Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 668
Filter
1.
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
2.
Front Immunol ; 15: 1398028, 2024.
Article in English | MEDLINE | ID: mdl-39148725

ABSTRACT

Introduction: A potential overlap in symptoms between post-acute COVID-19 syndrome and post-COVID-19 vaccination syndrome has been noted. We report a paired description of patients presenting with similar manifestations involving the central (CNS) or peripheral nervous system (PNS) following SARS-CoV-2 infection or vaccination, suggesting that both may have triggered similar immune-mediated neurological disorders in the presence of anti-idiotype antibodies directed against the ACE2 protein. Patients and methods: Four patients exhibited overlapping neurological manifestations following SARS-CoV-2 infection or vaccination: radiculitis, Guillain-Barré syndrome, and MRI-negative myelitis, respectively, sharing positivity for anti-ACE2 antibodies. Autoantibodies against AQP-4, MOG, GlyR, GAD, and amphiphysin, onconeural antibodies for CNS syndromes, and anti-ganglioside antibodies for PNS syndromes tested negative in all patients. Discussion: Anti-idiotype antibodies against ACE2 have been detected in patients who recovered from COVID-19 infection, and it has been hypothesized that such antibodies may mediate adverse events following SARS-CoV-2 infection or vaccination, resulting in the activation of the immune system against cells expressing ACE2, such as neurons. Our data reveal clinically overlapping syndromes triggered by SARS-CoV-2 infection or vaccination, sharing positivity for anti-ACE2 antibodies. Their presence, in the absence of other classic autoimmune markers of CNS or PNS involvement, suggests that they might play an active role in the context of an aberrant immune response. Conclusion: Anti-idiotype antibodies directed against ACE2 may be triggered by both SARS-CoV-2 infection and vaccination, possibly contributing to neurological autoimmune manifestations. Their pathogenic role, however, remains to be demonstrated in large-scale, more structured studies.


Subject(s)
Angiotensin-Converting Enzyme 2 , Autoantibodies , COVID-19 , SARS-CoV-2 , Humans , COVID-19/immunology , Male , SARS-CoV-2/immunology , Middle Aged , Autoantibodies/immunology , Autoantibodies/blood , Female , Angiotensin-Converting Enzyme 2/immunology , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/immunology , Aged , Antibodies, Anti-Idiotypic/immunology , Vaccination/adverse effects , Adult , Post-Acute COVID-19 Syndrome , Nervous System Diseases/immunology , Nervous System Diseases/etiology , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/etiology
3.
Front Immunol ; 15: 1404800, 2024.
Article in English | MEDLINE | ID: mdl-39156891

ABSTRACT

Background: Patients suffering from neurological symptoms after COVID-19 vaccination (post-COVID-19 vaccination syndrome (PCVS)) have imposed an increasing challenge on medical practice, as diagnostic precision and therapeutic options are lacking. Underlying autoimmune dysfunctions, including autoantibodies, have been discussed in neurological disorders after SARS-CoV-2 infection and vaccination. Here, we describe the frequency and targets of autoantibodies against peripheral nervous system tissues in PCVS. Methods: Sera from 50 PCVS patients with peripheral neurological symptoms after COVID-19 vaccination and 35 vaccinated healthy controls were used in this study. IgG autoreactivity was measured via indirect immunofluorescence assays on mouse sciatic nerve teased fibers. The frequencies of autoantibodies were compared between groups using Fisher's exact test. Serum anti-ganglioside antibodies were measured in ganglioside blots. Autoantibody target identification was performed using immunoprecipitation coupled to mass spectrometry. Subsequent target confirmation was conducted via cell-based assays and ELISA. Results: Compared with controls, PCVS patients had a significantly greater frequency of autoantibodies against peripheral nervous system structures (9/50(18%) vs 1/35(3%); p=0.04). Autoantibodies bound to paranodes (n=5), axons (n=4), Schmidt-Lanterman incisures (n=2) and Schwann cell nuclei (n=1). Conversely, antibodies against gangliosides were absent in PCVS patients. Target identification and subsequent confirmation revealed various subunits of neurofilaments as well as DFS-70 as autoantibody epitopes. Conclusion: Our data suggest that autoantibodies against nervous system tissue could be relevant in PCVS patients. Autoantibodies against neurofilaments and cell nuclei with so far non-established links to this disease spectrum should be further elucidated to determine their biomarker potential.


