Cellular and Humoral Immunity after COVID-19 Vaccination

A special issue of Vaccines (ISSN 2076-393X). This special issue belongs to the section "Cellular/Molecular Immunology".

Deadline for manuscript submissions: closed (30 June 2023) | Viewed by 25708

Special Issue Editor

Department of Nephrology, Transplantology and Internal Medicine, Medical University of Gdańsk, 80-210 Gdańsk, Poland
Interests: hypertension; chronic kidney disease; acute kidney injury; renin-angiotensin system; kidney transplantation; hemodialysis; COVID-19; vaccination
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

The SARS-CoV-2 pandemic is spreading around the world with death and paralysis in healthcare systems. Vaccination is the safest and most effective tool to achieve a control the pandemic and build herd immunity. Vaccines against COVID-19 have been developed that are based on various platforms, including mRNA and DNA, inactivated viruses, live attenuated viruses, protein subunits, virus-like particles, and  other viruses as vectors. The aim of vaccination is to generate a protective antibody titer to prevent infection and create long-term immune memory by the pool of memory cells, including T cells, high-affinity memory B cells, and plasma cells, all able to attenuate the severity of the disease course. Many aspects of the immune response following vaccination against COVID-19  remain unclear. This Special Issue of the Vaccines will summarize the current state of knowledge, fields to explore in particular referring (but not limited to) the following issues:

  • cellular and humoral response in immunocompetent and immunocompromised population.
  • differences in the immune response between vaccines
  • duration of specific humoral and cellular immunity after primary and booster vaccination
  • schemes of vaccination enhancing immune responses in immunocompromised patients
  • comparing immune response after vaccination with the natural immunity of convalescents
  • local and systemic reactogenicity and long-term safety of different vaccines

In this Special Issue, original research articles and reviews are welcome. We look forward to your contributions.

Prof. Dr. Leszek Tylicki
Guest Editor

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
  • SARS-CoV-2
  • humoral immunity
  • cellular immunity
  • vaccine
  • vaccination

Related Special Issue

Published Papers (12 papers)

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

Editorial

Jump to: Research, Review

4 pages, 181 KiB  
Editorial
Immunity after Vaccination against COVID-19
by Leszek Tylicki
Vaccines 2023, 11(11), 1723; https://doi.org/10.3390/vaccines11111723 - 17 Nov 2023
Cited by 1 | Viewed by 873
Abstract
The outbreak of the COVID-19 pandemic at the turn of 2019 and 2020 posed a substantial challenge for the world [...] Full article
(This article belongs to the Special Issue Cellular and Humoral Immunity after COVID-19 Vaccination)

