Humoral and Cellular Response after COVID-19 Vaccination

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 (31 December 2023) | Viewed by 24465

Special Issue Editor


E-Mail Website
Guest Editor
Department of Nephrology and Hypertension, Rabin Medical Center, Sackler School of Medicine, Tel-Aviv University, Petah Tikva, Israel
Interests: SARS-CoV-2 vaccines; T and B cell immune response

Special Issue Information

Dear Colleagues,

COVID-19 vaccines are effective for infection outbreak control. However, waning immunity over time and the emergence of new SARS-CoV-2 variants have caused immune resistance to current vaccines.

This Special Issue focuses on the long-term humoral and cellular responses after COVID-19 vaccination.

We are interested in manuscripts that focus on emerging issues in COVID-19 vaccination immune response as well as factors influencing current vaccines’ effectiveness. Papers identifying novel strategies for improving immune response in immunocompromised patients are also of interest.

Since the long-term efficacy of COVID-19 vaccines is still unknown, we are calling for studies on both humoral and cellular long-term immunity as well as those investigating the immunity effectiveness of additional booster vaccine doses.

The submission of original articles, systematic reviews, and short communications on related topics are welcome.

Dr. Michal Herman-Edelstein
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

  • SARS-CoV-2 vaccines
  • booster dose (second and third doses)
  • T- and B-cell immune response
  • humoral immune response
  • neutralizing antibodies
  • anti-SARS-CoV-2 spike antibodies

Published Papers (10 papers)

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

Research

Jump to: Review, Other

16 pages, 4560 KiB  
Article
SARS-CoV-2-Specific CD8+ T-Cells in Blood but Not in the Lungs of Vaccinated K18-hACE2 Mice after Infection
by Flavia Ferrantelli, Francesco Manfredi, Chiara Chiozzini, Patrizia Leone, Katherina Pugliese, Massimo Spada, Antonio Di Virgilio, Andrea Giovannelli, Mauro Valeri, Andrea Cara, Zuleika Michelini, Mauro Andreotti and Maurizio Federico
Vaccines 2023, 11(9), 1433; https://doi.org/10.3390/vaccines11091433 - 30 Aug 2023
Viewed by 924
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 enters the host by infecting nasal ciliated cells. Then, the virus can spread towards the oropharyngeal cavity and the pulmonary tissues. The antiviral adaptive immunity is promptly induced in response to the virus’s detection, with virus-specific T-lymphocytes [...] Read more.
Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 enters the host by infecting nasal ciliated cells. Then, the virus can spread towards the oropharyngeal cavity and the pulmonary tissues. The antiviral adaptive immunity is promptly induced in response to the virus’s detection, with virus-specific T-lymphocytes appearing before antiviral antibodies. Both the breadth and potency of antiviral CD8+ T-cell immunity have a key role in containing viral spread and disease severity. Current anti-SARS-CoV-2 vaccines do not impede the virus’s replication in the upper respiratory tract, and there is consensus on the fact that the best potency of the antiviral immune response in both blood and the upper respiratory tract can be reached upon infection in vaccinees (i.e., breakthrough infection). However, whether the antiviral CD8+ T-cells developing in response to the breakthrough infection in the upper respiratory tract diffuse to the lungs is also still largely unknown. To fill the gap, we checked the CD8+ T-cell immunity elicited after infection of K18-hACE2 transgenic mice both at 3 weeks and 3 months after anti-spike vaccination. Virus-specific CD8+ T-cell immunity was monitored in both blood and the lungs before and after infection. By investigating the de novo generation of the CD8+ T-cells specific for SARS-CoV-2 viral proteins, we found that both membrane (M) and/or nucleocapsid (N)-specific CD8+ T-cells were induced at comparable levels in the blood of both unvaccinated and vaccinated mice. Conversely, N-specific CD8+ T-cells were readily found in the lungs of the control mice but were either rare or absent in those of vaccinated mice. These results support the idea that the hybrid cell immunity developing after asymptomatic/mild breakthrough infection strengthens the antiviral cell immunity in the lungs only marginally, implying that the direct exposition of viral antigens is required for the induction of an efficient antiviral cell immunity in the lungs. Full article
(This article belongs to the Special Issue Humoral and Cellular Response after COVID-19 Vaccination)
Show Figures

