Svoboda | Graniru | BBC Russia | Golosameriki | Facebook
 
 

COVID-19 Vaccines and Immune Response

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 (29 February 2024) | Viewed by 38286

Special Issue Editors


E-Mail Website
Guest Editor
Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
Interests: coronaviruses; MERS-COV; SARS-COV-2; animal models; zoonoses; antiviral

E-Mail Website
Guest Editor Assistant
Clinique Saint-Pierre of Ottignies, Ottignies, Belgium
Interests: immunology

E-Mail Website
Guest Editor
Faculty of Medicine, University of Namur, Namur, Belgium
Interests: pharmacy; life sciences; medicine; drug innovation; thrombosis and hemostasis

E-Mail Website
Guest Editor
Clinique St. Luc Bouge, Namur, Belgium
Interests: analytics; immunoassay; thyroid; statistics; quality control; COVID-19

Special Issue Information

Dear Colleagues,

The COVID-19 pandemic is still ongoing and continues to represent a huge threat to global public health and prosperity around the world. Vaccination constitutes one of our most powerful tools in curbing this pandemic and has allowed us to return to a somewhat normal lifestyle. On the other hand, the vaccination strategy is continuously evolving regarding the number of recommended administrations or the recent introduction of bivalent vaccines. This Special Issue aims to describe recent findings in terms of vaccine response, in light of the evolution of vaccination strategy. Particular attention will be given to articles (original research articles, reviews or commentaries) assessing the immune response in innovative ways, such as the T-cell response, new in vitro or cell-based assays. Studies on populations characterized by lower serological responses (hematological malignancies, transplanted people, immunsuppressed patients, etc.) are especially encouraged.

Dr. Abdullah Algaissi
Jean-Louis Bayart
Dr. Julien Favresse
Dr. Jonathan Douxfils
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

  • SARS-CoV-2
  • Omicron
  • vaccines
  • immunogenicity
  • T-cell
  • B-cell
  • neutralization

Published Papers (21 papers)

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

Research

Jump to: Review, Other

18 pages, 586 KiB  
Article
Correlates of Breakthrough SARS-CoV-2 Infections in People with HIV: Results from the CIHR CTN 328 Study
by Cecilia T. Costiniuk, Terry Lee, Joel Singer, Yannick Galipeau, Corey Arnold, Marc-André Langlois, Judy Needham, Mohammad-Ali Jenabian, Ann N. Burchell, Hasina Samji, Catharine Chambers, Sharon Walmsley, Mario Ostrowski, Colin Kovacs, Darrell H. S. Tan, Marianne Harris, Mark Hull, Zabrina L. Brumme, Hope R. Lapointe, Mark A. Brockman, Shari Margolese, Enrico Mandarino, Suzanne Samarani, Bertrand Lebouché, Jonathan B. Angel, Jean-Pierre Routy, Curtis L. Cooper and Aslam H. Anisadd Show full author list remove Hide full author list
Vaccines 2024, 12(5), 447; https://doi.org/10.3390/vaccines12050447 - 23 Apr 2024
Viewed by 443
Abstract
COVID-19 breakthrough infection (BTI) can occur despite vaccination. Using a multi-centre, prospective, observational Canadian cohort of people with HIV (PWH) receiving ≥2 COVID-19 vaccines, we compared the SARS-CoV-2 spike (S) and receptor-binding domain (RBD)-specific IgG levels 3 and 6 months post second dose, [...] Read more.
COVID-19 breakthrough infection (BTI) can occur despite vaccination. Using a multi-centre, prospective, observational Canadian cohort of people with HIV (PWH) receiving ≥2 COVID-19 vaccines, we compared the SARS-CoV-2 spike (S) and receptor-binding domain (RBD)-specific IgG levels 3 and 6 months post second dose, as well as 1 month post third dose, in PWH with and without BTI. BTI was defined as positivity based on self-report measures (data up to last study visit) or IgG data (up to 1 month post dose 3). The self-report measures were based on their symptoms and either a positive PCR or rapid antigen test. The analysis was restricted to persons without previous COVID-19 infection. Persons without BTI remained COVID-19-naïve until ≥3 months following the third dose. Of 289 participants, 92 developed BTI (31.5 infections per 100 person-years). The median days between last vaccination and BTI was 128 (IQR 67, 176), with the most cases occurring between the third and fourth dose (n = 59), corresponding to the Omicron wave. In analyses adjusted for age, sex, race, multimorbidity, hypertension, chronic kidney disease, diabetes and obesity, a lower IgG S/RBD (log10 BAU/mL) at 1 month post dose 3 was significantly associated with BTI, suggesting that a lower IgG level at this time point may predict BTI in this cohort of PWH. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

19 pages, 11679 KiB  
Article
Innate Responses to the Former COVID-19 Vaccine Candidate CVnCoV and Their Relation to Reactogenicity and Adaptive Immunogenicity
by Olaf-Oliver Wolz, Dominik Vahrenhorst, Gianluca Quintini, Christina Lemberg, Sven D. Koch, Sarah-Katharina Kays, Lisa Walz, Neeraja Kulkarni, Michael Fehlings, Peter Wengenmayer, Jana Heß, Lidia Oostvogels, Sandra Lazzaro, Philipp von Eisenhart-Rothe and Philipp Mann
Vaccines 2024, 12(4), 388; https://doi.org/10.3390/vaccines12040388 - 06 Apr 2024
Viewed by 988
Abstract
Vaccines are highly effective at preventing severe coronavirus disease (COVID-19). With mRNA vaccines, further research is needed to understand the association between immunogenicity and reactogenicity, which is defined as the physical manifestation of an inflammatory response to a vaccination. This study analyzed the [...] Read more.
Vaccines are highly effective at preventing severe coronavirus disease (COVID-19). With mRNA vaccines, further research is needed to understand the association between immunogenicity and reactogenicity, which is defined as the physical manifestation of an inflammatory response to a vaccination. This study analyzed the immune response and reactogenicity in humans, post immunization, to the former SARS-CoV-2 mRNA investigational vaccine CVnCoV (CV-NCOV-001 and CV-NCOV-002 clinical trials). Immunogenicity was investigated using whole-blood RNA sequencing, serum cytokine levels, and SARS-CoV-2-specific antibodies. The T cell responses in peripheral blood were assessed using intracellular cytokine staining (ICS) and high-dimensional profiling in conjunction with SARS-CoV-2 antigen-specificity testing via mass cytometry. Reactogenicity was graded after participants’ first and second doses of CVnCoV using vaccine-related solicited adverse events (AEs). Finally, a Spearman correlation was performed between reactogenicity, humoral immunity, and serum cytokine levels to assess the relationship between reactogenicity and immunogenicity post CVnCoV vaccination. Our findings showed that the gene sets related to innate and inflammatory immune responses were upregulated one day post CVnCoV vaccination, while the gene sets related to adaptive immunity were upregulated predominantly one week after the second dose. The serum levels of IFNα, IFNγ, IP-10, CXCL11, IL-10, and MCP-1 increased transiently, peaking one day post vaccination. CD4+ T cells were induced in all vaccinated participants and low frequencies of CD8+ T cells were detected by ex vivo ICS. Using mass cytometry, SARS-CoV-2 spike-specific CD8+ T cells were induced and were characterized as having an activated effector memory phenotype. Overall, the results demonstrated a positive correlation between vaccine-induced systemic cytokines, reactogenicity, and adaptive immunity, highlighting the importance of the balance between the induction of innate immunity to achieve vaccine efficacy and ensuring low reactogenicity. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