Research

Jump to: Editorial, Review

16 pages, 1101 KiB  
Article
A Serological Analysis of the Humoral Immune Responses of Anti-RBD IgG, Anti-S1 IgG, and Anti-S2 IgG Levels Correlated to Anti-N IgG Positivity and Negativity in Sicilian Healthcare Workers (HCWs) with Third Doses of the mRNA-Based SARS-CoV-2 Vaccine: A Retrospective Cohort Study
by Nicola Serra, Maria Andriolo, Ignazio Butera, Giovanni Mazzola, Consolato Maria Sergi, Teresa Maria Assunta Fasciana, Anna Giammanco, Maria Chiara Gagliano, Antonio Cascio and Paola Di Carlo
Vaccines 2023, 11(7), 1136; https://doi.org/10.3390/vaccines11071136 - 23 Jun 2023
Viewed by 1144
Abstract
Background: With SARS-CoV-2 antibody tests on the market, healthcare providers must be confident that they can use the results to provide actionable information to understand the characteristics and dynamics of the humoral response and antibodies (abs) in SARS-CoV-2-vaccinated patients. In this way, the [...] Read more.
Background: With SARS-CoV-2 antibody tests on the market, healthcare providers must be confident that they can use the results to provide actionable information to understand the characteristics and dynamics of the humoral response and antibodies (abs) in SARS-CoV-2-vaccinated patients. In this way, the study of the antibody responses of healthcare workers (HCWs), a population that is immunocompetent, adherent to vaccination, and continuously exposed to different virus variants, can help us understand immune protection and determine vaccine design goals. Methods: We retrospectively evaluated antibody responses via multiplex assays in a sample of 538 asymptomatic HCWs with a documented complete vaccination cycle of 3 doses of mRNA vaccination and no previous history of infection. Our sample was composed of 49.44% males and 50.56% females, with an age ranging from 21 to 71 years, and a mean age of 46.73 years. All of the HCWs’ sera were collected from April to July 2022 at the Sant’Elia Hospital of Caltanissetta to investigate the immunologic responses against anti-RBD, anti-S1, anti-S2, and anti-N IgG abs. Results: A significant difference in age between HCWs who were positive and negative for anti-N IgG was observed. For anti-S2 IgG, a significant difference between HCWs who were negative and positive compared to anti-N IgG was observed only for positive HCWs, with values including 10 (U/mL)–100 (U/mL); meanwhile, for anti-RBD IgG and anti-S1 IgG levels, there was only a significant difference observed for positive HCWs with diluted titers. For the negative values of anti-N IgG, among the titer dilution levels of anti-RBD, anti-S1, and anti-S2 IgG, the anti-S2 IgG levels were significantly lower than the anti-RBD and anti-S1 levels; in addition, the anti-S1 IgG levels were significantly lower than the anti-RBD IgG levels. For the anti-N IgG positive levels, only the anti-S2 IgG levels were significantly lower than the anti-RBD IgG and anti-S1 IgG levels. Finally, a logistic regression analysis showed that age and anti-S2 IgG were negative and positive predictors of anti-N IgG levels, respectively. The analysis between the vaccine type and mixed mRNA combination showed higher levels of antibodies in mixed vaccinated HCWs. This finding disappeared in the anti-N positive group. Conclusions: Most anti-N positive HCWs showed antibodies against the S2 domain and were young subjects. Therefore, the authors suggest that including the anti-SARS-CoV-2-S2 in antibody profiles can serve as a complementary testing approach to qRT-PCR for the early identification of asymptomatic infections in order to reduce the impact of potential new SARS-CoV-2 variants. Our serological investigation on the type of mRNA vaccine and mixed mRNA vaccines shows that future investigations on the serological responses in vaccinated asymptomatic patients exposed to previous infection or reinfection are warranted for updated vaccine boosters. Full article
(This article belongs to the Special Issue Cellular and Humoral Immunity after COVID-19 Vaccination)
Show Figures

Figure 1

12 pages, 1463 KiB  
Article
Humoral Response after SARS-CoV-2 Vaccination in Prostate Cancer Patients
by Agata Błaszczuk, Dominika Sikora, Jacek Kiś, Ewa Stępień, Bartłomiej Drop and Małgorzata Polz-Dacewicz
Vaccines 2023, 11(4), 770; https://doi.org/10.3390/vaccines11040770 - 30 Mar 2023
Viewed by 1727
Abstract
Cancer is an important public health problem. Prostate cancer is one of the most common cancers among men. In Poland, the incidence of this type of cancer is constantly growing. Considering the appearance of a new coronavirus in December 2019 (SARS-CoV-2) and the [...] Read more.
Cancer is an important public health problem. Prostate cancer is one of the most common cancers among men. In Poland, the incidence of this type of cancer is constantly growing. Considering the appearance of a new coronavirus in December 2019 (SARS-CoV-2) and the fact that oncology patients, including those with prostate cancer, are particularly vulnerable to infection, it is recommended to get vaccinated against COVID-19. In our study, we determined the level and prevalence of antibodies against SARS-CoV-2 IgG in patients with prostate cancer compared to the control group and whether the patients’ ages affected the level of antibodies. PCa patients and controls were divided into two age groups: 50–59 years and 60–70 years. We also analyzed the level of antibodies in patients belonging to the relevant risk groups for prostate cancer (the European Society of Urology risk group classification of prostate cancer). For the study, we used the Microblot-Array COVID-19 IgG test to detect antibodies against the three main SARS-CoV-2 antigens: NCP, RBD, and S2. Our results showed that prostate cancer patients had significantly lower levels of anti-SARS-CoV-2 IgG antibodies compared to controls. In addition, age also affected the decrease in the number of IgG antibodies. The level of antibodies in the intermediate/high-risk group was lower compared to the low-risk group. Full article
(This article belongs to the Special Issue Cellular and Humoral Immunity after COVID-19 Vaccination)
Show Figures