Subject(s)
Autoantibodies , COVID-19 Vaccines , COVID-19 , Immunoglobulin G , SARS-CoV-2 , Humans , Autoantibodies/immunology , Autoantibodies/blood , Male , Female , Immunoglobulin G/immunology , Immunoglobulin G/blood , Middle Aged , COVID-19/immunology , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Aged , Mice , Animals , Adult , Vaccination , Gangliosides/immunology , Peripheral Nerves/immunology
4.
BMC Infect Dis ; 24(1): 841, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164637

ABSTRACT

BACKGROUND: According to Norwegian registries, 91% of individuals ≥ 16 years had received ≥ 1 dose of COVID-19 vaccine by mid-July 2022, whereas less than 2% of children < 12 years were vaccinated. Confirmed COVID-19 was reported for 27% of the population, but relaxation of testing lead to substantial underreporting. We have characterized the humoral immunity to SARS-CoV-2 in Norway in the late summer of 2022 by estimating the seroprevalence and identifying antibody profiles based on reactivity to Wuhan or Omicron-like viruses in a nationwide cross-sectional collection of residual sera, and validated our findings using cohort sera. METHODS: 1,914 anonymized convenience sera and 243 NorFlu-cohort sera previously collected from the Oslo-area with reported infection and vaccination status were analyzed for antibodies against spike, the receptor-binding domain (RBD) of the ancestral Wuhan strain and Omicron BA.2 RBD, and nucleocapsid (N). Samples were also tested for antibodies inhibiting RBD-ACE2 interaction. Neutralization assays were performed on subsets of residual sera against B.1, BA.2, XBB.1.5 and BQ.1.1. RESULTS: The national seroprevalence estimate from vaccination and/or infection was 99.1% (95% CrI 97.0-100.0%) based on Wuhan (spike_W and RBD_W) and RBD_BA2 antibodies. Sera from children < 12 years had 2.2 times higher levels of antibodies against RBD_BA2 than RBD_W and their seroprevalence estimate showed a 14.4 percentage points increase when also including anti-RBD_BA2 antibodies compared to Wuhan-antibodies alone. 50.3% (95% CI 45.0-55.5%) of residual sera from children and 38.1% (95% CI 36.0-40.4%) of all residual sera were positive for anti-N-antibodies. By combining measurements of binding- and ACE2-RBD-interaction-inhibiting antibodies, reactivity profiles indicative of infection and vaccination history were identified and validated using cohort sera. Residual sera with a profile indicative of hybrid immunity were able to neutralize newer Omicron variants XBB.1.5 and BQ.1.1. CONCLUSIONS: By late summer of 2022, most of the Norwegian population had antibodies to SARS-CoV-2, and almost all children had been infected. Antibody profiles indicated that children mostly had experienced a primary Omicron infection, while hybrid immunity was common among adults. The finding that sera displaying hybrid immunity could neutralize newer Omicron variants indicates that Wuhan-like priming of the immune response did not have a harmful imprinting effect and that infections induce cross-reacting antibodies against future variants.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Humans , COVID-19/epidemiology , COVID-19/immunology , Antibodies, Viral/blood , SARS-CoV-2/immunology , Norway/epidemiology , Seroepidemiologic Studies , Child , Adult , Adolescent , Middle Aged , Male , Child, Preschool , Female , Young Adult , COVID-19 Vaccines/immunology , Aged , Infant , Cross-Sectional Studies , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Spike Glycoprotein, Coronavirus/immunology
5.
BMC Neurol ; 24(1): 291, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164636