15 pages, 2508 KiB  
Article
SARS-CoV-2 Neutralizing Antibodies in Three African Countries Following Multiple Distinct Immune Challenges
by Diary Juliannie Ny Mioramalala, Rila Ratovoson, Paul Alain Tagnouokam-Ngoupo, Hermine Abessolo Abessolo, Joseph Marie Mindimi Nkodo, Georges Bouting Mayaka, Pierre Claude Tsoungui Atangana, Fanirisoa Randrianarisaona, Pulchérie Pélembi, Romaric Nzoumbou-Boko, Cathy Sandra Goimelle Coti-Reckoundji, Alexandre Manirakiza, Anjanirina Rahantamalala, Rindra Vatosoa Randremanana, Mathurin Cyrille Tejiokem and Matthieu Schoenhals
Vaccines 2024, 12(4), 363; https://doi.org/10.3390/vaccines12040363 - 27 Mar 2024
Viewed by 603
Abstract
Background: The COVID-19 pandemic has affected Madagascar, Cameroon, and the Central African Republic (CAR), with each experiencing multiple waves by mid-2022. This study aimed to evaluate immunity against SARS-CoV-2 strains Wuhan (W) and BA.2 (BA.2) among healthcare workers (HCWs) in these countries, focusing [...] Read more.
Background: The COVID-19 pandemic has affected Madagascar, Cameroon, and the Central African Republic (CAR), with each experiencing multiple waves by mid-2022. This study aimed to evaluate immunity against SARS-CoV-2 strains Wuhan (W) and BA.2 (BA.2) among healthcare workers (HCWs) in these countries, focusing on vaccination and natural infection effects. Methods: HCWs’ serum samples were analyzed for neutralizing antibodies (nAbs) against W and BA.2 variants, with statistical analyses comparing responses between countries and vaccination statuses. Results: Madagascar showed significantly higher nAb titers against both strains compared to CAR and Cameroon. Vaccination notably increased nAb levels against W by 2.6-fold in CAR and 1.8-fold in Madagascar, and against BA.2 by 1.6-fold in Madagascar and 1.5-fold in CAR. However, in Cameroon, there was no significant difference in nAb levels between vaccinated and unvaccinated groups. Conclusion: This study highlights the complex relationship between natural and vaccine-induced immunity, emphasizing the importance of assessing immunity in regions with varied epidemic experiences and low vaccination rates. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

17 pages, 5866 KiB  
Article
Long-Term Dynamic Changes in Hybrid Immunity over Six Months after Inactivated and Adenoviral Vector Vaccination in Individuals with Previous SARS-CoV-2 Infection
by Nungruthai Suntronwong, Sitthichai Kanokudom, Chompoonut Auphimai, Thanunrat Thongmee, Suvichada Assawakosri, Preeyaporn Vichaiwattana, Ritthideach Yorsaeng, Thaneeya Duangchinda, Warangkana Chantima, Pattarakul Pakchotanon, Pornjarim Nilyanimit, Donchida Srimuan, Thaksaporn Thatsanathorn, Natthinee Sudhinaraset, Nasamon Wanlapakorn and Yong Poovorawan
Vaccines 2024, 12(2), 180; https://doi.org/10.3390/vaccines12020180 - 10 Feb 2024
Viewed by 1057
Abstract
Numerous studies have largely focused on short-term immunogenicity in recovered individuals post mRNA vaccination. However, understanding the long-term durability, particularly in inactivated and adenoviral vectored vaccines, remains limited. We evaluated antibody responses, omicron variant neutralization, and IFN-γ responses in 119 previously infected individuals [...] Read more.
Numerous studies have largely focused on short-term immunogenicity in recovered individuals post mRNA vaccination. However, understanding the long-term durability, particularly in inactivated and adenoviral vectored vaccines, remains limited. We evaluated antibody responses, omicron variant neutralization, and IFN-γ responses in 119 previously infected individuals vaccinated with CoronaVac or ChAdOx1 up to six months post-vaccination. Both vaccines elicited robust immune responses in recovered individuals, surpassing those who were infection-naïve, and these persisted above pre-vaccination levels for six months. However, antibody levels declined over time (geometric mean ratio (GMR) = 0.52 for both vaccines). Notably, neutralizing activities against omicron declined faster in ChAdOx1 (GMR = 0.6) compared to CoronaVac recipients (GMR = 1.03). While the first dose of ChAdOx1 adequately induced immune responses in recovered individuals, a second dose demonstrated advantages in omicron variant neutralization and slower decline. Although both vaccines induced T cell responses, the median IFN-γ level at six months returned to pre-vaccination levels. However, more individuals exhibited reactive T cell responses. Extending the interval (13–15 months) between infection and vaccination could enhance antibody levels and broaden neutralization. Together, these findings demonstrate a robust humoral and cellular response that was sustained for at least six months after vaccination, thus guiding optimal vaccination strategies based on prior infection and vaccine platforms. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