Figure 1

13 pages, 1074 KiB  
Article
SARS-CoV-2 m-RNA Vaccine Response in Immunocompromised Patients: A Monocentric Study Comparing Cancer, People Living with HIV, Hematopoietic Stem Cell Transplant Patients and Lung Transplant Recipients
by Natacha Bordry, Anne-Claire Mamez, Chiara Fedeli, Chloé Cantero, Cyril Jaksic, Pilar Ustero Alonso, Caroline Rayroux, Gregory Berra, Vera Portillo, Maeva Puntel, Sabine Yerly, Sébastien Bugeia, Garance Gutknecht, Mariagrazia Di Marco, Nicolas Mach, Paola Marina Soccal, Yves Chalandon, Alexandra Calmy and Alfredo Addeo
Vaccines 2023, 11(8), 1284; https://doi.org/10.3390/vaccines11081284 - 26 Jul 2023
Cited by 1 | Viewed by 1743
Abstract
Immunocompromised patients (ICPs) have a higher risk of developing severe forms of COVID-19 and experience a higher burden of complications and mortality than the general population. However, recent studies have suggested that the antibody response to SARS-CoV-2 mRNA vaccines could be highly variable [...] Read more.
Immunocompromised patients (ICPs) have a higher risk of developing severe forms of COVID-19 and experience a higher burden of complications and mortality than the general population. However, recent studies have suggested that the antibody response to SARS-CoV-2 mRNA vaccines could be highly variable among different ICPs. Using a collaborative, monocentric, prospective cohort study, we assessed anti-SARS-CoV-2 spike protein antibody titers following two and three doses of mRNA vaccines in four groups of ICPs (cancer [n = 232]: hematopoietic stem cell transplant [HSCT; n = 126] patients; people living with HIV [PLWH; n = 131]; and lung transplant [LT; n = 39] recipients) treated at Geneva University Hospitals; and healthy individuals (n = 49). After primo-vaccination, the highest anti-S antibody geometric mean titer (IU/mL) was observed in healthy individuals (2417 IU/mL [95% CI: 2327–2500]), the PLWH group (2024 IU/mL [95% CI:1854–2209]) and patients with cancer (840 IU/mL [95% CI: 625–1129]), whereas patients in the HSCT and LT groups had weaker antibody responses (198 IU/mL [95% CI: 108–361] and 7.3 IU/mL [95% CI: 2.5–22]). The booster dose conferred a high antibody response after 1 month in both PLWH (2500 IU/mL) and cancer patients (2386 IU/mL [95% CI: 2182–2500]), a moderate response in HSCT patients (521 IU/mL [95% CI: 306–885]) and a poor response in LT recipients (84 IU/mL [95% CI: 18–389]). Contemporary treatment with immunosuppressive drugs used in transplantation or chemotherapy was associated with a poor response to vaccination. Our findings confirmed the heterogeneity of the humoral response after mRNA vaccines among different ICPs and the need for personalized recommendations for each of these different groups. Full article
(This article belongs to the Special Issue Humoral and Cellular Response after COVID-19 Vaccination)
Show Figures