ABSTRACT

BACKGROUND: To investigate the safety (adverse events [AEs] and post-vaccination multiple sclerosis [MS] activity within 6 weeks), clinical efficacy (protection against coronavirus disease 2019 [COVID-19]), and vaccine-induced humoral immunogenicity (SARS-CoV-2 neutralizing antibody, anti-nucleocapsid IgG, and anti-spike IgG) of the Sinopharm (BBIBP-CorV) vaccine among people with MS (PwMS) receiving different disease-modifying therapies (DMTs). METHODS: This prospective cohort study was conducted between November 2021 and May 2022. PwMS were followed for six months after the 2nd dose of vaccination. Antibody responses were measured 2-16 weeks after the 2nd dose injection. Multivariate logistic regression was employed to assess the impact of each DMT on dichotomous antibody responses, adjusting for age, sex, MS phenotype, expanded disability status scale, disease duration, and vaccination-antibody titration interval. RESULTS: Among the 261 screened PwMS, 209 (aged 38.23 ± 9.73 years, female: 70.8%; relapsing-remitting MS: 80.4%) were included. The frequencies of experiencing non-serious AEs and post-vaccination MS activity were 66.0% and 4.8%, respectively. Breakthrough COVID-19 infection was observed in 14.8% of the PwMS. A subcohort of 125 PwMS was assessed for antibody responses. Positive neutralizing antibodies, anti-nucleocapsid IgG, and anti-spike IgG were detected in 36.8%, 35.2%, and 52.0% of the PwMS, respectively. Multivariate regression indicated a 96% (OR: 0.04 [95% CI: 0.00, 0.51], P = 0.013), 93% (OR: 0.07 [0.01, 0.64], P = 0.019), and 89% (OR: 0.11 [0.01, 0.96], P = 0.045) reduced odds of positive neutralizing antibody, anti-nucleocapsid IgG, and anti-spike IgG, respectively, among fingolimod-receivers. Additionally, anti-CD20s-receivers had 88% (OR: 0.12 [0.02, 0.85], P = 0.034) lower odds of being positive for anti-nucleocapsid IgG. CONCLUSIONS: BBIBP-CorV appeared to be well tolerated in PwMS, with promising clinical efficacy. However, a suboptimal humoral response was observed in PwMS receiving fingolimod and anti-CD20s. Future research should investigate the relationship between humoral responses and the frequency and severity of COVID-19 infection across various DMTs.


Subject(s)
Antibodies, Neutralizing , COVID-19 Vaccines , COVID-19 , Multiple Sclerosis , Humans , Female , Male , COVID-19/prevention & control , COVID-19/immunology , COVID-19/epidemiology , Adult , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Prospective Studies , Middle Aged , Multiple Sclerosis/immunology , Multiple Sclerosis/drug therapy , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , SARS-CoV-2/immunology , Immunogenicity, Vaccine/immunology , Cohort Studies , Treatment Outcome , Immunoglobulin G/blood , Immunoglobulin G/immunology , Vaccines, Inactivated
6.
Article in English | MEDLINE | ID: mdl-39146978

ABSTRACT

Vaccines targeting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) have been pivotal in curtailing the spread of infection. Health care workers, as frontline responders, were among the first to receive vaccination to mitigate coronavirus disease in 2019 (COVID-19) transmission. This study aimed to assess the humoral response elicited by mRNA vaccines, specifically measuring antibodies against the spike S1 protein, a marker of immune response. A cohort of 649 health care workers received three doses of mRNA vaccine, with antibody levels evaluated before and after each dose within a 2- to 3-week interval. Participants were stratified into groups based on prior exposure to the virus: those without prior contact (440 individuals) and those with a history of infection (209 individuals). Among the latter, cases of SARS-CoV-2 infection ranged from asymptomatic (92 individuals) to mild symptomatic (117 individuals). Participants with a history of infection exhibited elevated levels of IgG antibodies against the S1 protein prior to vaccination. Notably, both immunoglobulin IgA class (IgA) and immunoglobulin IgG class (IgG) antibody responses increased significantly post-vaccination, peaking after the second dose for IgG and after the third dose for IgA. Interestingly, the immune response to the vaccine did not vary significantly based on the symptomatic or asymptomatic nature of prior infection. Furthermore, the study findings indicate that completion of the vaccination regimen led to sustained antibody production lasting between 6 months and 9 months. This study underscores the robust and enduring humoral response elicited by mRNA vaccines, particularly among health care workers, irrespective of prior SARS-CoV-2 exposure.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Health Personnel , Immunity, Humoral , Immunoglobulin G , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Vaccination , Humans , COVID-19/immunology , COVID-19/prevention & control , Antibodies, Viral/blood , Antibodies, Viral/immunology , SARS-CoV-2/immunology , Male , Spike Glycoprotein, Coronavirus/immunology , Female , Middle Aged , Adult , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Immunoglobulin G/blood , Immunoglobulin G/immunology , mRNA Vaccines , Immunoglobulin A/blood , Immunoglobulin A/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/administration & dosage , Antibody Formation/immunology
7.
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
8.
Virol J ; 21(1): 192, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39160532

