Advances in COVID-19 Vaccines and Neutralizing Antibody

A special issue of Vaccines (ISSN 2076-393X). This special issue belongs to the section "COVID-19 Vaccines and Vaccination".

Deadline for manuscript submissions: closed (10 February 2024) | Viewed by 30633

Special Issue Editors


E-Mail Website
Guest Editor
Department of Cancer Biology, Cardinal Bernardin Cancer Center, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153, USA
Interests: COVID-19; molecular virology; neutralizing antibodies; aging; telomere biology; cancer; genomic instability; cancer immunotherapy; vaccine

E-Mail Website
Guest Editor
Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
Interests: COVID-19; neutralizing antibodies; cancer immunotherapy; mucosal immunology

E-Mail Website
Guest Editor
Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
Interests: immunology; cancer immunotherapy; infectious disease; innate immunity; mucosal immunology; T cell biology

E-Mail Website
Guest Editor
Department of Pediatrics, Division of Infectious Disease, School of Medicine, Emory University, Atlanta, GA, USA
Interests: COVID-19; neutralizing antibodies; COVID vaccine; MISC; COVID infection and vaccination in children

Special Issue Information

Dear Colleagues,

The rapid and unprecedented spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the resulting coronavirus disease 2019 (COVID-19) has caused millions of deaths worldwide. It has also put pressure on the healthcare industry everywhere. In order to control the spread of coronavirus disease, as many as 31 COVID-19 vaccines have been approved for emergency use to date, and over 10 billion doses have been administered globally. However, many new variants of SARS-CoV-2 have emerged since the vaccine development, with high transmissibility and an ability to escape the immune responses. 

Antibody immune response is important for the clearance of the virus and critical for the generation of memory response to prevent reinfection. SARS-CoV-2 generates a virus-specific IgM, and IgG and neutralizing IgG response in the days following infection. However, the IgG response wanes over time and in some cases leads to only partial protection. There is a need for further research in this field to understand the measure/degree of long-term protection conferred by the vaccine, vaccine-driven pathology, and immune differences among patients. This necessitates the critical evaluation of vaccine efficiency to expedite vaccine research.  

In this Special Issue, we invite articles investigating antibody response to coronavirus, including but not limited to the role of neutralizing antibodies in SARS-CoV-2 infection, mechanisms that lead to viral escape, immune parameters correlated with cytokine release syndrome, and T cell and B cell dynamics in coronavirus infection. Research in this field will aid in further understanding the pathogenesis of the disease and in the development of improved vaccine strategies. 

We look forward to receiving your contributions.

Dr. Rishi Jaiswal
Dr. Srijani Basu
Dr. Suman Gupta
Dr. Sneh Lata Gupta
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Vaccines is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • COVID-19
  • neutralizing antibodies
  • B cells
  • T cells
  • IgG

Published Papers (13 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Editorial

Jump to: Research, Review, Other

3 pages, 177 KiB  
Editorial
An Assessment of the Bivalent Vaccine as a Second Booster for COVID-19
by Sneh Lata Gupta and Rishi K. Jaiswal
Vaccines 2023, 11(1), 79; https://doi.org/10.3390/vaccines11010079 - 29 Dec 2022
Cited by 5 | Viewed by 1938
Abstract
In the USA, two monovalent COVID-19 mRNA vaccines are primarily used for vaccination [...] Full article
(This article belongs to the Special Issue Advances in COVID-19 Vaccines and Neutralizing Antibody)

