBD became seronegative; no one from the control group became negative. However, in IBD patients treated with TNF-α inhibitors, antibody levels declined more rapidly than in controls. Overall, vaccination with mRNA-1273 led to higher antibody levels than with BNT162b2. Importantly, booster vaccination increased antibody levels >8-fold in seroresponders and induced anamnestic responses even in those with undetectable pre-booster antibody levels. Nevertheless, in IBD patients with TNF-α inhibitors even after booster vaccination, antibody levels were lower than in untreated IBD patients and controls. Conclusion: Immunomonitoring of vaccine-specific antibody and cellular responses seems advisable to identify vaccination failures and consequently establishing personalized vaccination schedules, including shorter booster intervals, and helps to improve vaccine effectiveness in all patients with secondary immunodeficiencies. Trial registration: EudraCT Number: 2021-000291-11.


Subject(s)
COVID-19 , Inflammatory Bowel Diseases , Multiple Myeloma , BNT162 Vaccine , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Immunization, Secondary , Immunocompromised Host , Immunoglobulin G , Immunologic Memory , Multiple Myeloma/therapy , Prospective Studies , RNA, Messenger , SARS-CoV-2 , Tumor Necrosis Factor-alpha , Vaccination
8.
Vaccines (Basel) ; 10(5)2022 May 11.
Article in English | MEDLINE | ID: covidwho-1869846

ABSTRACT

BACKGROUND: Two years into the pandemic, vaccination remains the most effective option to prevent coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Preliminary studies suggest vaccination efficacy in patients with inflammatory bowel diseases (IBD), but little is known about its impact on chronic intestinal inflammation. Here we assessed the mucosal inflammatory activity in patients with IBD before and after immunization with the mRNA-1273 (Moderna) vaccine by measurement of fecal calprotectin (fCP). METHODS: In 42 patients with IBD, the baseline fCP levels obtained prior to the first vaccine were compared with the highest levels measured during and after two doses of vaccination. Patients' sera were collected after the second dose to evaluate anti-SARS-CoV-2 antibodies' titers. RESULTS: We observed a significant fCP elevation in 31% of patients after any dose. Vedolizumab was identified as the only agent associated with an fCP increase (OR 12.4, 95% CI [1.6; 120.2], p = 0.0171). Gastrointestinal adverse events were reported in 9.5% of all subjects and in 75% of cases accompanied by an fCP increase. Anti-SARS-CoV-2 antibodies associated only weakly with the fCP increase after the first dose (p = 0.04). CONCLUSIONS: Our findings support possible collinearity in pathways of SARS-CoV-2 antigen expression and the pathogenesis of IBD.

9.
Commun Med (Lond) ; 1: 13, 2021.
Article in English | MEDLINE | ID: covidwho-1860406

ABSTRACT

Background: In spring 2020, at the beginning of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic in Europe, we set up an assay system for large-scale testing of virus-specific and neutralising antibodies including their longevity. Methods: We analysed the sera of 1655 adult employees for SARS-CoV-2-specific antibodies using the S1 subunit of the spike protein of SARS-CoV-2. Sera containing S1-reactive antibodies were further evaluated for receptor-binding domain (RBD)- and nucleocapsid protein (NCP)-specific antibodies in relation to the neutralisation test (NT) results at three time points over six months. Results: We detect immunoglobulin G (IgG) and/or IgA antibodies reactive to the S1 protein in 10.15% (n = 168) of the participants. In total, 0.97% (n = 16) are positive for S1-IgG, 0.91% (n = 15) were S1-IgG- borderline and 8.28% (n = 137) exhibit only S1-IgA antibodies. Of the 168 S1-reactive sera, 8.33% (n = 14) have detectable RBD-specific antibodies and 6.55% (n = 11) NCP-specific antibodies. The latter correlates with NTs (kappa coefficient = 0.8660) but start to decline after 3 months. RBD-specific antibodies correlate most closely with the NT (kappa = 0.9448) and only these antibodies are stable for up to six months. All participants with virus-neutralising antibodies report symptoms, of which anosmia and/or dysgeusia correlate most closely with the detection of virus-neutralising antibodies. Conclusions: RBD-specific antibodies are most reliably detected post-infection, independent of the number/severity of symptoms, and correlate with neutralising antibodies at least for six months. They thus qualify best for large-scale seroepidemiological evaluation of both antibody reactivity and virus neutralisation.