Figure 1

14 pages, 5852 KiB  
Article
Trivalent SARS-CoV-2 S1 Subunit Protein Vaccination Induces Broad Humoral Responses in BALB/c Mice
by Muhammad S. Khan, Eun Kim, Shaohua Huang, Thomas W. Kenniston and Andrea Gambotto
Vaccines 2023, 11(2), 314; https://doi.org/10.3390/vaccines11020314 - 31 Jan 2023
Cited by 5 | Viewed by 3599
Abstract
This paper presents a novel approach for improving the efficacy of COVID-19 vaccines against emergent SARS-CoV-2 variants. We have evaluated the immunogenicity of unadjuvanted wild-type (WU S1-RS09cg) and variant-specific (Delta S1-RS09cg and OM S1-RS09cg) S1 subunit protein vaccines delivered either as a monovalent [...] Read more.
This paper presents a novel approach for improving the efficacy of COVID-19 vaccines against emergent SARS-CoV-2 variants. We have evaluated the immunogenicity of unadjuvanted wild-type (WU S1-RS09cg) and variant-specific (Delta S1-RS09cg and OM S1-RS09cg) S1 subunit protein vaccines delivered either as a monovalent or a trivalent antigen in BALB/c mice. Our results show that a trivalent approach induced a broader humoral response with more coverage against antigenically distinct variants, especially when compared to monovalent Omicron-specific S1. This trivalent approach was also found to have increased or equivalent ACE2 binding inhibition, and increased S1 IgG endpoint titer at early timepoints, against SARS-CoV-2 spike variants when compared monovalent Wuhan, Delta, or Omicron S1. Our results demonstrate the utility of protein subunit vaccines against COVID-19 and provide insights into the impact of variant-specific COVID-19 vaccine approaches on the immune response in the current SARS-CoV-2 variant landscape. Particularly, our study provides insight into effects of further increasing valency of currently approved SARS-CoV-2 vaccines, a promising approach for improving protection to curtail emerging viral variants. Full article
(This article belongs to the Special Issue Humoral and Cellular Response after COVID-19 Vaccination)
Show Figures

Figure 1

18 pages, 4070 KiB  
Article
Effector-Memory B-Lymphocytes and Follicular Helper T-Lymphocytes as Central Players in the Immune Response in Vaccinated and Nonvaccinated Populations against SARS-CoV-2
by Lorenzo Islas-Vazquez, Marisa Cruz-Aguilar, Henry Velazquez-Soto, Aida Jiménez-Corona, Sonia Mayra Pérez-Tapia and Maria C. Jimenez-Martinez
Vaccines 2022, 10(10), 1761; https://doi.org/10.3390/vaccines10101761 - 20 Oct 2022
Cited by 2 | Viewed by 2754
Abstract
Vaccines have been recognized as having a central role in controlling the COVID-19 pandemic; however, most vaccine development research is focused on IgG-induced antibodies. Here, we analyzed the generation of IgGs related to SARS-CoV-2 and the changes in B- and T-lymphocyte proportions following [...] Read more.
Vaccines have been recognized as having a central role in controlling the COVID-19 pandemic; however, most vaccine development research is focused on IgG-induced antibodies. Here, we analyzed the generation of IgGs related to SARS-CoV-2 and the changes in B- and T-lymphocyte proportions following vaccination against COVID-19. We included samples from 69 volunteers inoculated with the Pfizer-BioNTech (BNT162b2), Astra Zeneca (AZD1222 Covishield), or Sputnik V (Gam-COVID-Vac) vaccines. IgGs related to SARS-CoV-2 increased after the first vaccine dose compared with the nonvaccinated group (Pfizer, p = 0.0001; Astra Zeneca, p < 0.0001; Sputnik V, p = 0.0089). The results of the flow cytometry analysis of B- and T-lymphocytes showed a higher proportion of effector-memory B-lymphocytes in both first and second doses when compared with the nonvaccinated subjects. FcRL4+ cells were increased in second-dose-vaccinated COVID-19(−) and recovered COVID-19(+) participants when compared with the nonvaccinated participants. COVID-19(−) participants showed a lower proportion of follicular helper T-lymphocytes (TFH) in the second dose when compared with the first-vaccine-dose and nonvaccinated subjects. In conclusion, after the first vaccine dose, immunization against SARS-CoV-2 induces IgG production, and this could be mediated by TFH and effector-memory B-lymphocytes. Our data can be used in the design of vaccine schedules to evaluate immuno-bridging from a cellular point of view. Full article
(This article belongs to the Special Issue Humoral and Cellular Response after COVID-19 Vaccination)
Show Figures