Research

Jump to: Editorial, Review, Other

10 pages, 1412 KiB  
Article
The Equal Neutralizing Effectiveness of BNT162b2, ChAdOx1 nCoV-19, and Sputnik V Vaccines in the Palestinian Population
by Alexia Damour, Muriel Faure, Nicolas Landrein, Jessica Ragues, Narda Ardah, Haneen Dhaidel, Marie-Edith Lafon, Harald Wodrich and Walid Basha
Vaccines 2024, 12(5), 493; https://doi.org/10.3390/vaccines12050493 - 3 May 2024
Viewed by 512
Abstract
Since the beginning of the COVID-19 pandemic, different viral vector-based and mRNA vaccines directed against the SARS-CoV-2 “S” spike glycoprotein have been developed and have shown a good profile in terms of safety and efficacy. Nevertheless, an unbiased comparison of vaccination efficiency, including [...] Read more.
Since the beginning of the COVID-19 pandemic, different viral vector-based and mRNA vaccines directed against the SARS-CoV-2 “S” spike glycoprotein have been developed and have shown a good profile in terms of safety and efficacy. Nevertheless, an unbiased comparison of vaccination efficiency, including post-vaccination neutralizing activity, between the different vaccines remains largely unavailable. This study aimed to compare the efficacy of one mRNA (BNT162b2) and two non-replicating adenoviral vector vaccines (ChAdOx1 nCoV-19 and Sputnik V) in a cohort of 1120 vaccinated Palestinian individuals who received vaccines on an availability basis and which displayed a unique diversity of genetic characteristics. We assessed the level of anti-S antibodies and further determined the antibody neutralizing activity in 261 of those individuals vaccinated with BNT162b2a (121), ChAdOx1 (72) or Sputnik V (68). Our results showed no significant difference in the distribution of serum-neutralizing activity or S-antibody serum levels for the three groups of vaccines, proving equivalence in efficacy for the three vaccines under real-life conditions. In addition, none of the eight demographic parameters tested had an influence on vaccination efficacy. Regardless of the vaccine type, the vaccination campaign ultimately played a pivotal role in significantly reducing the morbidity and mortality associated with COVID-19 in Palestine. Full article
(This article belongs to the Special Issue Advances in COVID-19 Vaccines and Neutralizing Antibody)
Show Figures

Figure 1

12 pages, 1197 KiB  
Article
Analysis of the Presence and Levels of IgG Antibodies Directed against the S1 Protein Receptor Binding Domain and the N Protein of SARS-CoV-2 in Patients with Multiple Sclerosis Treated with Immunomodulatory Therapies
by Joanna Kulikowska, Katarzyna Kapica-Topczewska, Monika Gudowska-Sawczuk, Agnieszka Kulczyńska-Przybik, Marcin Bazylewicz, Anna Mirończuk, Agata Czarnowska, Waldemar Brola, Barbara Mroczko, Jan Kochanowicz and Alina Kułakowska
Vaccines 2024, 12(3), 255; https://doi.org/10.3390/vaccines12030255 - 29 Feb 2024
Viewed by 821
Abstract
The coronavirus 2019 disease (COVID-19) course and serological statuses of patients with relapsing–remitting multiple sclerosis (RRMS), treated with disease-modifying therapies (DMTs) are generally parallel that of the general population. Over the pandemic’s course, however, a notable increase in the number of RRMS patients [...] Read more.
The coronavirus 2019 disease (COVID-19) course and serological statuses of patients with relapsing–remitting multiple sclerosis (RRMS), treated with disease-modifying therapies (DMTs) are generally parallel that of the general population. Over the pandemic’s course, however, a notable increase in the number of RRMS patients who received vaccination against severe acute respiratory coronavirus 2 (SARS-CoV-2) and those who had COVID-19 (symptomatic and asymptomatic) was reported. This virus and/or vaccination likely influenced DMT-treated RRMS patients’ serological statuses regarding the presence of SARS-CoV-2 antibodies and their quantitative expression. This investigation assesses the presence and levels of the antibody directed against the S1 protein receptor binding domain (SRBD) and against the N protein of SARS-CoV-2 in 38 DMT-treated RRMS patients. The findings indicate that people vaccinated against SARS-CoV-2 exhibited significantly higher levels of IgG antibodies against S1-RBD at both assessment points. Patients with a prior history of COVID-19 demonstrated statistically significant increases in anti-N antibodies at visit 1, whereas such statistical significance was not observed at visit 2. DMT-treated RRMS patients generated neutralizing antibodies following vaccination and/or COVID-19 infection. Nevertheless, it is noteworthy that antibody levels more accurately reflect the serological status and exhibit a stronger correlation with vaccination than just the presence of antibodies. Full article
(This article belongs to the Special Issue Advances in COVID-19 Vaccines and Neutralizing Antibody)
Show Figures