ABSTRACT

BACKGROUND: Natural infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or vaccination triggers antibody production against key viral antigens. However, there is limited evidence on the levels of antibodies produced in naturally infected individuals compared to those vaccinated in Ethiopia. Therefore, we aimed to detect and compare SARS-CoV-2 antibodies produced by naturally infected and vaccinated individuals. MATERIALS AND METHODS: We conducted a multicenter cross-sectional study among a total of 355 naturally infected and 355 vaccinated individuals from November 2022 to April 2023 at 10 selected health facilities in Addis Ababa, Ethiopia. We enrolled the participants consecutively upon their arrival at health facilities until the required sample size was achieved. We used a structured questionnaire to collect data on the demographic and clinical characteristics of the participants. We also collected 3-5 ml of blood samples from all participants and tested for anti-Spike (anti-S) and anti-nucleocapsid (anti-N) antibodies using Cobas 6000. We utilized frequency, mean, or median to describe the data, the Mann-Whitney U test to compare groups, and a generalized linear regression model to assess factors associated with anti-S antibody concentration. We analyzed the data with SPSS version 26, and the level of significance was set at P-value < 0.05. RESULTS: Of the naturally infected participants, 352 (99.5%) had anti-S antibodies and all (100%) had anti-N antibodies, whereas among vaccinated participants, all (100%) had anti-S antibodies, while 323 (91.6%) had anti-N antibodies. Anti-S antibodies produced by vaccinated individuals were significantly (P < 0.001) higher than those produced as a result of natural infection. Being young (P = 0.004), having hypertension (P < 0.001), and having diabetes (P < 0.001) were significantly associated with lower anti-S antibody levels, while being recently vaccinated and having a higher number of vaccine doses were significantly associated with higher anti-S antibody concentrations in vaccinated participants. Having diabetes (P < 0.001) were significantly associated with lower anti-S concentrations in participants who were naturally infected. CONCLUSION: There is a high seropositivity rate in both naturally infected and vaccinated individuals. However, vaccinated individuals had higher levels of SARS-CoV-2 antibodies than those who were naturally infected, which highlights the significant contribution of vaccination in increasing the protection of COVID-19 in Ethiopia.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Humans , Ethiopia/epidemiology , Cross-Sectional Studies , COVID-19/immunology , Antibodies, Viral/blood , Male , Female , Adult , SARS-CoV-2/immunology , Middle Aged , Young Adult , COVID-19 Vaccines/immunology , Adolescent , Vaccination , Spike Glycoprotein, Coronavirus/immunology , Aged , Child
9.
Diagn Microbiol Infect Dis ; 110(2): 116425, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39098282