Figure 1

13 pages, 1992 KiB  
Article
Evaluation of Immunogenicity to Three Doses of the SARS-CoV-2 BNT162b2 mRNA Vaccine in Lung Transplant Patients
by Mariasilvia Guardiani, Maria Antonella Zingaropoli, Francesco Cogliati Dezza, Anastasia Centofanti, Carolina Carillo, Eeva Tortellini, Federica Dominelli, Anna Napoli, Cosmo Del Borgo, Aurelia Gaeta, Federico Venuta, Vincenzo Vullo, Miriam Lichtner, Maria Rosa Ciardi, Claudio Maria Mastroianni and Gianluca Russo
Vaccines 2022, 10(10), 1642; https://doi.org/10.3390/vaccines10101642 - 30 Sep 2022
Cited by 4 | Viewed by 1375
Abstract
The aim of the study was to explore the humoral and T-cell response in lung transplant (LuT) patients. Two-time points were considered, before (T0) and after (Tpost) the third dose of the BNT162b2 mRNA vaccine, comparing LuT with healthy donors (HD). LuT patients [...] Read more.
The aim of the study was to explore the humoral and T-cell response in lung transplant (LuT) patients. Two-time points were considered, before (T0) and after (Tpost) the third dose of the BNT162b2 mRNA vaccine, comparing LuT with healthy donors (HD). LuT patients showed a lower serologic response against SARS-CoV-2 compared with HD at both time-points (p = 0.0001 and p = 0.0011, respectively). A lower percentage of IFNγ+orIL2+orTNFα+CD4+ and CD8+ T-cells LuT patients was observed in LuT patients compared with HD at T0 (CD4+: p = 0.0001; CD8+: p = 0.0005) and Tpost (CD4+: p = 0.0028; CD8+: p = 0.0114), as well as in the percentage of IFNγ+IL2+TNFα+CD4+ T-cells (T0: p = 0.0247; Tpost: p = 0.0367). Finally, at Tpost, a lower percentage of IFNγ+IL2+TNFα+ CD8+ T-cells in LuT patients compared with HD was found (p = 0.0147). LuT patients were stratified according to the lowest cut-off value for the detection of a humoral response (4.81 BAU/mL) at T0, into responder (R) and non-responder (NR) groups. In the R group, no differences in the percentage of IFNγ+or IL2+orTNFα+ and IFNγ+IL2+TNFα+CD4+ and CD8+ T-cells compared with HD at both time-points were observed. Otherwise, in the NR group, lower percentages of IFNγ+IL2+TNFα+CD4+ T-cells compared with the R group (T0: p = 0.0159; Tpost: p = 0.0159), as well as compared with the HD, at both time-points, were observed (T0: p = 0.0064; Tpost: p = 0.0064). These data seem to confirm that some LuT patients can mount cellular responses even in the absence of a positive humoral response (>33.8 BAU/mL), although this cellular response is dysfunctional and partially detrimental. Full article
(This article belongs to the Special Issue Humoral and Cellular Response after COVID-19 Vaccination)
Show Figures

Figure 1

9 pages, 2186 KiB  
Article
Validation of a Novel Fluorescent Lateral Flow Assay for Rapid Qualitative and Quantitative Assessment of Total Anti-SARS-CoV-2 S-RBD Binding Antibody Units (BAU) from Plasma or Fingerstick Whole-Blood of COVID-19 Vaccinees
by Nadin Younes, Duaa W. Al-Sadeq, Farah M. Shurrab, Hadeel T. Zedan, Haissam Abou-Saleh, Bushra Y. Abo-Halawa, Fatima M. AlHamaydeh, Amira E. Elsharafi, Hanin I. Daas, Swapna Thomas, Sahar Aboalmaaly, Afra Al Farsi, Reeham Al-Buainain, Samar Ataelmannan, Jiji Paul, Amana Salih Al Saadi, Hadi M. Yassine, Amin F. Majdalawieh, Ahmed Ismail, Laith J. Abu-Raddad and Gheyath K. Nasrallahadd Show full author list remove Hide full author list
Vaccines 2022, 10(8), 1318; https://doi.org/10.3390/vaccines10081318 - 15 Aug 2022
Cited by 3 | Viewed by 2219
Abstract
Background: Limited commercial LFA assays are available to provide a reliable quantitative measurement of the total binding antibody units (BAU/mL) against the receptor-binding domain of the SARS-CoV-2 spike protein (S-RBD). Aim: This study aimed to evaluate the performance of the fluorescence LFA FinecareTM [...] Read more.
Background: Limited commercial LFA assays are available to provide a reliable quantitative measurement of the total binding antibody units (BAU/mL) against the receptor-binding domain of the SARS-CoV-2 spike protein (S-RBD). Aim: This study aimed to evaluate the performance of the fluorescence LFA FinecareTM 2019-nCoV S-RBD test along with its reader (Model No.: FS-113) against the following reference methods: (i) the FDA-approved GenScript surrogate virus-neutralizing assay (sVNT); and (ii) three highly performing automated immunoassays: BioMérieux VIDAS®3, Ortho VITROS®, and Mindray CL-900i®. Methods: Plasma from 488 vaccinees was tested by all aforementioned assays. Fingerstick whole-blood samples from 156 vaccinees were also tested by FinecareTM. Results and conclusions: FinecareTM showed 100% specificity, as none of the pre-pandemic samples tested positive. Equivalent FinecareTM results were observed among the samples taken from fingerstick or plasma (Pearson correlation r = 0.9, p < 0.0001), suggesting that fingerstick samples are sufficient to quantitate the S-RBD BAU/mL. A moderate correlation was observed between FinecareTM and sVNT (r = 0.5, p < 0.0001), indicating that FinecareTM can be used for rapid prediction of the neutralizing antibody (nAb) post-vaccination. FinecareTM BAU results showed strong correlation with VIDAS®3 (r = 0.6, p < 0.0001) and moderate correlation with VITROS® (r = 0.5, p < 0.0001) and CL-900i® (r = 0.4, p < 0.0001), suggesting that FinecareTM can be used as a surrogate for the advanced automated assays to measure S-RBD BAU/mL. Full article
(This article belongs to the Special Issue Humoral and Cellular Response after COVID-19 Vaccination)
Show Figures