Figure 1

13 pages, 1690 KiB  
Article
Long-Term Immunogenicity and Safety of a Homologous Third Dose Booster Vaccination with TURKOVAC: Phase 2 Clinical Study Findings with 32-Week Post-Booster Follow-Up
by Zafer Sezer, Shaikh Terkis Islam Pavel, Ahmet Inal, Hazel Yetiskin, Busra Kaplan, Muhammet Ali Uygut, Ahmet Furkan Aslan, Adnan Bayram, Mumtaz Mazicioglu, Gamze Kalin Unuvar, Zeynep Ture Yuce, Gunsu Aydin, Refika Kamuran Kaya, Ihsan Ates, Ates Kara and Aykut Ozdarendeli
Vaccines 2024, 12(2), 140; https://doi.org/10.3390/vaccines12020140 - 29 Jan 2024
Viewed by 1460
Abstract
Vaccine-induced immunity wanes over time and warrants booster doses. We investigated the long-term (32 weeks) immunogenicity and safety of a third, homologous, open-label booster dose of TURKOVAC, administered 12 weeks after completion of the primary series in a randomized, controlled, double-blind, phase 2 [...] Read more.
Vaccine-induced immunity wanes over time and warrants booster doses. We investigated the long-term (32 weeks) immunogenicity and safety of a third, homologous, open-label booster dose of TURKOVAC, administered 12 weeks after completion of the primary series in a randomized, controlled, double-blind, phase 2 study. Forty-two participants included in the analysis were evaluated for neutralizing antibodies (NAbs) (with microneutralization (MNT50) and focus reduction (FRNT50) tests), SARS-CoV-2 S1 RBD (Spike S1 Receptor Binding Domain), and whole SARS-CoV-2 (with ELISA) IgGs on the day of booster injection and at weeks 1, 2, 4, 8, 16, 24, and 32 thereafter. Antibody titers increased significantly from week 1 and remained higher than the pre-booster titers until at least week 4 (week 8 for whole SARS-CoV-2) (p < 0.05 for all). Seroconversion (titers ≥ 4-fold compared with pre-immune status) persisted 16 weeks (MNT50: 6-fold; FRNT50: 5.4-fold) for NAbs and 32 weeks for S1 RBD (7.9-fold) and whole SARS-CoV-2 (9.4-fold) IgGs. Nine participants (20.9%) tested positive for SARS-CoV-2 RT-PCR between weeks 8 and 32 of booster vaccination; none of them were hospitalized or died. These findings suggest that boosting with TURKOVAC can provide effective protection against COVID-19 for at least 8 weeks and reduce the severity of the disease. Full article
(This article belongs to the Special Issue Advances in COVID-19 Vaccines and Neutralizing Antibody)
Show Figures