ABSTRACT

The study aimed to evaluate the effect of booster dose COVID-19 vaccines on prevention and humoral immune response in individuals with different vaccination schemes during the period BA.4 and BA.5 omicron sub-variants were globally dominant. The study included 146 individuals who preferred different vaccination schemes for booster doses. Anti-spike/RBD-IgG and neutralizing antibody levels were measured 28 days after the booster dose vaccination upon their consent. There is no significant difference between median antibody titers detected according to different vaccination schemes. SARS-CoV-2 neutralizing antibody inhibition percentages were detected significantly higher in serum samples before and after the last booster dose in 2 BNT162b2+1 BNT162b2(99.42 %), 2 BNT162b2 + 2 BNT162b2(99.42 %), and 2 BNT162b2 + 3 BNT162b2(99.42 %) vaccination schemes (p = 0.004, p = 0.044, p = 0.002,respectively). The study indicated that a booster vaccination dose provides a high level of protection against severe COVID-19 and death. We think that the variant-specific pancoronavirus vaccines will be necessary to protect against breakthrough infections.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization, Secondary , SARS-CoV-2 , Humans , COVID-19/prevention & control , COVID-19/immunology , Antibodies, Viral/blood , Male , Female , SARS-CoV-2/immunology , Prospective Studies , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Middle Aged , Antibodies, Neutralizing/blood , Adult , BNT162 Vaccine/immunology , BNT162 Vaccine/administration & dosage , Aged , Immunoglobulin G/blood , Vaccination , Spike Glycoprotein, Coronavirus/immunology , Antibody Formation/immunology
11.
Curr Microbiol ; 81(10): 317, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-39164527

ABSTRACT

Throughout history, infectious diseases have plagued humanity, with outbreaks occurring regularly worldwide. Not every outbreak affects people globally; however, in the case of Coronavirus Disease 2019 (COVID-19), caused by a novel coronavirus (SARS-CoV-2), it reached a pandemic level within a remarkably short period. Fortunately, advancements in medicine and biotechnology have facilitated swift responses to the disease, resulting in the development of therapeutics and vaccines. Nevertheless, the persistent spread of the virus and the emergence of new variants underscore the necessity for protective interventions, leading researchers to seek more effective vaccines. Despite the presence of various types of vaccines, including mRNA and inactivated vaccines against SARS-CoV-2, new platforms have been investigated since the pandemic, and research on bacterial membrane vesicles (BMVs) has demonstrated their potential as a novel COVID-19 vaccine platform. Researchers have explored different strategies for BMV-based COVID-19 vaccines, such as mixing the vesicles with antigenic components of the virus due to their adjuvant capacity or decorating the vesicles with the viral antigens to create adjuvanted delivery systems. These approaches have presented promising results in inducing robust immune responses, but obstacles such as reproducibility in obtaining and homogeneous characterization of BMVs remain in developing vesicle-based vaccines. Overall, the development of BMV-based vaccines represents a novel and promising strategy in the fight against COVID-19. Additional research and clinical trials are needed to further evaluate the potential of these vaccines to offer long-lasting protection against SARS-CoV-2 and its evolving variants.


Subject(s)
COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , SARS-CoV-2/immunology , SARS-CoV-2/genetics , COVID-19/prevention & control , COVID-19/immunology , Humans , Animals
12.
Article in English | MEDLINE | ID: mdl-39165016

ABSTRACT

Objectives: To compare serological evidence of prior severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection with linked coronavirus disease 2019 (COVID-19) case notification data in Victoria, Australia, and to determine in vitro SARS-CoV-2 neutralisation activity based on prior infection and vaccination history. Design, setting, participants: Four cross-sectional serological surveys were conducted between 30 June and 31 October 2022 (a period of Omicron BA.4/BA.5 dominance) using 1,974 residual serum samples obtained from the Victorian Infectious Diseases Reference Laboratory. Serological results were linked to COVID-19 case notification and vaccination data. Surrogate virus neutralisation testing was performed to obtain in vitro inhibition estimates by anti-nucleocapsid serostatus and COVID-19 vaccination history. Main outcome measures: Adjusted anti-SARS-CoV-2 spike and nucleocapsid seropositivity by sex, age and region of residence; adjusted proportion of cases notified by anti-nucleocapsid serostatus, age and number of COVID-19 vaccination doses received; adjusted percentage in vitro inhibition against wildtype and Omicron BA.4/BA.5 SARS-CoV-2 variants by anti-nucleocapsid serostatus and COVID-19 vaccination history. Results: The prevalence of anti-SARS-CoV-2 nucleocapsid antibodies was inversely proportional to age. In October 2022, prevalence was 84% (95% confidence interval [95% CI]: 75-93%) among 18-29-year-olds, compared to 39% (95% CI: 27-52%) among ≥ 80-year-olds. In most age groups, approximately 40% of COVID-19 cases appear to have been notified via existing surveillance mechanisms. Case notification was highest among individuals older than 80 years and people who had received COVID-19 vaccine booster doses. In vitro neutralisation of Omicron BA.4/BA.5 sub-variants was highest for individuals with evidence of both prior infection and booster vaccination. Conclusions: Under-notification of SARS-CoV-2 infections in the Victorian population is not uniform across age and vaccination strata. Seroprevalence data that give insights into case notification behaviour provide additional context for the interpretation of existing COVID-19 surveillance information.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Vaccination , Humans , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19/immunology , Male , Female , Victoria/epidemiology , Middle Aged , Adult , SARS-CoV-2/immunology , Aged , Adolescent , Young Adult , Antibodies, Viral/blood , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Cross-Sectional Studies , Seroepidemiologic Studies , Aged, 80 and over , Child , Child, Preschool , Age Factors , Infant , Antibodies, Neutralizing/blood
13.
Bull Exp Biol Med ; 177(2): 221-224, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39093475