Figure 1

20 pages, 5489 KiB  
Article
Anti-TNFα Treatment Impairs Long-Term Immune Responses to COVID-19 mRNA Vaccine in Patients with Inflammatory Bowel Diseases
by Keren Masha Rabinowitz, Michal Navon, Hadar Edelman-Klapper, Eran Zittan, Ariella Bar-Gil Shitrit, Idan Goren, Irit Avni-Biron, Jacob E. Ollech, Lev Lichtenstein, Hagar Banai-Eran, Henit Yanai, Yifat Snir, Maor H. Pauker, Adi Friedenberg, Adva Levy-Barda, Arie Segal, Yelena Broitman, Eran Maoz, Baruch Ovadia, Maya Aharoni Golan, Eyal Shachar, Shomron Ben-Horin, Nitsan Maharshak, Michal Mor, Haim Ben Zvi, Rami Eliakim, Revital Barkan, Tali Sharar-Fischler, Sophy Goren, Noy Krugliak, Edward Pichinuk, Michael Mor, Michal Werbner, Joel Alter, Hanan Abu-Taha, Kawsar Kaboub, Moshe Dessau, Meital Gal-Tanamy, Dani Cohen, Natalia T. Freund, Iris Dotan and on behalf of the Responses to COVID-19 Vaccine Israeli IBD Groupadd Show full author list remove Hide full author list
Vaccines 2022, 10(8), 1186; https://doi.org/10.3390/vaccines10081186 - 26 Jul 2022
Cited by 9 | Viewed by 2721
Abstract
Patients with inflammatory bowel disease (IBD) treated with anti-tumor-necrosis factor-alpha (TNFα) exhibited lower serologic responses one-month following the second dose of the COVID-19 BNT162b2 vaccine compared to those not treated with anti-TNFα (non-anti-TNFα) or to healthy controls (HCs). We comprehensively analyzed long-term humoral [...] Read more.
Patients with inflammatory bowel disease (IBD) treated with anti-tumor-necrosis factor-alpha (TNFα) exhibited lower serologic responses one-month following the second dose of the COVID-19 BNT162b2 vaccine compared to those not treated with anti-TNFα (non-anti-TNFα) or to healthy controls (HCs). We comprehensively analyzed long-term humoral responses, including anti-spike (S) antibodies, serum inhibition, neutralization, cross-reactivity and circulating B cell six months post BNT162b2, in patients with IBD stratified by therapy compared to HCs. Subjects enrolled in a prospective, controlled, multi-center Israeli study received two BNT162b2 doses. Anti-S levels, functional activity, specific B cells, antigen cross-reactivity, anti-nucleocapsid levels, adverse events and IBD disease score were detected longitudinally. In total, 240 subjects, 151 with IBD (94 not treated with anti-TNFα and 57 treated with anti-TNFα) and 89 HCs participated. Six months after vaccination, patients with IBD treated with anti-TNFα had significantly impaired BNT162b2 responses, specifically, more seronegativity, decreased specific circulating B cells and cross-reactivity compared to patients untreated with anti-TNFα. Importantly, all seronegative subjects were patients with IBD; of those, >90% were treated with anti-TNFα. Finally, IBD activity was unaffected by BNT162b2. Altogether these data support the earlier booster dose administration in these patients. Full article
(This article belongs to the Special Issue Humoral and Cellular Response after COVID-19 Vaccination)
Show Figures