Figure 1

22 pages, 1542 KiB  
Article
A SARS-CoV-2 Vaccine Designed for Manufacturability Results in Unexpected Potency and Non-Waning Humoral Response
by Elliot Campbell, Julie Dobkin, Louis J. Osorio, Afsal Kolloli, Santhamani Ramasamy, Ranjeet Kumar, Derek B. Sant’Angelo, Selvakumar Subbian, Lisa K. Denzin and Stephen Anderson
Vaccines 2023, 11(4), 832; https://doi.org/10.3390/vaccines11040832 - 12 Apr 2023
Cited by 1 | Viewed by 7843
Abstract
The rapid development of several highly efficacious SARS-CoV-2 vaccines was an unprecedented scientific achievement that saved millions of lives. However, now that SARS-CoV-2 is transitioning to the endemic stage, there exists an unmet need for new vaccines that provide durable immunity and protection [...] Read more.
The rapid development of several highly efficacious SARS-CoV-2 vaccines was an unprecedented scientific achievement that saved millions of lives. However, now that SARS-CoV-2 is transitioning to the endemic stage, there exists an unmet need for new vaccines that provide durable immunity and protection against variants and can be more easily manufactured and distributed. Here, we describe a novel protein component vaccine candidate, MT-001, based on a fragment of the SARS-CoV-2 spike protein that encompasses the receptor binding domain (RBD). Mice and hamsters immunized with a prime-boost regimen of MT-001 demonstrated extremely high anti-spike IgG titers, and remarkably this humoral response did not appreciably wane for up to 12 months following vaccination. Further, virus neutralization titers, including titers against variants such as Delta and Omicron BA.1, remained high without the requirement for subsequent boosting. MT-001 was designed for manufacturability and ease of distribution, and we demonstrate that these attributes are not inconsistent with a highly immunogenic vaccine that confers durable and broad immunity to SARS-CoV-2 and its emerging variants. These properties suggest MT-001 could be a valuable new addition to the toolbox of SARS-CoV-2 vaccines and other interventions to prevent infection and curtail additional morbidity and mortality from the ongoing worldwide pandemic. Full article
(This article belongs to the Special Issue Advances in COVID-19 Vaccines and Neutralizing Antibody)
Show Figures

Figure 1

11 pages, 1510 KiB  
Article
SARS-CoV-2-Neutralizing Antibody Response and Correlation of Two Serological Assays with Microneutralization
by Amal Souiri, Sanaâ Lemriss, Bouchra El Maliki, Hamadi Falahi, Elmostafa El Fahime and Saâd El Kabbaj
Vaccines 2023, 11(3), 590; https://doi.org/10.3390/vaccines11030590 - 3 Mar 2023
Cited by 3 | Viewed by 1661
Abstract
SARS-CoV-2 has caused a huge pandemic affecting millions of people and resulting innumerous deaths. A better understanding of the correlation between binding antibodies and neutralizing antibodies is necessary to address protective immunity post-infection or vaccination. Here, we investigate the humoral immune response and [...] Read more.
SARS-CoV-2 has caused a huge pandemic affecting millions of people and resulting innumerous deaths. A better understanding of the correlation between binding antibodies and neutralizing antibodies is necessary to address protective immunity post-infection or vaccination. Here, we investigate the humoral immune response and the seroprevalence of neutralizing antibodies following vaccination with adenovirus-based vector in 177 serum samples. A Microneutralization (MN) assay was used as a reference method to assess whether neutralizing antibody titers correlated with a positive signal in two commercially available serological tests:a rapid lateral flow immune-chromatographic assay (LFIA) and an enzyme-linked Fluorescence Assay (ELFA). Neutralizing antibodies were detected in most serum samples (84%). COVID-19 convalescent individuals showed high antibody titers and significant neutralizing activity. Spearman correlation coefficients between the serological and neutralization results ranged from 0.8 to 0.9, suggesting a moderate to strong correlation between commercial immunoassays test results (LFIA and ELFA) and virus neutralization. Full article
(This article belongs to the Special Issue Advances in COVID-19 Vaccines and Neutralizing Antibody)
Show Figures