Figure 1

8 pages, 1160 KiB  
Article
Antibody Response after SARS-CoV-2 Infection with the Delta and Omicron Variant
by Agata Błaszczuk, Aleksander Michalski, Dominika Sikora, Maria Malm, Bartłomiej Drop and Małgorzata Polz-Dacewicz
Vaccines 2022, 10(10), 1728; https://doi.org/10.3390/vaccines10101728 - 16 Oct 2022
Cited by 5 | Viewed by 1469
Abstract
The SARS-CoV-2 virus caused a worldwide COVID-19 pandemic. So far, 6,120,834 confirmed cases of COVID-19 with 116,773 deaths have been reported in Poland. According to WHO, a total of 54,662,485 vaccine doses have been administered. New variants emerge that become dominant. The aim [...] Read more.
The SARS-CoV-2 virus caused a worldwide COVID-19 pandemic. So far, 6,120,834 confirmed cases of COVID-19 with 116,773 deaths have been reported in Poland. According to WHO, a total of 54,662,485 vaccine doses have been administered. New variants emerge that become dominant. The aim of this study was a comparison of antibody level after infection caused by Delta and Omicron variants. The study included 203 persons who underwent mild COVID-19 despite two doses of vaccine. The obtained results indicate that a significantly lower titer was observed in patients with the Omicron variant infection. Therefore, these patients may be at risk of reinfection with new strains of the Omicron variant. Due to the possibility of reinfection, booster vaccinations are necessary. Further epidemiological and clinical studies are necessary to develop new prevention strategies. Full article
(This article belongs to the Special Issue Cellular and Humoral Immunity after COVID-19 Vaccination)
Show Figures

Figure 1

10 pages, 915 KiB  
Article
Antibodies to NCP, RBD and S2 SARS-CoV-2 in Vaccinated and Unvaccinated Healthcare Workers
by Agata Błaszczuk, Aleksander Michalski, Maria Malm, Bartłomiej Drop and Małgorzata Polz-Dacewicz
Vaccines 2022, 10(8), 1169; https://doi.org/10.3390/vaccines10081169 - 22 Jul 2022
Cited by 5 | Viewed by 1553
Abstract
In a few months, the SARS-CoV-2 virus caused a worldwide COVID-19 pandemic. In Poland, 6 million cases of the disease and 113,000 deaths from COVID-19 have been reported. Healthcare workers (HCWs) constitute one of the main COVID-19 risk groups. The Microblot-Array COVID-19 IgG [...] Read more.
In a few months, the SARS-CoV-2 virus caused a worldwide COVID-19 pandemic. In Poland, 6 million cases of the disease and 113,000 deaths from COVID-19 have been reported. Healthcare workers (HCWs) constitute one of the main COVID-19 risk groups. The Microblot-Array COVID-19 IgG assay was used to detect antibodies against three major SARS-CoV-2 antigens: nucleocapsid (NCP), RBD, and Spike 2 (S2). The aim of our study was to determine the seroprevalence and titer of anti-SARS-CoV-2 IgG antibodies—NCP, RBD, and S2—as markers of the humoral response in vaccinated and unvaccinated HCWs. The study included 203 persons who were divided into four groups: “COVID-19 Vaccinated”, “COVID-19 Unvaccinated”, “Non-COVID-19 Vaccinated”, and “Non-COVID-19 Unvaccinated”. The obtained results indicate that both seroprevalence and the antibody titer are the highest in the “COVID-19 Vaccinated” group. There is no so-called sterile vaccination, and after 6 months from the second dose of vaccine, most vaccinated people have a fairly high level of antibodies. We suggest that multiple vaccination and continuous testing are necessary. The Microblot-Array assay can distinguish between antibodies acquired after infection and/or vaccination. Full article
(This article belongs to the Special Issue Cellular and Humoral Immunity after COVID-19 Vaccination)
Show Figures