13 pages, 3268 KiB  
Article
Immune Evasion of SARS-CoV-2 Omicron Subvariants XBB.1.5, XBB.1.16 and EG.5.1 in a Cohort of Older Adults after ChAdOx1-S Vaccination and BA.4/5 Bivalent Booster
by Rafael Rahal Guaragna Machado, Érika Donizetti Candido, Andressa Simoes Aguiar, Vanessa Nascimento Chalup, Patricia Romão Sanches, Erick Gustavo Dorlass, Deyvid Emanuel Amgarten, João Renato Rebello Pinho, Edison Luiz Durigon and Danielle Bruna Leal Oliveira
Vaccines 2024, 12(2), 144; https://doi.org/10.3390/vaccines12020144 - 30 Jan 2024
Viewed by 1944
Abstract
The recently emerged SARS-CoV-2 Omicron sublineages, including the BA.2-derived XBB.1.5 (Kraken), XBB.1.16 (Arcturus), and EG.5.1 (Eris), have accumulated several spike mutations that may increase immune escape, affecting vaccine effectiveness. Older adults are an understudied group at significantly increased risk of severe COVID-19. Here [...] Read more.
The recently emerged SARS-CoV-2 Omicron sublineages, including the BA.2-derived XBB.1.5 (Kraken), XBB.1.16 (Arcturus), and EG.5.1 (Eris), have accumulated several spike mutations that may increase immune escape, affecting vaccine effectiveness. Older adults are an understudied group at significantly increased risk of severe COVID-19. Here we report the neutralizing activities of 177 sera samples from 59 older adults, aged 62–97 years, 1 and 4 months after vaccination with a 4th dose of ChAdOx1-S (Oxford/AstraZeneca) and 3 months after a 5th dose of Comirnaty Bivalent Original/Omicron BA.4/BA.5 vaccine (Pfizer-BioNTech). The ChAdOx1-S vaccination-induced antibodies neutralized efficiently the ancestral D614G and BA.4/5 variants, but to a much lesser extent the XBB.1.5, XBB.1.16, and EG.5.1 variants. The results showed similar neutralization titers between XBB.1.16 and EG.5.1 and were lower compared to XBB.1.5. Sera from the same individuals boosted with the bivalent mRNA vaccine contained higher neutralizing antibody titers, providing a better cross-protection against Omicron XBB.1.5, XBB.1.16 and EG.5.1 variants. Previous history of infection during the epidemiological waves of BA.1/BA.2 and BA.4/BA.5, poorly enhanced neutralization activity of serum samples against XBBs and EG.5.1 variants. Our data highlight the continued immune evasion of recent Omicron subvariants and support the booster administration of BA.4/5 bivalent vaccine, as a continuous strategy of updating future vaccine booster doses to match newly emerged SARS-CoV-2 variants. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

16 pages, 3525 KiB  
Article
Host Genetic Variation Impacts SARS-CoV-2 Vaccination Response in the Diversity Outbred Mouse Population
by Marta C. Cruz Cisneros, Elizabeth J. Anderson, Brea K. Hampton, Breantié Parotti, Sanjay Sarkar, Sharon Taft-Benz, Timothy A. Bell, Matthew Blanchard, Jacob A. Dillard, Kenneth H. Dinnon III, Pablo Hock, Sarah R. Leist, Emily A. Madden, Ginger D. Shaw, Ande West, Ralph S. Baric, Victoria K. Baxter, Fernando Pardo-Manuel de Villena, Mark T. Heise and Martin T. Ferris
Vaccines 2024, 12(1), 103; https://doi.org/10.3390/vaccines12010103 - 20 Jan 2024
Cited by 1 | Viewed by 1466
Abstract
The COVID-19 pandemic led to the rapid and worldwide development of highly effective vaccines against SARS-CoV-2. However, there is significant individual-to-individual variation in vaccine efficacy due to factors including viral variants, host age, immune status, environmental and host genetic factors. Understanding those determinants [...] Read more.
The COVID-19 pandemic led to the rapid and worldwide development of highly effective vaccines against SARS-CoV-2. However, there is significant individual-to-individual variation in vaccine efficacy due to factors including viral variants, host age, immune status, environmental and host genetic factors. Understanding those determinants driving this variation may inform the development of more broadly protective vaccine strategies. While host genetic factors are known to impact vaccine efficacy for respiratory pathogens such as influenza and tuberculosis, the impact of host genetic variation on vaccine efficacy against COVID-19 is not well understood. To model the impact of host genetic variation on SARS-CoV-2 vaccine efficacy, while controlling for the impact of non-genetic factors, we used the Diversity Outbred (DO) mouse model. We found that DO mice immunized against SARS-CoV-2 exhibited high levels of variation in vaccine-induced neutralizing antibody responses. While the majority of the vaccinated mice were protected from virus-induced disease, similar to human populations, we observed vaccine breakthrough in a subset of mice. Importantly, we found that this variation in neutralizing antibody, virus-induced disease, and viral titer is heritable, indicating that the DO serves as a useful model system for studying the contribution of genetic variation of both vaccines and disease outcomes. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

13 pages, 2276 KiB  
Article
Real-World Study: Hybrid Immunity against SARS-CoV-2 Influences the Antibody Levels and Persistency Lasting More than One Year
by Sitthichai Kanokudom, Jira Chansaenroj, Suvichada Assawakosri, Nungruthai Suntronwong, Ritthideach Yorsaeng, Lakkhana Wongsrisang, Ratchadawan Aeemjinda, Preeyaporn Vichaiwattana, Sirapa Klinfueng, Thaksaporn Thatsanathorn, Sittisak Honsawek and Yong Poovorawan
Vaccines 2023, 11(11), 1693; https://doi.org/10.3390/vaccines11111693 - 07 Nov 2023
Viewed by 888
Abstract
This study investigated the impact of hybrid immunity on antibody responses in the participants who received two to seven doses of the COVID-19 vaccine. The study was conducted between April and June 2023. Out of 771 serum samples analyzed, 71.7% exhibited hybrid immunity [...] Read more.
This study investigated the impact of hybrid immunity on antibody responses in the participants who received two to seven doses of the COVID-19 vaccine. The study was conducted between April and June 2023. Out of 771 serum samples analyzed, 71.7% exhibited hybrid immunity (positive for total anti-N Ig), while 28.3% showed vaccine-induced immunity (negative for total anti-N Ig). Participants were categorized based on the number of vaccine doses: 2, 3, 4, and ≥5. The findings highlight a trend where a higher number of vaccine doses received was associated with a lower infection rate. There was no significant difference in total RBD Ig levels between those who received 3, 4, or ≥5 doses in both the hybrid immunity and vaccination alone groups across all observed durations as follows: <6 months, 6 to <9 months, 9 to <12 months, and ≥12 months. Hybrid immunity consistently maintained higher total RBD Ig levels and durability compared to vaccination alone, with estimated half-lives (T1/2) of 189.5 days versus 106.8 days for vaccine alone. This investigation underscored the potential benefit of hybrid immunity and raised questions about the optimal strategies for further vaccine dosing. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