Figure 1

12 pages, 1305 KiB  
Article
Lack of Evidence on Association between Iron Deficiency and COVID-19 Vaccine-Induced Neutralizing Humoral Immunity
by Arwa A. Faizo, Asma A. Bawazir, Majed N. Almashjary, Ahmed M. Hassan, Fadi S. Qashqari, Ahmed S. Barefah, Sherif A. El-Kafrawy, Thamir A. Alandijany and Esam I. Azhar
Vaccines 2023, 11(2), 327; https://doi.org/10.3390/vaccines11020327 - 1 Feb 2023
Cited by 2 | Viewed by 3085
Abstract
Iron is a crucial micronutrient for immunity induction in response to infections and vaccinations. This study aimed to investigate the effect of iron deficiency on COVID-19-vaccine-induced humoral immunity. We investigated the effectiveness of COVID-19 vaccines (BNT162b2, mRNA-1273, and ChAdOx nCov-2019) in iron-deficient individuals [...] Read more.
Iron is a crucial micronutrient for immunity induction in response to infections and vaccinations. This study aimed to investigate the effect of iron deficiency on COVID-19-vaccine-induced humoral immunity. We investigated the effectiveness of COVID-19 vaccines (BNT162b2, mRNA-1273, and ChAdOx nCov-2019) in iron-deficient individuals (n = 63) and provide a side-by-side comparison to healthy controls (n = 67). The presence of anti-SARS-CoV-2 spike (S) and anti-nucleocapsid (NP) IgG were assessed using in-house S- and NP-based ELISA followed by serum neutralization test (SNT). High concordance between S-based ELISA and SNT results was observed. The prevalence of neutralizing antibodies was 95.24% (60/63) in the study group and 95.52% (64/67) in the controls with no significant difference. The presence/absence of past infection, period since vaccination, vaccine type, and being iron-deficient or having iron-deficiency anemia did not exert any significant effect on the prevalence or titer of anti-SARS-CoV-2 neutralizing antibodies. NP-based ELISA identified individuals unaware of exposure to SARS-CoV-2. Moreover, absence of anti-NP IgG was noted in participants who were previously diagnosed with COVID-19 suggesting the unpredictability of after-infection immunity. To sum up, this study demonstrated an initial lack of evidence on the association between iron deficiency and the effectiveness of COVID-19-vaccine-induced neutralizing humoral immunity. Similar studies with larger sample size remain necessary to obtain comprehensive conclusions about the effect or lack of effect of iron on COVID-19-vaccine effectiveness. Full article
(This article belongs to the Special Issue Advances in COVID-19 Vaccines and Neutralizing Antibody)
Show Figures

Figure 1

11 pages, 1466 KiB  
Article
Anti-SARS-CoV-2 IgM Secondary Response Was Suppressed by Preexisting Immunity in Vaccinees: A Prospective, Longitudinal Cohort Study over 456 Days
by Qiu-Yan Xu, Lin Xie, Xin-Qi Zheng, Xian-Ming Liang, Zhi-Juan Jia, Yan-Yun Liu, Xiao-Yu Liang, Li-Li Liu, Tian-Ci Yang and Li-Rong Lin
Vaccines 2023, 11(1), 188; https://doi.org/10.3390/vaccines11010188 - 16 Jan 2023
Cited by 2 | Viewed by 1856
Abstract
To obtain more insight into IgM in anti-SARS-CoV-2 immunity a prospective cohort study was carried out in 32 volunteers to longitudinally profile the kinetics of the anti-SARS-CoV-2 IgM response induced by administration of a three-dose inactivated SARS-CoV-2 vaccine regimen at 19 serial time [...] Read more.
To obtain more insight into IgM in anti-SARS-CoV-2 immunity a prospective cohort study was carried out in 32 volunteers to longitudinally profile the kinetics of the anti-SARS-CoV-2 IgM response induced by administration of a three-dose inactivated SARS-CoV-2 vaccine regimen at 19 serial time points over 456 days. The first and second doses were considered primary immunization, while the third dose was considered secondary immunization. IgM antibodies showed a low secondary response that was different from the other three antibodies (neutralizing, total, and IgG antibodies). There were 31.25% (10/32) (95% CI, 14.30–48.20%) of participants who never achieved a positive IgM antibody conversion over 456 days after vaccination. The seropositivity rate of IgM antibodies was 68.75% (22/32) (95% CI, 51.80–85.70%) after primary immunization. Unexpectedly, after secondary immunization the seropositivity response rate was only 9.38% (3/32) (95% CI, 1.30–20.10%), which was much lower than that after primary immunization (p = 0.000). Spearman’s correlation analysis indicated a poor correlation of IgM antibodies with the other three antibodies. IgM response in vaccinees was completely different from the response patterns of neutralizing, total, and IgG antibodies following both the primary immunization and the secondary immunization and was suppressed by pre-existing immunity induced by primary immunization. Full article
(This article belongs to the Special Issue Advances in COVID-19 Vaccines and Neutralizing Antibody)
Show Figures