Figure 1

10 pages, 2480 KiB  
Article
Evaluation of Safety and Immunogenicity of BNT162B2 mRNA COVID-19 Vaccine in IBD Pediatric Population with Distinct Immune Suppressive Regimens
by Nicola Cotugno, Enrica Franzese, Giulia Angelino, Donato Amodio, Erminia Francesca Romeo, Francesca Rea, Simona Faraci, Renato Tambucci, Elisa Profeti, Emma Concetta Manno, Veronica Santilli, Gioacchino Andrea Rotulo, Chiara Pighi, Chiara Medri, Elena Morrocchi, Luna Colagrossi, Giuseppe Rubens Pascucci, Diletta Valentini, Alberto Villani, Paolo Rossi, Paola De Angelis and Paolo Palmaadd Show full author list remove Hide full author list
Vaccines 2022, 10(7), 1109; https://doi.org/10.3390/vaccines10071109 - 11 Jul 2022
Cited by 9 | Viewed by 2015
Abstract
Patients affected by Inflammatory Bowel Disease (IBD) present higher risk for infection and suboptimal response upon vaccination. The immunogenicity of SARS-CoV2 vaccination is still largely unknown in adolescents or young adults affected by IBD (pIBD). We investigated the safety and immunogenicity of the [...] Read more.
Patients affected by Inflammatory Bowel Disease (IBD) present higher risk for infection and suboptimal response upon vaccination. The immunogenicity of SARS-CoV2 vaccination is still largely unknown in adolescents or young adults affected by IBD (pIBD). We investigated the safety and immunogenicity of the BNT162B2 mRNA COVID-19 vaccine in 27 pIBD, as compared to 30 healthy controls (HC). Immunogenicity was measured by anti-SARS-CoV2 IgG (anti-S and anti-trim Ab) before vaccination, after 21 days (T21) and 7 days after the second dose (T28). The safety profile was investigated by close monitoring and self-reported adverse events. Vaccination was well tolerated, and short-term adverse events reported were only mild to moderate. Three out of twenty-seven patients showed IBD flare after vaccination, but no causal relationship could be established. Overall, pIBD showed a good humoral response upon vaccination compared to HC; however, pIBD on anti-TNFα treatment showed lower anti-S Ab titers compared to patients receiving other immune-suppressive regimens (p = 0.0413 at first dose and p = 0.0301 at second dose). These data show that pIBD present a good safety and immunogenicity profile following SARS-CoV-2 mRNA vaccination. Additional studies on the impact of specific immune-suppressive regimens, such as anti TNFα, on immunogenicity should be further investigated on larger cohorts. Full article
(This article belongs to the Special Issue Cellular and Humoral Immunity after COVID-19 Vaccination)
Show Figures

Graphical abstract

9 pages, 942 KiB  
Article
Cellular Immunity—The Key to Long-Term Protection in Individuals Recovered from SARS-CoV-2 and after Vaccination
by Dragan Primorac, Petar Brlek, Vid Matišić, Vilim Molnar, Kristijan Vrdoljak, Renata Zadro and Marijo Parčina
Vaccines 2022, 10(3), 442; https://doi.org/10.3390/vaccines10030442 - 14 Mar 2022
Cited by 20 | Viewed by 4112
Abstract
Previous clinical and epidemiological studies have shown that over time antibody titers decrease, and they do not provide long-term mucosa protection against SARS-CoV-2 infection. Additionally, the increase in breakthrough infections that occur more frequently in the vaccinated than in the study participants with [...] Read more.
Previous clinical and epidemiological studies have shown that over time antibody titers decrease, and they do not provide long-term mucosa protection against SARS-CoV-2 infection. Additionally, the increase in breakthrough infections that occur more frequently in the vaccinated than in the study participants with previous SARS-CoV-2 infection has recently become a priority public health concern. We measured the amount of interferon-gamma (Quan-T-Cell ELISA) and the level of antibodies (Anti-SARS-CoV-2 QuantiVac ELISA IgG) in the blood of the same patients simultaneously to compare cellular and humoral immunity. A total of 200 study participants (before Omicron variant appearance) were divided into four groups whose levels of cellular and humoral immunity we compared: study participants previously infected with SARS-CoV-2 (group 1); study participants vaccinated with EMA-approved vaccines (group 2); study participants previously infected with SARS-CoV-2, and vaccination history (group 3); and study participants without a history of SARS-CoV-2 infection or vaccination (group 4). Our results showed that study participants who received one of the EMA-approved vaccines and who recovered from COVID-19 (group 3) had significantly higher levels of cellular immunity and antibody titers in comparison with groups 1 and 2. Additionally, we have noticed that the study participants previously infected with SARS-CoV-2 and the study participants vaccinated with EMA-approved vaccines had a long-lasting cellular immunity. Furthermore, antibody levels showed a negative correlation with time since the last contact with a viral antigen, while cellular immunity within 20 months showed as long-term protection. Moreover, out of 200 study participants, only 1 study participant who recovered from COVID-19 (0.5%) was re-infected, while a total of 6 study participants (3%) were infected with SARS-CoV-2 after receiving the vaccine. This study suggests that cellular immunity—unlike humoral immunity, thanks to memory T cells—represents long-term protection in individuals recovered from SARS-CoV-2 and after vaccination. Full article
(This article belongs to the Special Issue Cellular and Humoral Immunity after COVID-19 Vaccination)
Show Figures