10.
Front Med (Lausanne) ; 9: 822316, 2022.
Article in English | MEDLINE | ID: covidwho-1731799

ABSTRACT

In a SARS-CoV-2 seroprevalence study conducted with 1,655 working adults in spring of 2020, 12 of the subjects presented with positive neutralization test (NT) titers (>1:10). They were here followed up for 1 year to assess their Ab persistence. We report that 7/12 individuals (58%) had NT_50 titers ≥1:50 and S1-specific IgG ≥50 BAU/ml 1 year after mild COVID-19 infection. S1-specific IgG were retained until a year when these levels were at least >60 BAU/ml at 3 months post-infection. For both the initial fast and subsequent slow decline phase of Abs, we observed a significant correlation between NT_50 titers and S1-specific IgG and thus propose S1-IgG of 60 BAU/ml 3 months post-infection as a potential threshold to predict neutralizing Ab persistence for 1 year. NT_50 titers and S1-specific IgG also correlated with circulating S1-specific memory B-cells. SARS-CoV-2-specific Ab levels after primary mRNA vaccination in healthy controls were higher (Geometric Mean Concentration [GMC] 3158 BAU/ml [CI 2592 to 3848]) than after mild COVID-19 infection (GMC 82 BAU/ml [CI 48 to 139]), but showed a stronger fold-decline within 5-6 months (0.20-fold, to GMC 619 BAU/ml [CI 479 to 801] vs. 0.56-fold, to GMC 46 BAU/ml [CI 26 to 82]). Of particular interest, the decline of both infection- and vaccine-induced Abs correlated with body mass index. Our data contribute to describe decline and persistence of SARS-CoV-2-specific Abs after infection and vaccination, yet the relevance of the maintained Ab levels for protection against infection and/or disease depends on the so far undefined correlate of protection.

12.
Vaccine ; 39(51): 7375-7378, 2021 12 17.
Article in English | MEDLINE | ID: covidwho-1488002

ABSTRACT

Patients undergoing immunosuppressive treatments have a higher need for protection against coronavirus disease (COVID19) that follows infection with the SARS-CoV-2 virus but their ability to respond sufficiently to COVID vaccines is uncertain. We retrospectively evaluated SARS-CoV-2 spike subunit 1 (S1)-specific antibody levels after two mRNA doses in 242 patients with underlying chronic inflammatory, hematooncological or metabolic diseases and in solid organ transplant recipients. S1-specific antibodies were measured 30 days after the second dose. In 15.9% of these patients, no S1-specific antibodies were detectable. Non-responsiveness was linked to administration of B-cell depleting therapies as well as to ongoing therapies that block lymphocyte trafficking (Fingolimod) or inhibit T cell proliferation (Tacrolimus). Thus, it is important to inform immunosuppressed patients about the risk of vaccine non-responsiveness and the necessity to maintain non-pharmaceutical protection measures. In these risk patients antibody testing and cellular analysis are helpful to estimate the benefit/responsiveness to further booster vaccinations.


Subject(s)
COVID-19 Vaccines , COVID-19 , Antibodies, Viral , Antibody Formation , Humans , RNA, Messenger , Retrospective Studies , SARS-CoV-2 , Vaccination
13.
Allergo J Int ; 30(5): 169-175, 2021.
Article in English | MEDLINE | ID: covidwho-1442203

ABSTRACT

BACKGROUND: Along with the newly approved vaccines against coronavirus disease 2019 (COVID-19), first reports of allergic or intolerance reactions were published. Subsequently, questions arose whether these vaccines pose an increased risk for intolerance reactions and whether allergic patients may be at higher risk for this. RESULTS: Allergic reactions following COVID-19 vaccinations have been reported, but mostly of mild severity and at normal (Moderna®) or only slightly increased frequency (BioNTech/Pfizer®) compared to established conventional vaccines. The risk of allergic reaction to the newly licensed vector vaccines (AstraZeneca®, Johnson&Johnson®) cannot be conclusively assessed yet, but also appears to be low. There is currently no evidence that patients with allergic diseases (atopic patients) react more frequently or more severely to these vaccines. It is currently assumed that intolerance reactions of the immediate-type are either type I allergic (IgE-mediated) reactions or occur via complement activation (CARPA, "complement activation-related pseudoallergy"). Polyethylene glycol (PEG) or polysorbate, which are present as stabilizers in the vaccines, are suspected as triggers for this. CONCLUSION: The data available so far do not show a significantly increased risk of immediate-type allergic reactions in atopic persons. In almost all cases, atopic patients can be vaccinated without problems. Standardized follow-up tests after suspected allergic reactions or CARPA-mediated reactions are currently limited.

15.
Allergo J Int ; 30(5): 155-168, 2021.
Article in English | MEDLINE | ID: covidwho-1283828

ABSTRACT

BACKGROUND: The vaccines against the coronavirus disease 2019 (COVID-19) approved in the European Union represent a decisive step in the fight against the pandemic. The application of these available vaccines to patients with pre-existing immunological conditions leads to a multitude of questions regarding efficacy, side effects and the necessary patient information. RESULTS: This review article provides insight into mechanisms of action of the currently available severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines and summarises the current state of science as well as expert recommendations regarding tolerability of the vaccines. In addition, the potential to develop protective immune responses is determined. A special focus is given on patients under immunosuppression or in treatment with immunomodulatory drugs. Special groups of the population such as children, pregnant women and the elderly are also considered. CONCLUSION: Despite the need for a patient-specific risk-benefit assessment, the consensus among experts is that patients with immunological diseases in particular benefit from the induced immune protection after COVID-19 vaccination and do not have an increased risk of side effects.

SELECTION OF CITATIONS
SEARCH DETAIL