Figure 1

10 pages, 1254 KiB  
Article
Long-Term Dynamic Humoral Response to SARS-CoV-2 mRNA Vaccines in Patients on Peritoneal Dialysis
by Borja Quiroga, María José Soler, Alberto Ortiz, Ron T. Gansevoort, Alba Leyva, José Rojas and Patricia de Sequera
Vaccines 2022, 10(10), 1738; https://doi.org/10.3390/vaccines10101738 - 18 Oct 2022
Cited by 4 | Viewed by 1658
Abstract
Introduction. Patients on peritoneal dialysis (PD) present an impaired humoral response against SARS-CoV-2, at least after the initial vaccination and booster dose. Until now, the effect of a fourth dose has not been established. The aim of the present study is to evaluate [...] Read more.
Introduction. Patients on peritoneal dialysis (PD) present an impaired humoral response against SARS-CoV-2, at least after the initial vaccination and booster dose. Until now, the effect of a fourth dose has not been established. The aim of the present study is to evaluate the long-term dynamics of the humoral response of PD patients to multiple doses of SARS-CoV-2 vaccines, focusing on the effect of the fourth dose. Methods. This is an analysis of the prospective and multicentric SENCOVAC study. We included patients on PD without additional immunosuppression that had received at least 3 SARS-CoV-2 mRNA vaccine doses. We evaluated anti-spike antibody titers after the initial vaccination, third and fourth doses, using prespecified fixed assessments (i.e., baseline, 28 days, 3, 6, and 12 months after completing the initial vaccine schedule). Breakthrough infections were also collected. Results. We included 164 patients on PD (69% males, 62 ± 13 years old). In patients who had received only two doses, the rates of positive humoral response progressively decreased from 96% at 28 days to 80% at 6 months, as did with anti-spike antibody titers. At 6 months, 102 (62%) patients had received the third vaccine dose. Patients with the third dose had higher rates of positive humoral response (p = 0.01) and higher anti-spike antibody titers (p < 0.001) at 6 months than those with only 2 doses. At 12 months, the whole cohort had received 3 vaccine doses, and 44 (27%) patients had an additional fourth dose. The fourth dose was not associated to higher rates of positive humoral response (100 vs. 97%, p = 0.466) or to statistically significant differences in anti-spike antibody titers as compared to three doses (p = 0.371) at 12 months. Prior antibody titers were the only predictor for subsequent higher anti-spike antibody titer (B 0.53 [95%CI 0.27–0.78], p < 0.001). The 2 (1.2%) patients that developed COVID-19 during follow-up had mild disease. Conclusions. PD presents an acceptable humoral response with three doses of SARS-CoV-2 vaccines that improve the progressive loss of anti-spike antibody titers following two vaccine doses. Full article
(This article belongs to the Special Issue Advances in COVID-19 Vaccines and Neutralizing Antibody)
Show Figures