13 pages, 4389 KiB  
Article
Cytokine and Chemokine Production in Mice Inoculated with NVX-CoV2373 (Nuvaxovid®) in Comparison with Omicron BA.4/5 Bivalent BNT162b2 (Comirnaty®)
by Tetsuo Nakayama, Takashi Ito, Ryoka Ishiyama and Kazuhiko Katayama
Vaccines 2023, 11(11), 1677; https://doi.org/10.3390/vaccines11111677 - 02 Nov 2023
Viewed by 1007
Abstract
A recombinant SARS-CoV-2 spike protein vaccine (NVX-CoV2373) has been licensed and has a lesser incidence of adverse events. To know the immunological mechanisms of adverse events, the production of cytokines and chemokines was investigated in mice inoculated with NVX-CoV2373. Serum IL-6 was detected [...] Read more.
A recombinant SARS-CoV-2 spike protein vaccine (NVX-CoV2373) has been licensed and has a lesser incidence of adverse events. To know the immunological mechanisms of adverse events, the production of cytokines and chemokines was investigated in mice inoculated with NVX-CoV2373. Serum IL-6 was detected on Day 1 of the first and second doses and the IFN-γ, IL-4, IL-10, TNF-α, and IL-6 levels increased on Day 1 of the second dose at the inoculation site. The enhanced production of the inflammatory chemokines (CCL2), homeostatic chemokine (CXCL13), and Th2 chemokine (CCL17) was observed at the inoculation site on Day 1 of the second dose. These findings were compared with data obtained following inoculation with BNT162b2 bivalent vaccine containing omicron BA.4/5. Significantly lower levels of inflammatory chemokines were detected on Day 1 after the first dose of NVX-CoV2373 in sera and inoculation site than those following inoculation with bivalent BNT162b2 (p < 0.01), reflecting a lower incidence of adverse events after immunization with NVX-CoV2373 in humans. NVX-CoV2373 induced significantly higher concentrations of IFN-γ, TNF-α, and IL-10 at the inoculation site obtained on Day 1 of the second dose (p < 0.05). Significant higher levels of Th2 chemokines, CCL11 and CCL17, were induced at the inoculation site on Day 1 of the second dose (p < 0.01) and they explain the booster IgG EIA antibody response after the second dose of NVX-CoV2373. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

15 pages, 1908 KiB  
Article
Comparative Immune Response after Vaccination with SOBERANA® 02 and SOBERANA® plus Heterologous Scheme and Natural Infection in Young Children
by Rocmira Pérez-Nicado, Chiara Massa, Laura Marta Rodríguez-Noda, Anja Müller, Rinaldo Puga-Gómez, Yariset Ricardo-Delgado, Beatriz Paredes-Moreno, Meiby Rodríguez-González, Marylé García-Ferrer, Ilianet Palmero-Álvarez, Aniurka Garcés-Hechavarría, Daniel G. Rivera, Yury Valdés-Balbín, Vicente Vérez-Bencomo, Dagmar García-Rivera and Barbara Seliger
Vaccines 2023, 11(11), 1636; https://doi.org/10.3390/vaccines11111636 - 25 Oct 2023
Viewed by 2882
Abstract
(1) Background: In children, SARS-CoV-2 infection is mostly accompanied by mild COVID-19 symptoms. However, multisystem inflammatory syndrome (MIS-C) and long-term sequelae are often severe complications. Therefore, the protection of the pediatric population against SARS-CoV-2 with effective vaccines is particularly important. Here, we compare [...] Read more.
(1) Background: In children, SARS-CoV-2 infection is mostly accompanied by mild COVID-19 symptoms. However, multisystem inflammatory syndrome (MIS-C) and long-term sequelae are often severe complications. Therefore, the protection of the pediatric population against SARS-CoV-2 with effective vaccines is particularly important. Here, we compare the humoral and cellular immune responses elicited in children (n = 15, aged 5–11 years) vaccinated with the RBD-based vaccines SOBERANA® 02 and SOBERANA® Plus combined in a heterologous scheme with those from children (n = 10, aged 4–11 years) who recovered from mild symptomatic COVID-19. (2) Methods: Blood samples were taken 14 days after the last dose for vaccinated children and 45–60 days after the infection diagnosis for COVID-19 recovered children. Anti-RBD IgG and ACE2-RBD inhibition were assessed by ELISA; IgA, cytokines, and cytotoxic-related proteins were determined by multiplex assays. Total B and T cell subpopulations and IFN-γ release were measured by multiparametric flow cytometry using a large panel of antibodies after in vitro stimulation with S1 peptides. (3) Results: Significant higher levels of specific anti-RBD IgG and IgA and ACE2-RBD inhibition capacity were found in vaccinated children in comparison to COVID-19 recovered children. Th1-like and Th2-like CD4+ T cells were also significantly higher in vaccinated subjects. IFN-γ secretion was higher in central memory CD4+ T cells of COVID-19 recovered children, but no differences between both groups were found in the CD4+ and CD8+ T cell effector, terminal effector, and naïve T cell subpopulations. In contrast to low levels of IL-4, high levels of IL-2, IL-6, IFN-γ, and IL-10 suggest a predominant Th1 cell polarization. Cytotoxic-related proteins granzyme A and B, perforin, and granulin were also found in the supernatant after S1 stimulation in both vaccinated and recovered children. (4) Conclusions: Vaccination with the heterologous scheme of SOBERANA® 02/SOBERANA® Plus induces a stronger antibody and cellular immune response compared to natural infections in young children. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

16 pages, 3032 KiB  
Article
Longitudinal Analysis of SARS-CoV-2-Specific Cellular and Humoral Immune Responses and Breakthrough Infection following BNT162b2/BNT162b2/BNT162b2 and ChAdOx1/ChAdOx1/BNT162b2 Vaccination: A Prospective Cohort in Naive Healthcare Workers
by Geon Young Ko, Jihyun Lee, Hyunjoo Bae, Ji Hyeong Ryu, Hye-Sun Park, Hyunhye Kang, Jin Jung, Ae-Ran Choi, Raeseok Lee, Dong-Gun Lee and Eun-Jee Oh
Vaccines 2023, 11(10), 1613; https://doi.org/10.3390/vaccines11101613 - 19 Oct 2023
Cited by 3 | Viewed by 1106
Abstract
Assessing immune responses post-SARS-CoV-2 vaccination is crucial for optimizing vaccine strategies. This prospective study aims to evaluate immune responses and breakthrough infection in 235 infection-naïve healthcare workers up to 13–15 months after initial vaccination in two vaccine groups (108 BNT/BNT/BNT and 127 ChAd/ChAd/BNT). [...] Read more.
Assessing immune responses post-SARS-CoV-2 vaccination is crucial for optimizing vaccine strategies. This prospective study aims to evaluate immune responses and breakthrough infection in 235 infection-naïve healthcare workers up to 13–15 months after initial vaccination in two vaccine groups (108 BNT/BNT/BNT and 127 ChAd/ChAd/BNT). Immune responses were assessed using the interferon-gamma enzyme-linked immunospot (ELISPOT) assay, total immunoglobulin, and neutralizing activity through surrogate virus neutralization test at nine different time points. Both groups exhibited peak responses one to two months after the second or third dose, followed by gradual declines over six months. Notably, the ChAd group exhibited a gradual increase in ELISPOT results, but their antibody levels declined more rapidly after reaching peak response compared to the BNT group. Six months after the third dose, both groups had substantial cellular responses, with superior humoral responses in the BNT group (p < 0.05). As many as 55 breakthrough infection participants displayed higher neutralization activities against Omicron variants, but similar cellular responses compared to 127 infection-naïve individuals, suggesting cross-immunity. Distinct neutralization classifications (<30%, >80% inhibition) correlated with different ELISPOT results. Our study reveals diverse immune response patterns based on vaccine strategies and breakthrough infections, emphasizing the importance of understanding these dynamics for optimized vaccination decisions. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