ABSTRACT

Squalene-based adjuvant compositions that can provide effective induction of specific humoral immune response have been developed. Recombinant receptor-binding domain (RBD) of surface S-protein of SARS-CoV-2 was used to evaluate the properties of the composition. Immunization of mice with the developed squalene-based compositions in combination with RBD allows obtaining high titers of specific antibodies: from 105 to 2×106. The blood sera from immunized mice exhibit neutralizing activity against SARS-CoV-2 Delta variant (B.1.617.2) with a titer up to 1:2000.


Subject(s)
Adjuvants, Immunologic , Antibodies, Neutralizing , Antibodies, Viral , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Squalene , Squalene/immunology , Animals , Antibodies, Neutralizing/immunology , Adjuvants, Immunologic/pharmacology , SARS-CoV-2/immunology , Mice , Antibodies, Viral/immunology , Antibodies, Viral/blood , Spike Glycoprotein, Coronavirus/immunology , COVID-19/immunology , COVID-19/virology , COVID-19 Vaccines/immunology , Female , Humans , Mice, Inbred BALB C , Immunity, Humoral/drug effects
14.
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
15.
Nat Commun ; 15(1): 6811, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39122676

ABSTRACT

Resting memory B cells can be divided into classical or atypical groups, but the heterogenous marker expression on activated memory B cells makes similar classification difficult. Here, by longitudinal analysis of mass cytometry and CITE-seq data from cohorts with COVID-19, bacterial sepsis, or BNT162b2 mRNA vaccine, we observe that resting B cell memory consist of classical CD45RB+ memory and CD45RBlo memory, of which the latter contains of two distinct groups of CD11c+ atypical and CD23+ non-classical memory cells. CD45RB levels remain stable in these cells after activation, thereby enabling the tracking of activated B cells and plasmablasts derived from either CD45RB+ or CD45RBlo memory B cells. Moreover, in both COVID-19 patients and mRNA vaccination, CD45RBlo B cells formed the majority of SARS-CoV2 specific memory B cells and correlated with serum antibodies, while CD45RB+ memory are activated by bacterial sepsis. Our results thus identify that stably expressed CD45RB levels can be exploited to trace resting memory B cells and their activated progeny, and suggest that atypical and non-classical CD45RBlo memory B cells contribute to SARS-CoV-2 infection and vaccination.


Subject(s)
BNT162 Vaccine , COVID-19 , Leukocyte Common Antigens , Memory B Cells , SARS-CoV-2 , Humans , COVID-19/immunology , Leukocyte Common Antigens/metabolism , SARS-CoV-2/immunology , Memory B Cells/immunology , BNT162 Vaccine/immunology , Male , Antibodies, Viral/immunology , Antibodies, Viral/blood , Middle Aged , Female , COVID-19 Vaccines/immunology , Vaccination , Adult , Immunologic Memory/immunology , mRNA Vaccines/immunology , B-Lymphocytes/immunology , Aged
17.
PLoS One ; 19(8): e0302338, 2024.
Article in English | MEDLINE | ID: mdl-39102410