Figure 1

9 pages, 515 KiB  
Article
Relationship between Humoral Response in COVID-19 and Seasonal Influenza Vaccination
by Barbara Poniedziałek, Ewelina Hallmann, Dominika Sikora, Karol Szymański, Katarzyna Kondratiuk, Jakub Żurawski, Piotr Rzymski and Lidia Brydak
Vaccines 2022, 10(10), 1621; https://doi.org/10.3390/vaccines10101621 - 27 Sep 2022
Cited by 7 | Viewed by 2573
Abstract
There is evidence that vaccination against seasonal influenza can improve innate immune responses to COVID-19 and decrease disease severity. However, less is known about whether it could also impact the humoral immunity in SARS-CoV-2 infected patients. The present study aimed to compare the [...] Read more.
There is evidence that vaccination against seasonal influenza can improve innate immune responses to COVID-19 and decrease disease severity. However, less is known about whether it could also impact the humoral immunity in SARS-CoV-2 infected patients. The present study aimed to compare the SARS-CoV-2 specific humoral responses (IgG antibodies against nucleocapsid; anti-N, receptor binding domain; anti-RBD, subunit S2; anti-S2, and envelope protein; anti-E) between non-hospitalized, COVID-19 unvaccinated, and mild COVID-19 convalescent patients who were and were not vaccinated against influenza during the 2019/2020 epidemic season (n = 489 and n = 292, respectively). The influenza-vaccinated group had significantly higher frequency and titers of anti-N antibodies (75 vs. 66%; mean 559 vs. 520 U/mL) and anti-RBD antibodies (85 vs. 76%; mean 580 vs. 540 U/mL). The prevalence and concentrations of anti-S2 and anti-E antibodies did not differ between groups (40–43%; mean 370–375 U/mL and 1.4–1.7%; mean 261–294 U/mL) and were significantly lower compared to those of anti-RBD and anti-N. In both groups, age, comorbidities, and gender did not affect the prevalence and concentrations of studied antibodies. The results indicate that influenza vaccination can improve serum antibody levels produced in response to SARS-CoV-2 infection. Full article
(This article belongs to the Special Issue Advances in COVID-19 Vaccines and Neutralizing Antibody)
Show Figures

Figure 1

Review

Jump to: Editorial, Research, Other

14 pages, 1032 KiB  
Review
Children’s SARS-CoV-2 Infection and Their Vaccination
by Sneh Lata Gupta, Rohit Tyagi, Atika Dhar, Neelam Oswal, Ankita Khandelwal and Rishi Kumar Jaiswal
Vaccines 2023, 11(2), 418; https://doi.org/10.3390/vaccines11020418 - 12 Feb 2023
Cited by 5 | Viewed by 2965
Abstract
SARS-CoV-2, a novel coronavirus, causes respiratory tract infections and other complications in affected individuals, and has resulted in numerous deaths worldwide. The unprecedented pace of its transmission worldwide, and the resultant heavy burden on healthcare systems everywhere, prompted efforts to have effective therapeutic [...] Read more.
SARS-CoV-2, a novel coronavirus, causes respiratory tract infections and other complications in affected individuals, and has resulted in numerous deaths worldwide. The unprecedented pace of its transmission worldwide, and the resultant heavy burden on healthcare systems everywhere, prompted efforts to have effective therapeutic strategies and vaccination candidates available to the global population. While aged and immunocompromised individuals form a high-risk group for COVID-19 and have severe disease outcome, the rate of infections among children has also increased with the emergence of the Omicron variant. In addition, recent reports of threatening SARS-CoV-2-associated complications in children have brought to the forefront an urgent necessity for vaccination. In this article, we discuss the current scenario of SARS-CoV-2 infections in children with a special focus on the differences in their immune system response as compared to adults. Further, we describe the various available COVID-19 vaccines, including the recent bivalent vaccines for children, in detail, intending to increase willingness for their acceptance. Full article
(This article belongs to the Special Issue Advances in COVID-19 Vaccines and Neutralizing Antibody)
Show Figures