12 pages, 1393 KiB  
Article
Comparison of Humoral Response between Third and Fourth Doses of COVID-19 Vaccine in Hemodialysis Patients
by Yoosun Joo, Dae Kyu Kim, Yun Gi Jeon, Ah-Ra Kim, Hyeon Nam Do, Soo-Young Yoon, Jin Sug Kim, Su Woong Jung, Hyeon Seok Hwang, Ju-Young Moon, Kyung Hwang Jeong, Sang-Ho Lee, So-Young Kang and Yang Gyun Kim
Vaccines 2023, 11(10), 1584; https://doi.org/10.3390/vaccines11101584 - 12 Oct 2023
Cited by 1 | Viewed by 1040
Abstract
Dialysis patients are more likely to die or become hospitalized from coronavirus disease 2019 (COVID-19). Currently, only a few studies have evaluated the efficacy of a fourth booster vaccination in hemodialysis (HD) patients and there is not enough evidence to recommend for or [...] Read more.
Dialysis patients are more likely to die or become hospitalized from coronavirus disease 2019 (COVID-19). Currently, only a few studies have evaluated the efficacy of a fourth booster vaccination in hemodialysis (HD) patients and there is not enough evidence to recommend for or against a fourth booster vaccination. This study compared the humoral response and disease severity of patients on HD who received either three or four doses of COVID-19 vaccine. A total of 88 patients were enrolled. Humoral response to vaccination was measured by quantifying immunoglobulin G levels against the receptor binding domain of SARS-CoV-2 (anti-RBD IgG) at five different times and plaque reduction neutralization tests (PRNT) at two different times after vaccination over a period of 18 months. Antibody levels were measured at approximately two-month intervals after the first and second dose, then four months after the third dose, and then one to six months after the fourth dose of vaccine. PRNT was performed two months after the second and four months after the third dose of vaccine. We classified patients into four groups according to the number of vaccine doses and presence of COVID-19 infection. Severe infection was defined as hospital admission for greater than or equal to two weeks or death. There was no difference in antibody levels between naïve and infected patients except after a fourth vaccination, which was effective for increasing antibodies in infection-naïve patients. Age, sex, body mass index (BMI), dialysis vintage, and presence of diabetes mellitus (DM) did not show a significant correlation with antibody levels. Four patients who experienced severe COVID-19 disease tended to have lower antibody levels prior to infection. A fourth dose of SARS-CoV-2 vaccine significantly elevated antibodies in infection-naïve HD patients and may be beneficial for HD patients who have not been previously infected with SARS-CoV-2 for protection against severe infection. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

16 pages, 2632 KiB  
Article
Exploring the Effects of Vitamin D and Vitamin A Levels on the Response to COVID-19 Vaccine
by Hassan M. Kofahi, Baha’ R. Badran, Refat M. Nimer, Ali M. Atoom and Shefa’ M. Al Hersh
Vaccines 2023, 11(9), 1509; https://doi.org/10.3390/vaccines11091509 - 21 Sep 2023
Viewed by 1034
Abstract
COVID-19 vaccines were developed at an unprecedented speed in history. The factors affecting the response to COVID-19 vaccines are not clear. Herein, the effects of vitamin D and vitamin A (retinol) levels on the response to the BNT162b2 vaccine were explored. A total [...] Read more.
COVID-19 vaccines were developed at an unprecedented speed in history. The factors affecting the response to COVID-19 vaccines are not clear. Herein, the effects of vitamin D and vitamin A (retinol) levels on the response to the BNT162b2 vaccine were explored. A total of 124 vaccine recipients were recruited from the general population attending vaccination centers in Irbid, Jordan. Blood samples were collected immediately before receiving the first vaccine dose (D0) and three weeks later (D21). Baseline (D0) levels of 25-hydroxyvitamin D [25(OH)D], retinol, and SARS-CoV-2 S1 IgG antibodies were measured with ELISA. The response to the BNT162b2 vaccine was tested by measuring the levels and avidity of SARS-CoV-2 S1 IgG antibodies on D21. The participants were divided into two groups, unexposed and exposed, based on the D0 SARS-CoV-2 antibody results. No significant correlation was found between the levels of 25(OH)D or retinol and the levels, avidity, or fold increase of antibodies in both groups. Similarly, no significant difference in antibody response was found between 25(OH)D status groups, retinol status groups, or combined status groups. These findings show that the baseline vitamin D or vitamin A levels have no effect on the short-term response to a single dose of BNT162b2 vaccine. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

23 pages, 2511 KiB  
Article
Aerosol Inhalation of Chimpanzee Adenovirus Vectors (ChAd68) Expressing Ancestral or Omicron BA.1 Stabilized Pre–Fusion Spike Glycoproteins Protects Non–Human Primates against SARS-CoV-2 Infection
by Shen Wang, Mian Qin, Long Xu, Ting Mu, Ping Zhao, Bing Sun, Yue Wu, Lingli Song, Han Wu, Weicheng Wang, Xingwen Liu, Yanyan Li, Fengmei Yang, Ke Xu, Zhanlong He, Michel Klein and Ke Wu
Vaccines 2023, 11(9), 1427; https://doi.org/10.3390/vaccines11091427 - 28 Aug 2023
Viewed by 1167
Abstract
Current COVID-19 vaccines are effective countermeasures to control the SARS-CoV-2 virus pandemic by inducing systemic immune responses through intramuscular injection. However, respiratory mucosal immunization will be needed to elicit local sterilizing immunity to prevent virus replication in the nasopharynx, shedding, and transmission. In [...] Read more.
Current COVID-19 vaccines are effective countermeasures to control the SARS-CoV-2 virus pandemic by inducing systemic immune responses through intramuscular injection. However, respiratory mucosal immunization will be needed to elicit local sterilizing immunity to prevent virus replication in the nasopharynx, shedding, and transmission. In this study, we first compared the immunoprotective ability of a chimpanzee replication–deficient adenovirus–vectored COVID-19 vaccine expressing a stabilized pre–fusion spike glycoprotein from the ancestral SARS-CoV-2 strain Wuhan–Hu–1 (BV-AdCoV-1) administered through either aerosol inhalation, intranasal spray, or intramuscular injection in cynomolgus monkeys and rhesus macaques. Compared with intranasal administration, aerosol inhalation of BV-AdCoV-1 elicited stronger humoral and mucosal immunity that conferred excellent protection against SARS-CoV-2 infection in rhesus macaques. Importantly, aerosol inhalation induced immunity comparable to that obtained by intramuscular injection, although at a significantly lower dose. Furthermore, to address the problem of immune escape variants, we evaluated the merits of heterologous boosting with an adenovirus–based Omicron BA.1 vaccine (C68–COA04). Boosting rhesus macaques vaccinated with two doses of BV-AdCoV-1 with either the homologous or the heterologous C68–COA04 vector resulted in cross–neutralizing immunity against WT, Delta, and Omicron subvariants, including BA.4/5 stronger than that obtained by administering a bivalent BV-AdCoV-1/C68–COA04 vaccine. These results demonstrate that the administration of BV-AdCoV-1 or C68–COA04 via aerosol inhalation is a promising approach to prevent SARS-CoV-2 infection and transmission and curtail the pandemic spread. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