Figure 1

Review

Jump to: Research, Other

28 pages, 1769 KiB  
Review
Updated Insights into the T Cell-Mediated Immune Response against SARS-CoV-2: A Step towards Efficient and Reliable Vaccines
by Manish Dhawan, Ali A. Rabaan, Mahmoud M. Al Fawarah, Souad A. Almuthree, Roua A. Alsubki, Amal H. Alfaraj, Mutaib M. Mashraqi, Saleh A. Alshamrani, Wesam A. Abduljabbar, Ameen S. S. Alwashmi, Fatimah Al Ibrahim, Abdulmonem A. Alsaleh, Faryal Khamis, Jameela Alsalman, Manish Sharma and Talha Bin Emran
Vaccines 2023, 11(1), 101; https://doi.org/10.3390/vaccines11010101 - 01 Jan 2023
Cited by 14 | Viewed by 5992
Abstract
The emergence of novel variants of SARS-CoV-2 and their abilities to evade the immune response elicited through presently available vaccination makes it essential to recognize the mechanisms through which SARS-CoV-2 interacts with the human immune response. It is essential not only to comprehend [...] Read more.
The emergence of novel variants of SARS-CoV-2 and their abilities to evade the immune response elicited through presently available vaccination makes it essential to recognize the mechanisms through which SARS-CoV-2 interacts with the human immune response. It is essential not only to comprehend the infection mechanism of SARS-CoV-2 but also for the generation of effective and reliable vaccines against COVID-19. The effectiveness of the vaccine is supported by the adaptive immune response, which mainly consists of B and T cells, which play a critical role in deciding the prognosis of the COVID-19 disease. T cells are essential for reducing the viral load and containing the infection. A plethora of viral proteins can be recognized by T cells and provide a broad range of protection, especially amid the emergence of novel variants of SARS-CoV-2. However, the hyperactivation of the effector T cells and reduced number of lymphocytes have been found to be the key characteristics of the severe disease. Notably, excessive T cell activation may cause acute respiratory distress syndrome (ARDS) by producing unwarranted and excessive amounts of cytokines and chemokines. Nevertheless, it is still unknown how T-cell-mediated immune responses function in determining the prognosis of SARS-CoV-2 infection. Additionally, it is unknown how the functional perturbations in the T cells lead to the severe form of the disease and to reduced protection not only against SARS-CoV-2 but many other viral infections. Hence, an updated review has been developed to understand the involvement of T cells in the infection mechanism, which in turn determines the prognosis of the disease. Importantly, we have also focused on the T cells’ exhaustion under certain conditions and how these functional perturbations can be modulated for an effective immune response against SARS-CoV-2. Additionally, a range of therapeutic strategies has been discussed that can elevate the T cell-mediated immune response either directly or indirectly. Full article
(This article belongs to the Special Issue Humoral and Cellular Response after COVID-19 Vaccination)
Show Figures