Figure 1

28 pages, 4991 KiB  
Review
Therapeutic Role of Neutralizing Antibody for the Treatment against SARS-CoV-2 and Its Emerging Variants: A Clinical and Pre-Clinical Perspective
by Manojit Bhattacharya, Srijan Chatterjee, Bidyut Mallik, Ashish Ranjan Sharma and Chiranjib Chakraborty
Vaccines 2022, 10(10), 1612; https://doi.org/10.3390/vaccines10101612 - 26 Sep 2022
Cited by 12 | Viewed by 1950
Abstract
Since early 2020, the entire world has been facing a disastrous outbreak of the SARS-CoV-2 virus, with massive reporting of death and infections per day. Medical practitioners adopted certain measures such as convalescent plasma therapy, antibody treatment, and injecting vaccines to eradicate the [...] Read more.
Since early 2020, the entire world has been facing a disastrous outbreak of the SARS-CoV-2 virus, with massive reporting of death and infections per day. Medical practitioners adopted certain measures such as convalescent plasma therapy, antibody treatment, and injecting vaccines to eradicate the pandemic. In this review, we have primarily focused on the neutralizing antibodies presently under pre-clinical and clinical trials, focusing on their structures, binding affinity, mechanism of neutralization, and advantages over other therapeutics. We have also enlisted all the nAbs against SARS-CoV-2 and its emerging variants in different phases of clinical trials (phase-1, phase-II, and phase-III). The efficacy of administering antibody cocktails over the normal antibodies and their efficacy for the mutant variants of the SARS-CoV-2 virus in minimizing viral virulence is discussed. The potent neutralizing antibodies have eliminated many of the common problems posed by several other therapeutics. A common mechanism of the antibodies and their relevant sources have also been listed in this review. Full article
(This article belongs to the Special Issue Advances in COVID-19 Vaccines and Neutralizing Antibody)
Show Figures

Figure 1

Other

8 pages, 955 KiB  
Brief Report
Improving the Antigenicity of SARS-CoV-2 Vaccine Genes by Merging Mutations from Different Variants of Concern
by Susanne Herwig, Julia M. Adler, Daria Vladimirova, Jakob Trimpert, Jalid Sehouli and Günter Cichon
Vaccines 2024, 12(3), 248; https://doi.org/10.3390/vaccines12030248 - 27 Feb 2024
Viewed by 823
Abstract
During the COVID-19 pandemic, the early emergence of viral variants repeatedly undermined the effects of vaccination. Our aim here is to explore strategies for improving spike vaccine gene antigenicity by merging mutations from different variants of concern (VOCs) in a single vaccine gene. [...] Read more.
During the COVID-19 pandemic, the early emergence of viral variants repeatedly undermined the effects of vaccination. Our aim here is to explore strategies for improving spike vaccine gene antigenicity by merging mutations from different variants of concern (VOCs) in a single vaccine gene. To this end, newly developed recombinant vaccine genes were designed, cloned into adenoviral vectors, and applied to C57BL/6 mice; then, serum-neutralizing antibodies against the wildtype SARS-CoV-2 strains were determined in neutralization assays. The merger of mutations from different variants of concern (alpha, beta, gamma, and delta) in a single recombinant spike-based vaccine gene provided a substantial improvement in neutralizing immunity to all variants of concern, including the omicron strains. To date, only unmodified spike genes of the original SARS-CoV-2 Wuhan strain (B.1) or dominant variants (BA.1, BA.5, and XBB.1.5) have been used as vaccine genes. The employment of unmodified vaccine genes is afflicted by limited cross-protection among variant strains. In contrast, recombinant vaccine genes that combine mutations from different strains in a single gene hold the potential to broaden and improve immune protection and might help to reduce the need for frequent vaccine adaptations in the future. Full article
(This article belongs to the Special Issue Advances in COVID-19 Vaccines and Neutralizing Antibody)
Show Figures

Figure 1

Back to TopTop