ABSTRACT

BACKGROUND: SARS-CoV-2 vaccines are safe and effective against infection and severe COVID-19 disease worldwide. Certain co-morbid conditions cause immune dysfunction and may reduce immune response to vaccination. In contrast, those with co-morbidities may practice infection prevention strategies. Thus, the real-world clinical impact of co-morbidities on SARS-CoV-2 infection in the recent post-vaccination period is not well established. This study was performed to understand the epidemiology of Omicron breakthrough infection and evaluate associations with number of comorbidities in a vaccinated and boosted population. METHODS AND FINDINGS: A retrospective clinical cohort study was performed utilizing the Northwestern Medicine Enterprise Data Warehouse. Our study population was identified as fully vaccinated adults with at least one booster. The primary risk factor of interest was the number of co-morbidities. The primary outcome was the incidence and time to the first positive SARS-CoV-2 molecular test in the Omicron predominant era. Multivariable Cox modeling analyses to determine the hazard of SARS-CoV-2 infection were stratified by calendar time (Period 1: January 1 -June 30, 2022; Period 2: July 1 -December 31, 2022) due to violations in the proportional hazards assumption. In total, 133,191 patients were analyzed. During Period 1, having 3+ comorbidities was associated with increased hazard for breakthrough (HR = 1.16 CI 1.08-1.26). During Period 2 of the study, having 2 comorbidities (HR = 1.45 95% CI 1.26-1.67) and having 3+ comorbidities (HR 1.73, 95% CI 1.51-1.97) were associated with increased hazard for Omicron breakthrough. Older age was associated with decreased hazard in Period 1 of follow-up. Interaction terms for calendar time indicated significant changes in hazard for many factors between the first and second halves of the follow-up period. CONCLUSIONS: Omicron breakthrough is common with significantly higher risk for our most vulnerable patients with multiple co-morbidities. Age plays an important role in breakthrough infection with the highest incidence among young adults, which may be due to age-related behavioral factors. These findings reflect real-world differences in immunity and exposure risk behaviors for populations vulnerable to COVID-19.


Subject(s)
COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Humans , COVID-19/epidemiology , COVID-19/prevention & control , COVID-19/virology , Male , Female , Middle Aged , SARS-CoV-2/immunology , Incidence , Risk Factors , Adult , Retrospective Studies , COVID-19 Vaccines/immunology , Aged , Chicago/epidemiology , Comorbidity , Vaccination , Immunization, Secondary , Young Adult , Breakthrough Infections
18.
Sci Adv ; 10(32): eadn7187, 2024 Aug 09.
Article in English | MEDLINE | ID: mdl-39110802

ABSTRACT

Over the past few decades, the development of potent and safe immune-activating adjuvant technologies has become the heart of intensive research in the constant fight against highly mutative and immune evasive viruses such as influenza, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and human immunodeficiency virus (HIV). Herein, we developed a highly modular saponin-based nanoparticle platform incorporating Toll-like receptor agonists (TLRas) including TLR1/2a, TLR4a, and TLR7/8a adjuvants and their mixtures. These various TLRa-saponin nanoparticle adjuvant constructs induce unique acute cytokine and immune-signaling profiles, leading to specific T helper responses that could be of interest depending on the target disease for prevention. In a murine vaccine study, the adjuvants greatly improved the potency, durability, breadth, and neutralization of both COVID-19 and HIV vaccine candidates, suggesting the potential broad application of these adjuvant constructs to a range of different antigens. Overall, this work demonstrates a modular TLRa-SNP adjuvant platform that could improve the design of vaccines and affect modern vaccine development.