Figure 1

12 pages, 1801 KiB  
Article
Waning Humoral Response after COVID-19 mRNA Vaccination in Maintenance Dialysis Patients and Recovery after a Complementary Third Dose
by Bogdan Biedunkiewicz, Leszek Tylicki, Waldemar Ślizień, Monika Lichodziejewska-Niemierko, Małgorzata Dąbrowska, Alicja Kubanek, Sylwia Rodak, Karolina Polewska, Piotr Tylicki, Marcin Renke and Alicja Dębska-Ślizień
Vaccines 2022, 10(3), 433; https://doi.org/10.3390/vaccines10030433 - 11 Mar 2022
Cited by 11 | Viewed by 1801
Abstract
The aim of this study was to analyze the waning of anti-spike (S) antibodies after mRNA vaccination against COVID-19 in maintenance dialysis patients, and to assess the safety and effectiveness of the complementary third dose. This was a prospective, longitudinal study in which [...] Read more.
The aim of this study was to analyze the waning of anti-spike (S) antibodies after mRNA vaccination against COVID-19 in maintenance dialysis patients, and to assess the safety and effectiveness of the complementary third dose. This was a prospective, longitudinal study in which we analyzed the kinetics of antibodies up to six months after a two-dose vaccination (first protocol) in infection-naïve dialysis patients (IN-Ds), previously infected dialysis patients (PI-Ds) and subjects without chronic kidney disease (the controls), as well as their humoral response to the third dose of the same mRNA vaccine (second protocol). The respective reduction in antibody titer after 3 and 6 months by 82.9% and 93.03% in IN-Ds (n = 109), 73.4% and 93.36% in PI-Ds (n = 32) and 75.5% and 88.8% in the controls (n = 20) was demonstrated. Consequently, a protective antibody titer above 141 BAU/mL was found in only 47.7% and 23.8% of IN-Ds after 3 and 6 months, respectively. After the third vaccine dose, a significant increase in antibody titer was observed in all groups, with increases by a factor of ×51.6 in IN-Ds, ×30.1 in the controls and ×8.4 in PI-Ds. The median antibody titer after the third dose differed significantly between groups, and was the highest in PI-Ds: PI-Ds, 9090 (3300–15,000) BAU/mL; the controls, 6945 (2130–11,800); IN-Ds, 3715 (1470–7325) (p < 0.001). In conclusion, we observed similar degrees of antibody waning in all patients. After 3 months, over half of the infection-naïve dialysis patients had a very low antibody titer, and almost twenty percent of them had no antibodies at all. The humoral response to the third dose was very good, raising their titer of antibodies to a higher level than those in the general population who have received the primary two-dose scheme. The results support the administration of a complementary third dose of the mRNA vaccine for dialysis patients as soon as possible. Full article
(This article belongs to the Special Issue Cellular and Humoral Immunity after COVID-19 Vaccination)
Show Figures

Figure 1

10 pages, 1216 KiB  
Article
Humoral and Cellular Immune Responses of Solid Organ Transplant Patients on Belatacept to Three Doses of mRNA-Based Anti-SARS-CoV-2 Vaccine
by Florence Abravanel, Olivier Marion, Arnaud Del Bello, Thomas Beunon, Raphaelle Romieu-Mourez, Chloé Couat, Mélanie Pucelle, Laetitia Staes, Joelle Guitard, Laure Esposito, Stanislas Faguer, Nassim Kamar and Jacques Izopet
Vaccines 2022, 10(3), 354; https://doi.org/10.3390/vaccines10030354 - 24 Feb 2022
Cited by 8 | Viewed by 1864
Abstract
Background: Two doses of anti-SARS-CoV-2 mRNA-based vaccines are poorly immunogenic in solid organ transplant recipients (SOT). Methods: In total, 68 belatacept-treated SOT recipients followed at the Toulouse University Hospital were investigated. They were given three injections of the BNT162b2 mRNA COVID-19 vaccine. Their [...] Read more.
Background: Two doses of anti-SARS-CoV-2 mRNA-based vaccines are poorly immunogenic in solid organ transplant recipients (SOT). Methods: In total, 68 belatacept-treated SOT recipients followed at the Toulouse University Hospital were investigated. They were given three injections of the BNT162b2 mRNA COVID-19 vaccine. Their humoral response was assessed by determining anti-spike antibodies and neutralizing antibodies. The T-cell responses were assessed using an enzyme-linked immunospot assay that measured the interferon-γ produced by specific SARS-CoV-2 T-cells in a subgroup of 17 patients. Results: Only 23.5% of these patients developed a detectable anti-spike response. Moreover, the cellular and the humoral responses were well correlated. Patients with no humoral response were also without a detectable cellular response. Those belatacept-treated patients who developed an Anti-SARS-CoV-2 humoral response were younger, had been transplanted for longer, and had a higher lymphocyte count and a better glomerular filtration rate than those with no response. Finally, patients on tacrolimus plus belatacept produced a lower immune response. Conclusions: Belatacept-treated SOT recipients have a reduced immune response to anti-SARS-CoV-2 mRNA vaccination. The vaccine should be given quite separately from the belatacept infusion to improve immunogenicity. Studies to assess whether switching to another immunosuppressive regimen can improve the post-vaccination immune response would be useful. Full article
(This article belongs to the Special Issue Cellular and Humoral Immunity after COVID-19 Vaccination)
Show Figures

Figure 1

10 pages, 1176 KiB  
Article
Boosting Humoral Immunity from mRNA COVID-19 Vaccines in Kidney Transplant Recipients
by Leszek Tylicki, Alicja Dębska-Ślizień, Marta Muchlado, Zuzanna Ślizień, Justyna Gołębiewska, Małgorzata Dąbrowska and Bogdan Biedunkiewicz
Vaccines 2022, 10(1), 56; https://doi.org/10.3390/vaccines10010056 - 31 Dec 2021
Cited by 20 | Viewed by 2072
Abstract
Introduction: The immune response to the primary (two-dose) series of mRNA COVID-19 vaccines in kidney transplant recipients (KTRs) is very weak. We conducted a longitudinal observational study to compare the humoral response to a third, additional primary dose of mRNA vaccines between infection-naïve [...] Read more.
Introduction: The immune response to the primary (two-dose) series of mRNA COVID-19 vaccines in kidney transplant recipients (KTRs) is very weak. We conducted a longitudinal observational study to compare the humoral response to a third, additional primary dose of mRNA vaccines between infection-naïve (IN-KTRs) and previously infected KTRs (PI-KTRs). Methods: We measured the levels of anti-spike (anti-s) IgG antibodies before and 14–21 days after the third dose and, in the secondary analysis, we compared the antibody response to BNT162b2 versus mRNA-1273. The reactogenicity assessment included solicited local and systemic reactions. Results: A total of 112 KTRs were enrolled, including 83 IN-KTR and 29 PI-KTR, among whom seroconversion in anti-s antibodies after the primary two-dose vaccination was achieved in 45.78% and 100% of cases, respectively. After three months, a waning antibodies titer by 67.4% (IN-KTR) and 7.5% (PI-KTR) was observed. After the third dose of the mRNA vaccine, 71.08% (59/83) of IN-KTR and 96.5% (28/29) of PI-KTR samples were seroconverted with a median anti-s titer of 468.0 (195.0–1620.0) BAU/mL and 1629.0 (1205–1815) BAU/mL, respectively. Of those IN-KTR in whom the primary vaccination failed, 46.67% (21/45) of patients achieved seroconversion after the third dose. No serious adverse events after the third dose were reported. In strata analyses, after the third dose, 66% (40/60) of patients vaccinated with BNT162b2 and 82.6% (19/23) of patients vaccinated with mRNA-1273 seroconverted with a median anti-s titer of 384.5 (144–837) BAU/mL and 1620 (671–2040) BAU/mL, respectively. Conclusions: The use of a third dose of mRNA vaccine may be of benefit for KTR, especially for those in whom the primary vaccination failed. Vaccines with a higher dose of mRNA and a longer interval between doses of the primary vaccination, such as mRNA-1273, seem to be the preparations of choice in immunocompromised individuals. Full article
(This article belongs to the Special Issue Cellular and Humoral Immunity after COVID-19 Vaccination)
Show Figures

Figure 1

Review

Jump to: Editorial, Research

12 pages, 846 KiB  
Review
The Four Ws of the Fourth Dose COVID-19 Vaccines: Why, Who, When and What
by Ka-Wa Khong, Ruiqi Zhang and Ivan Fan-Ngai Hung
Vaccines 2022, 10(11), 1924; https://doi.org/10.3390/vaccines10111924 - 14 Nov 2022
Cited by 7 | Viewed by 2743
Abstract
With the emergence of SARS-CoV-2 variants, vaccine breakthrough is a major public health concern. With evidence of reduced neutralizing antibody activity against Omicron variants and fading antibody level after the third-dose booster vaccine, there are suggestions of a fourth-dose booster vaccine. In this [...] Read more.
With the emergence of SARS-CoV-2 variants, vaccine breakthrough is a major public health concern. With evidence of reduced neutralizing antibody activity against Omicron variants and fading antibody level after the third-dose booster vaccine, there are suggestions of a fourth-dose booster vaccine. In this review, the benefits of a fourth-dose booster is evaluated from four perspectives, including the effectiveness of the booster dose against virus variants (Why), susceptible groups of individuals who may benefit from additional booster dose (Who), selection of vaccine platforms to better enhance immunity (What) and appropriate intervals between the third and fourth booster dose (When). In summary, a fourth dose can temporarily boost the immune response against SARS-CoV-2 variants and can be considered for specific groups of individuals. A heterologous vaccine strategy using mRNA vaccine in individuals primed with inactivated vaccine may boost immunity against variants. The timing of the fourth dose should be individualized but an interval of 4 months after the third-dose booster is appropriate. A universal fourth booster dose is not necessary. Full article
(This article belongs to the Special Issue Cellular and Humoral Immunity after COVID-19 Vaccination)
Show Figures

Figure 1

28 pages, 675 KiB  
Review
Immunity after COVID-19 Recovery and Vaccination: Similarities and Differences
by Dorota Kamińska, Dominika Dęborska-Materkowska, Katarzyna Kościelska-Kasprzak, Oktawia Mazanowska, Agata Remiorz, Paweł Poznański, Magdalena Durlik and Magdalena Krajewska
Vaccines 2022, 10(7), 1068; https://doi.org/10.3390/vaccines10071068 - 03 Jul 2022
Cited by 8 | Viewed by 2884
Abstract
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is associated with a robust immune response. The development of systemic inflammation leads to a hyperinflammatory state due to cytokine release syndrome during severe COVID-19. The emergence of [...] Read more.
The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is associated with a robust immune response. The development of systemic inflammation leads to a hyperinflammatory state due to cytokine release syndrome during severe COVID-19. The emergence of many new SARS-CoV-2 variants across the world deteriorates the protective antiviral immunity induced after infection or vaccination. The innate immune response to SARS-CoV-2 is crucial for determining the fate of COVID-19 symptomatology. T cell-mediated immunity is the main factor of the antiviral immune response; moreover, SARS-CoV-2 infection initiates a rapid B-cell response. In this paper, we present the current state of knowledge on immunity after COVID-19 infection and vaccination. We discuss the mechanisms of immune response to various types of vaccines (nucleoside-modified, adenovirus-vectored, inactivated virus vaccines and recombinant protein adjuvanted formulations). This includes specific aspects of vaccination in selected patient populations with altered immune activity (the elderly, children, pregnant women, solid organ transplant recipients, patients with systemic rheumatic diseases or malignancies). We also present diagnostic and research tools available to study the anti-SARS-CoV-2 cellular and humoral immune responses. Full article
(This article belongs to the Special Issue Cellular and Humoral Immunity after COVID-19 Vaccination)
Show Figures

Figure 1

Back to TopTop