Figure 1

Other

Jump to: Research, Review

10 pages, 1182 KiB  
Case Report
Lack of Specific Immune Response after Five Doses of mRNA SARS-CoV-2 Vaccine in a Patient with CD4+ T-Cell Lymphopenia but Preserved Responses to CMV
by Trinidad Alba-Cano, Eduardo Fernández-Cruz, Roberto Alonso, Sara Muñoz-Gómez, Rebeca Pérez de Diego, Elena García Martínez, Paloma Sánchez-Mateos, Joaquín Navarro Caspistegui, Mónica Martín López and Juana Gil-Herrera
Vaccines 2024, 12(4), 386; https://doi.org/10.3390/vaccines12040386 - 06 Apr 2024
Viewed by 604
Abstract
Immunogenicity of SARS-CoV-2 mRNA vaccines is highly heterogeneous in patients with inborn errors of immunity (IEIs). This case report analyzes the immune response to mRNA COVID-19 two-dose primary vaccination followed by three boosters in an IEI patient with marked CD4+ T-cell cytopenia [...] Read more.
Immunogenicity of SARS-CoV-2 mRNA vaccines is highly heterogeneous in patients with inborn errors of immunity (IEIs). This case report analyzes the immune response to mRNA COVID-19 two-dose primary vaccination followed by three boosters in an IEI patient with marked CD4+ T-cell cytopenia and diminished thymic output, in comparison with that raised against latent, chronic cytomegalovirus (CMV) infection. Serum IgG antibodies anti-spike (S) protein of SARS-CoV-2 and anti-CMV were both determined by chemiluminescent microparticle immunoassays (CMIAs). SARS-CoV-2 and CMV memory CD4+ T-cell responses were simultaneously evaluated in vitro using an activation-induced marker (AIM) assay via multicolor flow cytometry. Throughout the 2-year follow-up that included the administration of five doses of SARS-CoV-2 mRNA vaccines, cellular anti-SARS-CoV-2-specific responses remained consistently negative, with extremely weak humoral responses, while the patient showed in vitro persistent CD4+ T-cell reactivity to CMV peptides and high-IgG CMV-specific titers. The assessment of immune responses to vaccines and prevalent viruses is essential in IEI patients in order to take adequate preventive measures. Full article
(This article belongs to the Special Issue Humoral and Cellular Response after COVID-19 Vaccination)
Show Figures

Figure 1

8 pages, 1339 KiB  
Brief Report
Prior COVID-19 Immunization Does Not Cause IgA- or IgG-Dependent Enhancement of SARS-CoV-2 Infection
by Melyssa Yaugel-Novoa, Blandine Noailly, Fabienne Jospin, Anne-Emmanuelle Berger, Louis Waeckel, Elisabeth Botelho-Nevers, Stéphanie Longet, Thomas Bourlet and Stéphane Paul
Vaccines 2023, 11(4), 773; https://doi.org/10.3390/vaccines11040773 - 31 Mar 2023
Viewed by 1699
Abstract
Antibody-dependent enhancement (ADE) can increase the rates and severity of infection with various viruses, including coronaviruses, such as MERS. Some in vitro studies on COVID-19 have suggested that prior immunization enhances SARS-CoV-2 infection, but preclinical and clinical studies have demonstrated the contrary. We [...] Read more.
Antibody-dependent enhancement (ADE) can increase the rates and severity of infection with various viruses, including coronaviruses, such as MERS. Some in vitro studies on COVID-19 have suggested that prior immunization enhances SARS-CoV-2 infection, but preclinical and clinical studies have demonstrated the contrary. We studied a cohort of COVID-19 patients and a cohort of vaccinated individuals with a heterologous (Moderna/Pfizer) or homologous (Pfizer/Pfizer) vaccination scheme. The dependence on IgG or IgA of ADE of infection was evaluated on the serum samples from these subjects (twenty-six vaccinated individuals and twenty-one PCR-positive SARS-CoV-2-infected patients) using an in vitro model with CD16- or CD89-expressing cells and the Delta (B.1.617.2 lineage) and Omicron (B.1.1.529 lineage) variants of SARS-CoV-2. Sera from COVID-19 patients did not show ADE of infection with any of the tested viral variants. Some serum samples from vaccinated individuals displayed a mild IgA-ADE effect with Omicron after the second dose of the vaccine, but this effect was abolished after the completion of the full vaccination scheme. In this study, FcγRIIIa- and FcαRI-dependent ADE of SARS-CoV-2 infection after prior immunization, which might increase the risk of severe disease in a second natural infection, was not observed. Full article
(This article belongs to the Special Issue Humoral and Cellular Response after COVID-19 Vaccination)
Show Figures

Figure 1

Back to TopTop