11 pages, 868 KiB  
Article
Blood CD8+ Naïve T-Cells Identify MS Patients with High Probability of Optimal Cellular Response to SARS-CoV-2 Vaccine
by Alexander Rodero-Romero, Susana Sainz de la Maza, José Ignacio Fernández-Velasco, Enric Monreal, Paulette Esperanza Walo-Delgado, Juan Luis Chico-García, Noelia Villarrubia, Fernando Rodríguez-Jorge, Rafael Rodríguez-Ramos, Jaime Masjuan, Lucienne Costa-Frossard and Luisa María Villar
Vaccines 2023, 11(9), 1399; https://doi.org/10.3390/vaccines11091399 - 22 Aug 2023
Viewed by 836
Abstract
This single-center study included 68 multiple sclerosis (MS) patients who received the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination from one of several approved vaccine preparations in Spain. Blood samples were collected one to three months after the second dose of the [...] Read more.
This single-center study included 68 multiple sclerosis (MS) patients who received the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination from one of several approved vaccine preparations in Spain. Blood samples were collected one to three months after the second dose of the vaccine had been administered. Cellular immune responses to the vaccine were assessed using QuantiFERON analysis, and peripheral blood mononuclear cell subsets were assayed using flow cytometry. Response associated with higher percentages of total lymphocytes, naïve CD4+ T-cells (p = 0.028), CD8+ T-cells (p = 0.013), and, mostly, naïve CD8+ T-cells (p = 0.0003). These results were confirmed by analyzing absolute numbers (p = 0.019; p = 0.002, and p = 0.0003, respectively). Naïve CD8 T-cell numbers higher than 17 cells/μL were closely associated with an optimal cellular response to SARS-CoV-2 vaccination (odds ratio: 24.0, confidence interval: 4.8–460.3; p = 0.0001). This finding clearly shows that independent of the treatment received, higher numbers of naïve CD8+ T-cells yield a strong cellular response to SARS-CoV-2 vaccines in MS patients. If this finding is validated with other viruses/vaccines, it could provide a good tool for identifying MS patients undergoing treatment who will develop strong cellular responses to anti-virus vaccines. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

17 pages, 1008 KiB  
Article
COVID-19 Vaccination Effectiveness in the General Population of an Italian Province: Two Years of Follow-Up
by Annalisa Rosso, Maria Elena Flacco, Graziella Soldato, Giuseppe Di Martino, Cecilia Acuti Martellucci, Roberto Carota, Marco De Benedictis, Graziano Di Marco, Rossano Di Luzio, Matteo Fiore, Antonio Caponetti and Lamberto Manzoli
Vaccines 2023, 11(8), 1325; https://doi.org/10.3390/vaccines11081325 - 04 Aug 2023
Cited by 2 | Viewed by 10730
Abstract
We carried out a cohort study on the overall population of the province of Pescara, Italy, to assess the real-world effectiveness of SARS-CoV-2 vaccination against infection, severe, or lethal COVID-19, two years after the start of the vaccination campaign. We included all the [...] Read more.
We carried out a cohort study on the overall population of the province of Pescara, Italy, to assess the real-world effectiveness of SARS-CoV-2 vaccination against infection, severe, or lethal COVID-19, two years after the start of the vaccination campaign. We included all the resident or domiciled subjects, and extracted the official demographic, vaccination, COVID-19, hospital and co-pay exemption datasets from 1 January 2021, up to 15 February 2023. Cox proportional hazards analyses were adjusted for gender, age, diabetes, hypertension, COPD, major cardio- and cerebrovascular events, cancer, and kidney diseases. Throughout the follow-up (466 days on average), 186,676 subjects received greater than or equal to three vaccine doses (of ChAdOx1 nCoV-19, BNT162b2, mRNA-1273, NVX-CoV2373, or JNJ-78436735), 47,610 two doses, 11,452 one dose, and 44,989 none. Overall, 40.4% of subjects were infected with SARS-CoV-2. Of them, 2.74% had severe or lethal (1.30%) COVID-19. As compared to the unvaccinated, the individuals who received greater than or equal to one booster dose showed a ≥85% lower risk of severe or lethal COVID-19. A massive impact of vaccination was found among the elderly: 22.0% of the unvaccinated, infected individuals died, as opposed to less than 3% of those who received greater than or equal to three vaccine doses. No protection against infection was observed, although this finding was certainly influenced by the Italian restriction policies to control the pandemic. Importantly, during the Omicron predominance period, only the group who received at least a booster dose showed a reduced risk of COVID-19-related death. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

11 pages, 1737 KiB  
Article
Recipient-Reported Reactogenicity of Different SARS-CoV-2 Vaccination Regimens among Healthcare Professionals and Police Staff in Germany
by Katharina Rau, Edgar von Heeringen, Nina Bühler, Stefan Wagenpfeil, Sören L. Becker and Sophie Schneitler
Vaccines 2023, 11(7), 1147; https://doi.org/10.3390/vaccines11071147 - 25 Jun 2023
Cited by 1 | Viewed by 1061
Abstract
The rapid availability of effective vaccines against SARS-CoV-2 was key during the COVID-19 pandemic. However, vaccine hesitancy and relatively low vaccine coverage rates among the general population and particularly vulnerable populations such as healthcare staff reduced the potential benefits of these vaccines. During [...] Read more.
The rapid availability of effective vaccines against SARS-CoV-2 was key during the COVID-19 pandemic. However, vaccine hesitancy and relatively low vaccine coverage rates among the general population and particularly vulnerable populations such as healthcare staff reduced the potential benefits of these vaccines. During the early phase of the pandemic, fear of vaccine-related adverse events was common among individuals who refused vaccination. Between March and May 2021, we comparatively assessed the self-reported reactogenicity of different SARS-CoV-2 prime-boost regimens using mRNA-based (BNT162b2 and mRNA-1273) and vector-based vaccines (ChAdOx1 nCoV-19) in (a) healthcare workers (HCW), and (b) police staff from southwest Germany. The majority of participants (71.8%; 1564/2176) received a homologous vaccination. Among HCW, 75.0% were female, whereas 70.0% of police staff were male. The most frequently reported reactions following the first vaccine administration were pain at the injection site (77.94%; 1696/2176), tiredness (51.75%; 1126/2176), and headache (40.44%; 880/2176), which were more commonly reported by HCW as compared to police staff. In homologous, mRNA-based and heterologous vaccination schedules, more reactions were reported after the second vaccine dose. We conclude that the frequency and intensity of self-perceived vaccine reactogenicity may differ between specific population groups and might be mitigated by tailored communication strategies. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

13 pages, 1709 KiB  
Article
Platelet Activation and Cytokine Release of Interleukin-8 and Interferon-Gamma-Induced Protein 10 after ChAdOx1 nCoV-19 Coronavirus Vaccine Injection
by Chih-Lung Shen, Tso-Fu Wang, Chao-Zong Liu and Yi-Feng Wu
Vaccines 2023, 11(2), 456; https://doi.org/10.3390/vaccines11020456 - 16 Feb 2023
Cited by 1 | Viewed by 1650 | Correction
Abstract
Coronavirus disease 2019 (COVID-19) vaccines are associated with serious thromboembolic or thrombocytopenic events including vaccine-induced immune thrombocytopenia and thrombosis and immune thrombocytopenia, particularly AZD1222/ChAdOx1. According to the proposed mechanism, COVID-19 vaccines stimulate inflammation and platelet activation. In this study, we analyzed the role [...] Read more.
Coronavirus disease 2019 (COVID-19) vaccines are associated with serious thromboembolic or thrombocytopenic events including vaccine-induced immune thrombocytopenia and thrombosis and immune thrombocytopenia, particularly AZD1222/ChAdOx1. According to the proposed mechanism, COVID-19 vaccines stimulate inflammation and platelet activation. In this study, we analyzed the role of AZD1222/ChAdOx1 vaccines in the activation of platelets and the release of anti-PF4 antibodies and inflammatory cytokines in a cohort of healthy donors without vaccine-induced immune thrombotic thrombocytopenia (VITT). Forty-eight healthy volunteers were enrolled in this study. Blood samples were collected from peripheral blood at three time points: before vaccination and 1 and 7 days after vaccination. Compared with the prevaccination data, a decrease in the leukocyte and platelet counts was observed 1 day after vaccination, which recovered 7 days after injection. The percentage of activated GPIIb/IIIa complex (PAC-1) under high ADP or thrombin receptor-activating peptide stimulation increased 1 day after vaccination. Furthermore, interleukin-8 (IL-8) and interferon-gamma-induced protein 10 (IP-10) increased significantly. Additionally, platelet activation and inflammation, with the release of cytokines, were observed; however, none of the individuals developed VITT. Mild thrombocytopenia with platelet activation and inflammation with an elevation of IL-8 and IP-10 were observed after AZ vaccination. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

11 pages, 1493 KiB  
Article
Neutralizing Antibodies as Predictors of Vaccine Breakthrough Infection in Healthcare Workers Vaccinated with or without a Heterologous Booster Dose: A Cohort Study during the Third COVID-19 Wave in Peru
by Miguel Hueda-Zavaleta, Juan C. Gómez de la Torre, José Alonso Cáceres-DelAguila, Cecilia Muro-Rojo, Nathalia De La Cruz-Escurra, Cesar Copaja-Corzo, Carlos J. Aragón-Ayala and Vicente A. Benítes-Zapata
Vaccines 2023, 11(2), 447; https://doi.org/10.3390/vaccines11020447 - 15 Feb 2023
Cited by 2 | Viewed by 1908
Abstract
We evaluated neutralizing antibody (NAbs) levels as a protective factor against vaccine breakthrough infection (VBI) in healthcare workers (HCWs) during the third COVID-19 wave in Peru. This retrospective cohort study employed the information from a private laboratory in Lima (Peru) of HCW who [...] Read more.
We evaluated neutralizing antibody (NAbs) levels as a protective factor against vaccine breakthrough infection (VBI) in healthcare workers (HCWs) during the third COVID-19 wave in Peru. This retrospective cohort study employed the information from a private laboratory in Lima (Peru) of HCW who received only two BBIBP-CorV vaccines or (additionally) a heterologous booster with BNT162b2. We evaluated the association between the VBI and the levels of NAbs at 21, 90, 180, and 210 days after the BBIBP-CorV second dose. NAbs were calculated with the cPass™ SARS-CoV-2 Neutralization Antibody Detection kit (surrogate virus neutralization test (sVNT)) and the Elecsys® anti-SARS-CoV-2 S Test. Of the 435 HCW evaluated, 31.72% had an infection previous to vaccination, 68.28% received a booster dose, and 23.21% had a VBI during the third wave. The variables associated with a lower risk of VBI were male sex (aRR: 0.43) and those who had (180 days after BBIBP-CorV inoculation) NAbs levels ≥ 60% (aRR: 0.58) and ≥90% (aRR: 0.59) on cPass™, and ≥500 with Elecsys® (aRR: 0.58). HCW whose NAbs persisted at higher levels six months after the BBIBP-CorV showed a lower risk of suffering from a VBI during the third COVID-19 wave. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

11 pages, 967 KiB  
Article
Durability of ChAdOx1 nCoV-19 (Covishield®) Vaccine Induced Antibody Response in Health Care Workers
by Alka Verma, Amit Goel, Harshita Katiyar, Prachi Tiwari, Mayank, Asari Sana, Dheeraj Khetan, Dharmendra Singh Bhadauria, Ajay Raja, Neelam Khokher, Shalimar, Ratendra Kumar Singh and Amita Aggarwal
Vaccines 2023, 11(1), 84; https://doi.org/10.3390/vaccines11010084 - 30 Dec 2022
Cited by 3 | Viewed by 1438
Abstract
(i) Background: ChAdOx1 nCoV-19 (Covishield®) vaccine is widely used in India. We studied the Covishield® induced antibody response and its durability among health care workers (HCWs) (ii) Method: HCWs received two doses (0.5 mL) four weeks apart. Blood specimens, collected [...] Read more.
(i) Background: ChAdOx1 nCoV-19 (Covishield®) vaccine is widely used in India. We studied the Covishield® induced antibody response and its durability among health care workers (HCWs) (ii) Method: HCWs received two doses (0.5 mL) four weeks apart. Blood specimens, collected before each dose, day (D)60, D150 and D270 after second dose, were tested for anti-spike antibody (ASAb) titre and neutralising antibody (%) (NAb) using Elecsys Anti-SARS-CoV-2 S (Roche) and SARS-CoV-2 NAb ELISA Kit (Invitrogen), respectively. Data are expressed as proportions and median (interquartile range) and compared using non-parametric (iii) Result: Among 135 HCWs (83 males; age 45 (37–53); 36 had pre-existing ASAb), 29 (21.5%) acquired COVID-19 after 60 (39–68) days of vaccination. ASAb titre before second dose and at D60, D150, D270 were 77.2 (19.4–329.4), 512 (114.5–9212), 149 (51.6–2283) and 2079 (433.9–8644) U/mL, respectively. Compared to those without pre-existing ASAb, titres were significantly higher before second dose (5929 vs. 41, p < 0.001), D60 (3395 vs. 234, p = 0.007) and D150 (1805 vs. 103, p < 0.001) in participants with pre-existing ASAb; NAb were also higher (80 vs. 18, p < 0.001) before second dose. Between those who acquired infection or not after vaccination, ASAb titres were comparable before second dose (77 vs. 78, p = 0.362) but significantly higher at D60 (14,019 vs. 317, p < 0.001) and D150 (2062 vs. 121, p = 0.002) in the former group, though NAb percentage were higher at D60 (87 vs. 27, p < 0.001) and D150 (79 vs. 25, p = 0.007) only (iv) Conclusions: Covishield® induces a higher antibody titre in those with pre-existing ASAb. The vaccine induced antibody starts falling 5 months after vaccination. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

Review

Jump to: Research, Other

17 pages, 874 KiB  
Review
Assessment of the Immune Response in Patients with Insulin Resistance, Obesity, and Diabetes to COVID-19 Vaccination
by Jędrzej Warpechowski, Paula Leszczyńska, Dominika Juchnicka, Adam Olichwier, Łukasz Szczerbiński and Adam Jacek Krętowski
Vaccines 2023, 11(7), 1203; https://doi.org/10.3390/vaccines11071203 - 05 Jul 2023
Cited by 2 | Viewed by 1820
Abstract
The SARS-CoV-19 pandemic overwhelmed multiple healthcare systems across the world. Patients with underlying medical conditions such as obesity or diabetes were particularly vulnerable, had more severe symptoms, and were more frequently hospitalized. To date, there have been many studies on the severity of [...] Read more.
The SARS-CoV-19 pandemic overwhelmed multiple healthcare systems across the world. Patients with underlying medical conditions such as obesity or diabetes were particularly vulnerable, had more severe symptoms, and were more frequently hospitalized. To date, there have been many studies on the severity of SARS-CoV-2 in patients with metabolic disorders, but data on the efficiency of vaccines against COVID-19 are still limited. This paper aims to provide a comprehensive overview of the effectiveness of COVID-19 vaccines in individuals with diabetes, insulin resistance, and obesity. A comparison is made between the immune response after vaccination in patients with and without metabolic comorbidities. Additionally, an attempt is made to highlight the mechanisms of immune stimulation affected by SARS-CoV-2 vaccines and how metabolic comorbidities modulate these mechanisms. The focus is on the most common COVID-19 vaccines, which include mRNA vaccines such as Pfizer-BioNTech and Moderna, as well as viral vector vaccines such as AstraZeneca and Johnson & Johnson. Furthermore, an effort is made to clarify how the functional differences between these vaccines may impact the response in individuals with metabolic disorders, drawing from available experimental data. This review summarizes the current knowledge regarding the post-vaccination response to COVID-19 in the context of metabolic comorbidities such as diabetes, insulin resistance, and obesity. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

Other

Jump to: Research, Review

11 pages, 416 KiB  
Systematic Review
ANCA Associated Glomerulonephritis Following SARS-CoV-2 Vaccination: A Case Series and Systematic Review
by Theerachai Thammathiwat, Athiphat Banjongjit, Kroonpong Iampenkhae, Natavudh Townamchai and Talerngsak Kanjanabuch
Vaccines 2023, 11(5), 983; https://doi.org/10.3390/vaccines11050983 - 15 May 2023
Cited by 6 | Viewed by 2217
Abstract
Vaccines against SARS-CoV-2 (COVID-19) proved beneficial for COVID-19 disease attenuation and preventing virus spreading. Cumulative reports of the rarity of antineutrophil cytoplasmic autoantibodies (ANCA)-associated vasculitis (AAV) raise concerns about its relationship with COVID-19 vaccination. Several case reports described ANCA-associated pauci-immune glomerulonephritis (ANCA-GN) following [...] Read more.
Vaccines against SARS-CoV-2 (COVID-19) proved beneficial for COVID-19 disease attenuation and preventing virus spreading. Cumulative reports of the rarity of antineutrophil cytoplasmic autoantibodies (ANCA)-associated vasculitis (AAV) raise concerns about its relationship with COVID-19 vaccination. Several case reports described ANCA-associated pauci-immune glomerulonephritis (ANCA-GN) following COVID-19 vaccination with some uniqueness. We systematically reviewed COVID-19 vaccine-induced ANCA-GN from PubMed, SCOPUS, and Cochrane library databases until 1 January 2023 according to PRISMA guidelines and presented our three cases. Twenty-six cases from 25 articles, including our 3 cases, were analyzed. Most cases were diagnosed following the second dose of the COVID-19 vaccine (59%) with a median (IQR) interval onset of 14 (16) days. The highest prevalence was related to the mRNA-type vaccine. Anti-myeloperoxidase (MPO) ANCA was far more common than the other ANCAs, with various positive autoantibodies. Fourteen cases (out of 29 cases, 48%) had extra-kidney AAV manifestation. Although severe kidney injury was observed in 10/29 (34%), remission was achieved in 89% (25/28) with no death. The mechanisms of the vaccine-inducing ANCA-GN were postulated here. Since ANCA-GN after the COVID-19 vaccine was rare, the benefit of the COVID-19 vaccine could outweigh the risk of ANCA-GN side effects in the pandemic era. Full article
(This article belongs to the Special Issue COVID-19 Vaccines and Immune Response)
Show Figures

Figure 1

Back to TopTop