Subject(s)
Adjuvants, Immunologic , COVID-19 Vaccines , Nanoparticles , SARS-CoV-2 , Saponins , Toll-Like Receptor Agonists , Animals , Humans , Mice , Adjuvants, Immunologic/pharmacology , Adjuvants, Vaccine/chemistry , AIDS Vaccines/immunology , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , COVID-19 Vaccines/immunology , Cytokines/metabolism , Nanoparticles/chemistry , Saponins/pharmacology , Saponins/chemistry , Saponins/immunology , SARS-CoV-2/immunology
19.
Front Immunol ; 15: 1383086, 2024.
Article in English | MEDLINE | ID: mdl-39119342

ABSTRACT

Current vaccines against COVID-19 elicit immune responses that are overall strong but wane rapidly. As a consequence, the necessary booster shots have contributed to vaccine fatigue. Hence, vaccines that would provide lasting protection against COVID-19 are needed, but are still unavailable. Cytomegaloviruses (CMVs) elicit lasting and uniquely strong immune responses. Used as vaccine vectors, they may be attractive tools that obviate the need for boosters. Therefore, we tested the murine CMV (MCMV) as a vaccine vector against COVID-19 in relevant preclinical models of immunization and challenge. We have previously developed a recombinant MCMV vaccine vector expressing the spike protein of the ancestral SARS-CoV-2 (MCMVS). In this study, we show that the MCMVS elicits a robust and lasting protection in young and aged mice. Notably, spike-specific humoral and cellular immunity was not only maintained but also even increased over a period of at least 6 months. During that time, antibody avidity continuously increased and expanded in breadth, resulting in neutralization of genetically distant variants, like Omicron BA.1. A single dose of MCMVS conferred rapid virus clearance upon challenge. Moreover, MCMVS vaccination controlled two variants of concern (VOCs), the Beta (B.1.135) and the Omicron (BA.1) variants. Thus, CMV vectors provide unique advantages over other vaccine technologies, eliciting broadly reactive and long-lasting immune responses against COVID-19.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , SARS-CoV-2/immunology , SARS-CoV-2/genetics , Mice , COVID-19 Vaccines/immunology , COVID-19/prevention & control , COVID-19/immunology , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Antibodies, Viral/immunology , Antibodies, Viral/blood , Muromegalovirus/immunology , Muromegalovirus/genetics , Female , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Mice, Inbred BALB C , Humans , Genetic Vectors , Immunity, Cellular , Immunity, Humoral , Disease Models, Animal
20.
Front Immunol ; 15: 1427501, 2024.
Article in English | MEDLINE | ID: mdl-39131157

ABSTRACT

Objective: to evaluate the immune response to the SARS-CoV-2 vaccines in adults with immune-mediated rheumatic diseases (IMRDs) in comparison to healthy individuals, observed 1-20 weeks following the fourth vaccine dose. Additionally, to evaluate the impact of immunosuppressive therapies, vaccination schedules, the time interval between vaccination and sample collection on the vaccine's immune response. Methods: We designed a longitudinal observational study conducted at the rheumatology department of Hospital de Copiapó. Neutralizing antibodies (Nabs) titers against the Wuhan and Omicron variant were analyzed between 1-20 weeks after administration of the fourth dose of the SARS-CoV-2 vaccine to 341 participants (218 IMRD patients and 123 healthy controls). 218 IMRD patients with rheumatoid arthritis (RA), psoriatic arthritis (PsA), ankylosing spondylitis (AS), systemic lupus erythematosus (SLE), systemic vasculitis (VS) and systemic scleroderma (SS) were analyzed. Results: Performing a comparison between the variants, Wuhan vs Omicron, we noticed that there were significant differences (p<0.05) in the level of the ID50, both for healthy controls and for patients with IMRDs. The humoral response of patients with IMRDs is significantly lower compared to healthy controls for the Omicron variant of SARS-CoV-2 (p = 0.0015). The humoral response of patients with IMRDs decreases significantly when the time interval between vaccination and sample collection is greater than 35 days. This difference was observed in the response, both for the Wuhan variant and for the Omicron variant. Conclusion: The IMRDs patients, the humoral response variation in the SARS-CoV-2 vaccine depends on doses and type of vaccine administered, the humoral response times and the treatment that these patients are receiving.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization, Secondary , Rheumatic Diseases , SARS-CoV-2 , Humans , Male , Middle Aged , Female , COVID-19/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Rheumatic Diseases/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , Adult , Aged , Longitudinal Studies , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL