1
|
Alirezaylavasani A, Skeie LG, Egner IM, Chopra A, Dahl TB, Prebensen C, Vaage JT, Halvorsen B, Lund-Johansen F, Tonby K, Reikvam DH, Stiksrud B, Holter JC, Dyrhol-Riise AM, Munthe LA, Kared H. Vaccine responses and hybrid immunity in people living with HIV after SARS-CoV-2 breakthrough infections. NPJ Vaccines 2024; 9:185. [PMID: 39384763 PMCID: PMC11464709 DOI: 10.1038/s41541-024-00972-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 09/19/2024] [Indexed: 10/11/2024] Open
Abstract
The COVID-19 pandemic posed a challenge for people living with HIV (PLWH), particularly immune non-responders (INR) with compromised CD4 T-cell reconstitution following antiretroviral therapy (CD4 count <350 cells per mm3). Their diminished vaccine responses raised concerns about their vulnerability to SARS-CoV-2 breakthrough infections (BTI). Our in-depth study here revealed chronic inflammation in PLWH and a limited anti-Spike IgG response after vaccination in INR. Nevertheless, the imprinting of Spike-specific B cells by vaccination significantly enhanced the humoral responses after BTI. Notably, the magnitude of cellular CD4 response in all PLWH was comparable to that in healthy donors (HD). However, the polyfunctionality and phenotype of Spike-specific CD8 T cells in INR differed from controls. The findings highlight the need for additional boosters with variant vaccines, and for monitoring ART adherence and the durability of both humoral and cellular anti-SARS-CoV-2 immunity in INR.
Collapse
Affiliation(s)
- Amin Alirezaylavasani
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Linda Gail Skeie
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Ingrid Marie Egner
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Adity Chopra
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Tuva Børresdatter Dahl
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Christian Prebensen
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - John Torgils Vaage
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Kristian Tonby
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Dag Henrik Reikvam
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Birgitte Stiksrud
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Jan Cato Holter
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Anne Ma Dyrhol-Riise
- Department of Infectious Diseases, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ludvig A Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| | - Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway.
- KG Jebsen Centre for B Cell Malignancies, University of Oslo, Oslo, Norway.
- Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| |
Collapse
|
2
|
Wietschel KA, Fechtner K, Antileo E, Abdurrahman G, Drechsler CA, Makuvise MK, Rose R, Voß M, Krumbholz A, Michalik S, Weiss S, Ulm L, Franikowski P, Fickenscher H, Bröker BM, Raafat D, Holtfreter S. Non-cross-reactive epitopes dominate the humoral immune response to COVID-19 vaccination - kinetics of plasma antibodies, plasmablasts and memory B cells. Front Immunol 2024; 15:1382911. [PMID: 38807606 PMCID: PMC11130424 DOI: 10.3389/fimmu.2024.1382911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/15/2024] [Indexed: 05/30/2024] Open
Abstract
Introduction COVID-19 vaccines are highly effective in inducing protective immunity. While the serum antibody response to COVID-19 vaccination has been studied in depth, our knowledge of the underlying plasmablast and memory B cell (Bmem) responses is still incomplete. Here, we determined the antibody and B cell response to COVID-19 vaccination in a naïve population and contrasted it with the response to a single influenza vaccination in a primed cohort. In addition, we analyzed the antibody and B cell responses against the four endemic human coronaviruses (HCoVs). Methods Measurement of specific plasma IgG antibodies was combined with functional analyses of antibody-secreting plasmablasts and Bmems. SARS-CoV-2- and HCoV-specific IgG antibodies were quantified with an in-house bead-based multiplexed immunoassay. Results The antibody and B cell responses to COVID-19 vaccination reflected the kinetics of a prime-boost immunization, characterized by a slow and moderate primary response and a faster and stronger secondary response. In contrast, the influenza vaccinees possessed robust immune memory for the vaccine antigens prior to vaccination, and the recall vaccination moderately boosted antibody production and Bmem responses. Antibody levels and Bmem responses waned several months after the 2nd COVID-19 vaccination, but were restored upon the 3rd vaccination. The COVID-19 vaccine-induced antibodies mainly targeted novel, non-cross-reactive S1 epitopes of the viral spike protein, while cross-reactive S2 epitopes were less immunogenic. Booster vaccination not only strongly enhanced neutralizing antibodies against an original SARS-CoV-2 strain, but also induced neutralizing antibodies against the Omicron BA.2 variant. We observed a 100% plasma antibody prevalence against the S1 subunits of HCoVs, which was not affected by vaccination. Discussion Overall, by complementing classical serology with a functional evaluation of plasmablasts and memory B cells we provide new insights into the specificity of COVID-19 vaccine-induced antibody and B cell responses.
Collapse
Affiliation(s)
- Kilian A. Wietschel
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Kevin Fechtner
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Elmer Antileo
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Goran Abdurrahman
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Chiara A. Drechsler
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | | | - Ruben Rose
- Institute for Infection Medicine, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Mathias Voß
- Institute for Infection Medicine, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Andi Krumbholz
- Institute for Infection Medicine, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
- Labor Dr. Krause und Kollegen MVZ GmbH, Kiel, Germany
| | - Stephan Michalik
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, University Medicine Greifswald, Greifswald, Germany
| | - Lena Ulm
- Friedrich Loeffler-Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Philipp Franikowski
- Institute for Educational Quality Improvement, Humboldt University of Berlin, Berlin, Germany
| | - Helmut Fickenscher
- Institute for Infection Medicine, Kiel University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Barbara M. Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Dina Raafat
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Silva Holtfreter
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
3
|
Grigoryan L, Feng Y, Bellusci L, Lai L, Wali B, Ellis M, Yuan M, Arunachalam PS, Hu M, Kowli S, Gupta S, Maysel-Auslender S, Maecker HT, Samaha H, Rouphael N, Wilson IA, Moreno AC, Suthar MS, Khurana S, Pillet S, Charland N, Ward BJ, Pulendran B. AS03 adjuvant enhances the magnitude, persistence, and clonal breadth of memory B cell responses to a plant-based COVID-19 vaccine in humans. Sci Immunol 2024; 9:eadi8039. [PMID: 38579013 DOI: 10.1126/sciimmunol.adi8039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 03/11/2024] [Indexed: 04/07/2024]
Abstract
Vaccine adjuvants increase the breadth of serum antibody responses, but whether this is due to the generation of antigen-specific B cell clones with distinct specificities or the maturation of memory B cell clones that produce broadly cross-reactive antibodies is unknown. Here, we longitudinally analyzed immune responses in healthy adults after two-dose vaccination with either a virus-like particle COVID-19 vaccine (CoVLP), CoVLP adjuvanted with AS03 (CoVLP+AS03), or a messenger RNA vaccination (mRNA-1273). CoVLP+AS03 enhanced the magnitude and durability of circulating antibodies and antigen-specific CD4+ T cell and memory B cell responses. Antigen-specific CD4+ T cells in the CoVLP+AS03 group at day 42 correlated with antigen-specific memory B cells at 6 months. CoVLP+AS03 induced memory B cell responses, which accumulated somatic hypermutations over 6 months, resulting in enhanced neutralization breadth of monoclonal antibodies. Furthermore, the fraction of broadly neutralizing antibodies encoded by memory B cells increased between day 42 and 6 months. These results indicate that AS03 enhances the antigenic breadth of B cell memory at the clonal level and induces progressive maturation of the B cell response.
Collapse
Affiliation(s)
- Lilit Grigoryan
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Yupeng Feng
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | | | - Lilin Lai
- Department of Pediatrics and Department of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory School of Medicine, Atlanta, GA 30329, USA
| | - Bushra Wali
- Department of Pediatrics and Department of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory School of Medicine, Atlanta, GA 30329, USA
| | - Madison Ellis
- Department of Pediatrics and Department of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory School of Medicine, Atlanta, GA 30329, USA
| | - Meng Yuan
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mengyun Hu
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sangeeta Kowli
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sheena Gupta
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Sofia Maysel-Auslender
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Holden T Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Hady Samaha
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nadine Rouphael
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
- Hope Clinic of Emory Vaccine Center, Emory University, Decatur, GA 30030, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, Scripps Research Institute, La Jolla, CA 92037, USA
| | - Alberto C Moreno
- Department of Medicine, Emory Vaccine Center, Emory National Primate Research Center, Emory University School of Medicine, Atlanta, GA 30329, USA
| | - Mehul S Suthar
- Department of Pediatrics and Department of Microbiology and Immunology, Emory Vaccine Center, Emory National Primate Research Center, Emory School of Medicine, Atlanta, GA 30329, USA
| | | | - Stéphane Pillet
- Medicago Inc., Québec, QC G1V 3V9, Canada
- Research Institute of the McGill University Health Center, 1001 Decarie St., Montréal, QC H4A 3J1, Canada
| | | | - Brian J Ward
- Medicago Inc., Québec, QC G1V 3V9, Canada
- Research Institute of the McGill University Health Center, 1001 Decarie St., Montréal, QC H4A 3J1, Canada
| | - Bali Pulendran
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
4
|
Sheng WH, Lin PH, Cheng YC, Wu YY, Hsieh MJ, Yang HC, Chang SY, Chang SC. Immunogenicity and safety of heterologous booster with protein-based COVID-19 vaccine (NVX-CoV2373) in healthy adults: A comparative analysis with mRNA vaccines. J Formos Med Assoc 2024; 123:340-346. [PMID: 37996322 DOI: 10.1016/j.jfma.2023.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/07/2023] [Accepted: 10/12/2023] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Information on the protein-based severe acute respiratory syndrome (SARS-CoV-2) vaccine-NVX-CoV2373 (Novavax), as a heterologous booster remains limited. We investigated the immunogenicity and adverse events of NVX-CoV2373 as a second booster and compared them with those of mRNA vaccines in healthy adults. METHODS Healthcare workers who had received an mRNA vaccine (mRNA-1273 or BNT-162b2) as the first booster (third dose) 12 weeks prior were recruited. Participants voluntarily received either NVX-CoV2373 or an mRNA vaccine as a second booster. Participants with a history of SARS-CoV-2 infection were excluded. The primary outcomes included serum anti-SARS-CoV-2 spike protein (SP) and neutralizing antibody titers against B.1.1.7 (Alpha), B.1.1.529 (Omicron) BA2, and BA5 variants on the 28th day after the boost. Secondary outcomes included new SARS-CoV-2 infections and adverse events reported during the study period. RESULTS A total of 160 participants were enrolled in this study. Compared with the mRNA vaccination group (n = 59), the NVX-CoV2373 vaccination group (n = 101) had significantly lower anti-SARS-CoV-2 SP antibody titers and neutralizing antibody titers against all variants tested after the boost. During the study period, higher rates of new SARS-CoV-2 infections and a lower incidence of adverse events were observed in the NVX-CoV2373 vaccination group. No significant differences in cellular immune responses were observed between the two groups. CONCLUSION Compared to a homologous mRNA booster vaccination, heterologous boosters with NVX-CoV2373 showed lower antibody responses, a higher incidence of new SARS-CoV-2 infections, and fewer adverse events.
Collapse
Affiliation(s)
- Wang-Huei Sheng
- Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan; School of Medicine, National Taiwan University College of Medicine, Taipei City, Taiwan
| | - Pin-Hung Lin
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Yu-Chen Cheng
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Yun Wu
- Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ming-Ju Hsieh
- Department of Emergency Medicine, National Taiwan University Hospital, Taipei, Taiwan; Occupational Safety and Health Office, National Taiwan University Hospital, Taipei, Taiwan
| | - Hung-Chih Yang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan; Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Sui-Yuan Chang
- Department of Clinical Laboratory Sciences and Medical Biotechnology, National Taiwan University College of Medicine, Taipei, Taiwan; Department of Laboratory Medicine, National Taiwan University Hospital, Taipei City, Taiwan
| | - Shan-Chwen Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei City, Taiwan; School of Medicine, National Taiwan University College of Medicine, Taipei City, Taiwan.
| |
Collapse
|
5
|
Röltgen K, Boyd SD. Antibody and B Cell Responses to SARS-CoV-2 Infection and Vaccination: The End of the Beginning. ANNUAL REVIEW OF PATHOLOGY 2024; 19:69-97. [PMID: 37738512 DOI: 10.1146/annurev-pathmechdis-031521-042754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/24/2023]
Abstract
As the COVID-19 pandemic has evolved during the past years, interactions between human immune systems, rapidly mutating and selected SARS-CoV-2 viral variants, and effective vaccines have complicated the landscape of individual immunological histories. Here, we review some key findings for antibody and B cell-mediated immunity, including responses to the highly mutated omicron variants; immunological imprinting and other impacts of successive viral antigenic variant exposures on antibody and B cell memory; responses in secondary lymphoid and mucosal tissues and non-neutralizing antibody-mediated immunity; responses in populations vulnerable to severe disease such as those with cancer, immunodeficiencies, and other comorbidities, as well as populations showing apparent resistance to severe disease such as many African populations; and evidence of antibody involvement in postacute sequelae of infection or long COVID. Despite the initial phase of the pandemic ending, human populations will continue to face challenges presented by this unpredictable virus.
Collapse
Affiliation(s)
- Katharina Röltgen
- Department of Medical Parasitology and Infection Biology, Swiss Tropical and Public Health Institute, Allschwil, Switzerland
- University of Basel, Basel, Switzerland
| | - Scott D Boyd
- Department of Pathology, Stanford University School of Medicine, Stanford, California, USA;
- Sean N. Parker Center for Allergy and Asthma Research, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
6
|
Pastore G, Polvere J, Fiorino F, Lucchesi S, Montesi G, Rancan I, Zirpoli S, Lippi A, Durante M, Fabbiani M, Tumbarello M, Montagnani F, Medaglini D, Ciabattini A. Homologous or heterologous administration of mRNA or adenovirus-vectored vaccines show comparable immunogenicity and effectiveness against the SARS-CoV-2 Omicron variant. Expert Rev Vaccines 2024; 23:432-444. [PMID: 38517153 DOI: 10.1080/14760584.2024.2333952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 03/19/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Heterologous prime-boost schedules have been employed in SARS-CoV-2 vaccination, yet additional data on immunogenicity and effectiveness are still needed. RESEARCH DESIGN AND METHODS Here, we measured the immunogenicity and effectiveness in the real-world setting of the mRNA booster dose in 181 subjects who had completed primary vaccination with ChAdOx1, BNT162b2, or mRNA1273 vaccines (IMMUNO_COV study; protocol code 18,869). The spike-specific antibody and B cell responses were analyzed up to 6 months after boosting. RESULTS After an initial slower antibody response, the heterologous ChAdOx1/mRNA prime-boost formulation elicited spike-specific IgG titers comparable to homologous approaches, while spike-specific B cells showed a higher percentage of CD21-CD27- atypical cells compared to homologous mRNA vaccination. Mixed combinations of BNT162b2 and mRNA-1273 elicited an immune response comparable with homologous strategies. Non-significant differences in the Relative Risk of infection, calculated over a period of 18 months after boosting, were reported among homologous or heterologous vaccination groups, indicating a comparable relative vaccine effectiveness. CONCLUSIONS Our data endorse the heterologous booster vaccination with mRNA as a valuable alternative to homologous schedules. This approach can serve as a solution in instances of formulation shortages and contribute to enhancing vaccine strategies for potential epidemics or pandemics.
Collapse
Affiliation(s)
- Gabiria Pastore
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Jacopo Polvere
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Department of Medicine and Surgery, LUM University "Giuseppe Degennaro"; Casamassima, Bari, Italy
| | - Simone Lucchesi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Giorgio Montesi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Ilaria Rancan
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, Siena, Italy
| | - Sara Zirpoli
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Arianna Lippi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, Siena, Italy
| | - Miriam Durante
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | | | - Mario Tumbarello
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, Siena, Italy
| | - Francesca Montagnani
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, Siena, Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
7
|
Panza F, Fiorino F, Pastore G, Fiaschi L, Tumbarello M, Medaglini D, Ciabattini A, Montagnani F, Fabbiani M. Does Nirmatrelvir/Ritonavir Influence the Immune Response against SARS-CoV-2, Independently from Rebound? Microorganisms 2023; 11:2607. [PMID: 37894265 PMCID: PMC10609571 DOI: 10.3390/microorganisms11102607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/14/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
Recurrence of coronavirus disease 19 (COVID-19) symptoms and SARS-CoV-2 viral load relapse have been reported in people treated with nirmatrelvir/ritonavir (NM/r). However, little is understood about the etiology of this phenomenon. Our aim was to investigate the relation between the host's immune response and viral rebound. We described three cases of COVID-19 rebound that occurred after treatment with nirmatrelvir/ritonavir (group A). In addition, we compared spike-specific antibody response and plasma cytokine/chemokine patterns of the rebound cases with those of (i) control patients treated with nirmatrelvir/ritonavir who did not show rebound (group B), and (ii) subjects not treated with any anti-SARS-CoV-2 drug (group C). The anti-spike antibodies and plasma cytokines/chemokines were similar in groups A and B. However, we observed a higher anti-BA.2 spike IgG response in patients without antiviral treatment (group C) [geometric mean titer 210,807, 5.1- and 8.2-fold higher compared to group A (p = 0.039) and group B (p = 0.032)]. Moreover, the patients receiving antiviral treatment (groups A-B) showed higher circulating levels of platelet-derived growth factor subunit B (PDGF-BB) and vascular endothelial growth Factors (VEGF) and lower levels of interleukin-9 (IL-9), interleukine-1 receptor antagonist (IL-1 RA), and regulated upon activation normal T cell expressed and presumably secreted chemokine (RANTES) when compared to group C. In conclusion, we observed lower anti-spike IgG levels and different cytokine patterns in nirmatrelvir/ritonavir-treated patients compared to those not treated with anti-SARS-CoV-2 drugs. This suggests that early antiviral treatment, by reducing viral load and antigen presentation, could mitigate the immune response against SARS-CoV-2. The clinical relevance of such observation should be further investigated in larger populations.
Collapse
Affiliation(s)
- Francesca Panza
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.P.); (L.F.); (M.T.)
- Infectious and Tropical Diseases Unit, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy;
| | - Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (G.P.); (D.M.); (A.C.)
- Department of Medicine and Surgery, LUM University “Giuseppe Degennaro”, Casamassima, 70010 Bari, Italy
| | - Gabiria Pastore
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (G.P.); (D.M.); (A.C.)
| | - Lia Fiaschi
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.P.); (L.F.); (M.T.)
| | - Mario Tumbarello
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.P.); (L.F.); (M.T.)
- Infectious and Tropical Diseases Unit, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy;
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (G.P.); (D.M.); (A.C.)
| | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.F.); (G.P.); (D.M.); (A.C.)
| | - Francesca Montagnani
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (F.P.); (L.F.); (M.T.)
- Infectious and Tropical Diseases Unit, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy;
| | - Massimiliano Fabbiani
- Infectious and Tropical Diseases Unit, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy;
| |
Collapse
|
8
|
Lopez-Gomez A, Pelaez-Prestel HF, Juarez I. Approaches to evaluate the specific immune responses to SARS-CoV-2. Vaccine 2023; 41:6434-6443. [PMID: 37770298 DOI: 10.1016/j.vaccine.2023.09.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/12/2023] [Accepted: 09/19/2023] [Indexed: 09/30/2023]
Abstract
The SARS-CoV-2 pandemic has a huge impact on public health and global economy, meaning an enormous scientific, political, and social challenge. Studying how infection or vaccination triggers both cellular and humoral responses is essential to know the grade and length of protection generated in the population. Nowadays, scientists and authorities around the world are increasingly concerned about the arrival of new variants, which have a greater spread, due to the high mutation rate of this virus. The aim of this review is to summarize the different techniques available for the study of the immune responses after exposure or vaccination against SARS-CoV-2, showing their advantages and limitations, and proposing suitable combinations of different techniques to achieve extensive information in these studies. We wish that the information provided here will helps other scientists in their studies of the immune response against SARS-CoV-2 after vaccination with new vaccine candidates or infection with upcoming variants.
Collapse
Affiliation(s)
- Ana Lopez-Gomez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Hector F Pelaez-Prestel
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain.
| | - Ignacio Juarez
- Department of Immunology, Ophthalmology and ENT, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
9
|
Agbarya A, Sarel I, Ziv-Baran T, Schwartz O, Shechtman Y, Kozlener E, Khoury R, Sheikh-Ahmad M, Saiegh L, Swaid F, Ahmad AA, Janzic U, Brenner R. Response Rate of the Third and Fourth Doses of the BNT162b2 Vaccine Administered to Cancer Patients Undergoing Active Anti-Neoplastic Treatments. Diseases 2023; 11:128. [PMID: 37873772 PMCID: PMC10594524 DOI: 10.3390/diseases11040128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/25/2023] Open
Abstract
The BNT162b2 vaccine is globally used for preventing morbidity and mortality related to COVID-19. Cancer patients have had priority for receiving the vaccine due to their diminished immunity. This study reports the response rate of administering the third and fourth vaccine doses to cancer patients receiving active anti-neoplastic treatment. A total of 142 patients received two doses of the mRNA-based BNT162b2 COVID-19 vaccine, while 76 and 25 patients received three and four doses, respectively. The efficacy of the humoral response following two vaccine doses was diminished in cancer patients, especially in the group of patients receiving chemotherapy. In a multivariate analysis, patients who received three and four BNT162b2 vaccine doses were more likely to have antibody titers in the upper tertile compared to patients who received two doses of the vaccine (odds ratio (OR) 7.62 (95% CI 1.38-42.12), p = 0.02 and 17.15 (95% CI 5.01-58.7), p < 0.01, respectively). Unlike the response after two doses, the third and fourth BNT162b2 vaccine booster doses had an increased efficacy of 95-100% in cancer patients while undergoing active treatment. This result could be explained by different mechanisms including the development of memory B cells.
Collapse
Affiliation(s)
- Abed Agbarya
- Bnai-Zion Medical Center, Oncology Institute, Haifa 3339419, Israel; (Y.S.); (E.K.); (R.K.); (M.S.-A.); (L.S.); (F.S.); (A.A.A.)
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | - Ina Sarel
- Edith Wolfson Medical Center, Oncology Institute, Holon 5822012, Israel;
| | - Tomer Ziv-Baran
- School of Public Health, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel;
| | - Orna Schwartz
- Microbiology and Immunology Laboratory, Edith Wolfson Medical Center, Holon 5822012, Israel;
| | - Yelena Shechtman
- Bnai-Zion Medical Center, Oncology Institute, Haifa 3339419, Israel; (Y.S.); (E.K.); (R.K.); (M.S.-A.); (L.S.); (F.S.); (A.A.A.)
| | - Ella Kozlener
- Bnai-Zion Medical Center, Oncology Institute, Haifa 3339419, Israel; (Y.S.); (E.K.); (R.K.); (M.S.-A.); (L.S.); (F.S.); (A.A.A.)
| | - Rasha Khoury
- Bnai-Zion Medical Center, Oncology Institute, Haifa 3339419, Israel; (Y.S.); (E.K.); (R.K.); (M.S.-A.); (L.S.); (F.S.); (A.A.A.)
| | - Mohammad Sheikh-Ahmad
- Bnai-Zion Medical Center, Oncology Institute, Haifa 3339419, Israel; (Y.S.); (E.K.); (R.K.); (M.S.-A.); (L.S.); (F.S.); (A.A.A.)
| | - Leonard Saiegh
- Bnai-Zion Medical Center, Oncology Institute, Haifa 3339419, Israel; (Y.S.); (E.K.); (R.K.); (M.S.-A.); (L.S.); (F.S.); (A.A.A.)
| | - Forat Swaid
- Bnai-Zion Medical Center, Oncology Institute, Haifa 3339419, Israel; (Y.S.); (E.K.); (R.K.); (M.S.-A.); (L.S.); (F.S.); (A.A.A.)
| | - Asala Abu Ahmad
- Bnai-Zion Medical Center, Oncology Institute, Haifa 3339419, Israel; (Y.S.); (E.K.); (R.K.); (M.S.-A.); (L.S.); (F.S.); (A.A.A.)
| | - Urska Janzic
- Department of Medical Oncology, University Clinic Golnik, 4202 Golnik, Slovenia;
| | - Ronen Brenner
- Edith Wolfson Medical Center, Oncology Institute, Holon 5822012, Israel;
| |
Collapse
|
10
|
Wang H, Yuan Y, Wu B, Xiao M, Wang Z, Diao T, Zeng R, Chen L, Lei Y, Long P, Guo Y, Lai X, Wen Y, Li W, Cai H, Song L, Ni W, Zhao Y, Ouyang K, Wang J, Wang Q, Liu L, Wang C, Pan A, Li X, Gong R, Wu T. Neutralization against SARS-CoV-2 Delta/Omicron variants and B cell response after inactivated vaccination among COVID-19 convalescents. Front Med 2023; 17:747-757. [PMID: 36738428 PMCID: PMC9898702 DOI: 10.1007/s11684-022-0954-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/20/2022] [Indexed: 02/05/2023]
Abstract
Emerging SARS-CoV-2 variants have made COVID-19 convalescents susceptible to re-infection and have raised concern about the efficacy of inactivated vaccination in neutralization against emerging variants and antigen-specific B cell response. To this end, a study on a long-term cohort of 208 participants who have recovered from COVID-19 was conducted, and the participants were followed up at 3.3 (Visit 1), 9.2 (Visit 2), and 18.5 (Visit 3) months after SARS-CoV-2 infection. They were classified into three groups (no-vaccination (n = 54), one-dose (n = 62), and two-dose (n = 92) groups) on the basis of the administration of inactivated vaccination. The neutralizing antibody (NAb) titers against the wild-type virus continued to decrease in the no-vaccination group, but they rose significantly in the one-dose and two-dose groups, with the highest NAb titers being observed in the two-dose group at Visit 3. The NAb titers against the Delta variant for the no-vaccination, one-dose, and two-dose groups decreased by 3.3, 1.9, and 2.3 folds relative to the wild-type virus, respectively, and those against the Omicron variant decreased by 7.0, 4.0, and 3.8 folds, respectively. Similarly, the responses of SARS-CoV-2 RBD-specific B cells and memory B cells were boosted by the second vaccine dose. Results showed that the convalescents benefited from the administration of the inactivated vaccine (one or two doses), which enhanced neutralization against highly mutated SARS-CoV-2 variants and memory B cell responses. Two doses of inactivated vaccine among COVID-19 convalescents are therefore recommended for the prevention of the COVID-19 pandemic, and vaccination guidelines and policies need to be updated.
Collapse
Affiliation(s)
- Hao Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Yuan
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bihao Wu
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Mingzhong Xiao
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
| | - Zhen Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Tingyue Diao
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Rui Zeng
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Chen
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yanshou Lei
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pinpin Long
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yi Guo
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuefeng Lai
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuying Wen
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wenhui Li
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Hao Cai
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Lulu Song
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Ni
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
| | - Youyun Zhao
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
| | - Kani Ouyang
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
| | - Jingzhi Wang
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China
| | - Qi Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Liu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chaolong Wang
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - An Pan
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaodong Li
- Affiliated Hospital of Hubei University of Chinese Medicine, Wuhan, 430061, China.
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, China.
| | - Rui Gong
- CAS Key Laboratory of Special Pathogens and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China.
| | - Tangchun Wu
- Ministry of Education and State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
11
|
Skarke C, Lordan R, Barekat K, Naik A, Mathew D, Ohtani T, Greenplate AR, Grant GR, Lahens NF, Gouma S, Troisi E, Sengupta A, Weljie AM, Meng W, Luning Prak ET, Lundgreen K, Bates P, Meng H, FitzGerald GA. Modulation of the Immune Response to Severe Acute Respiratory Syndrome Coronavirus 2 Vaccination by Nonsteroidal Anti-Inflammatory Drugs. J Pharmacol Exp Ther 2023; 386:198-204. [PMID: 37105582 PMCID: PMC10353078 DOI: 10.1124/jpet.122.001415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 01/13/2023] [Accepted: 02/09/2023] [Indexed: 04/29/2023] Open
Abstract
Evidence is scarce to guide the use of nonsteroidal anti-inflammatory drugs (NSAIDs) to mitigate severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine-related adverse effects, given the possibility of blunting the desired immune response. In this pilot study, we deeply phenotyped a small number of volunteers who did or did not take NSAIDs concomitant with SARS-CoV-2 immunizations to seek initial information on the immune response. A SARS-CoV-2 vaccine-specific receptor binding domain (RBD) IgG antibody response and efficacy in the evoked neutralization titers were evident irrespective of concomitant NSAID consumption. Given the sample size, only a large and consistent signal of immunomodulation would have been detectable, and this was not apparent. However, the information gathered may inform the design of a definitive clinical trial. Here we report a series of divergent omics signals that invites additional hypotheses testing. SIGNIFICANCE STATEMENT: The impact of nonsteroidal anti-inflammatory drugs (NSAIDs) on the immune response elicited by repeat severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) immunizations was profiled by immunophenotypic, proteomic, and metabolomic approaches in a clinical pilot study of small sample size. A SARS-CoV-2 vaccine-specific immune response was evident irrespective of concomitant NSAID consumption. The information gathered may inform the design of a definitive clinical trial.
Collapse
Affiliation(s)
- Carsten Skarke
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ronan Lordan
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kayla Barekat
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Amruta Naik
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Divij Mathew
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Takuya Ohtani
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Allison R Greenplate
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Gregory R Grant
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Nicholas F Lahens
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Sigrid Gouma
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Elizabeth Troisi
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Arjun Sengupta
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Aalim M Weljie
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Wenzhao Meng
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Eline T Luning Prak
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kendall Lundgreen
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Paul Bates
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Hu Meng
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Garret A FitzGerald
- Institute for Translational Medicine and Therapeutics (C.S., R.L., K.B., A.N., G.R.G., N.F.L., A.S., A.M.W., H.M., G.A.F.), Department of Medicine (C.S., G.A.F.), Institute for Immunology (D.M., T.O., A.R.G.), Immune Health (A.R.G.), Department of Microbiology (S.G., E.T., A.S., K.L., P.B.), Department of Systems Pharmacology and Translational Therapeutics (A.M.W.), and Department of Pathology and Laboratory Medicine (W.M., E.T.L.P.), University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
12
|
Masson LC, Servian CDP, Jardim VH, Dos Anjos D, Dorta ML, Batalha-Carvalho JV, Moro AM, Romão PRT, Souza M, Fiaccadori FS, Fonseca SG. Heterologous Booster with BNT162b2 Induced High Specific Antibody Levels in CoronaVac Vaccinees. Vaccines (Basel) 2023; 11:1183. [PMID: 37514999 PMCID: PMC10383528 DOI: 10.3390/vaccines11071183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/21/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
Immune responses after COVID-19 vaccination should be evaluated in different populations around the world. This study compared antibody responses induced by ChAdOx1 nCoV-19, CoronaVac, and BNT162b2 vaccines. Blood samples from vaccinees were collected pre- and post-vaccinations with the second and third doses. The study enrolled 78 vaccinees, of whom 62.8% were women, with the following median ages: 26 years-ChAdOx1 nCoV-19; 40 years-CoronaVac; 30 years-BNT162b2. Serum samples were quantified for anti-RBD IgG and anti-RBD IgA and anti-spike IgG by ELISA. After two vaccine doses, BNT162b2 vaccinees produced higher levels of anti-RBD IgA and IgG, and anti-spike IgG compared to ChAdOx1 nCoV-19 and CoronaVac vaccinees. The third dose booster with BNT162b2 induced higher levels of anti-RBD IgA and IgG, and anti-spike IgG in CoronaVac vaccinees. Individuals who reported a SARS-CoV-2 infection before or during the study had higher anti-RBD IgA and IgG production. In conclusion, two doses of the studied vaccines induced detectable levels of anti-RBD IgA and IgG and anti-spike IgG in vaccinees. The heterologous booster with BNT162b2 increased anti-RBD IgA and IgG and anti-spike IgG levels in CoronaVac vaccinees and anti-RBD IgA levels in ChAdOx1 nCoV-19 vaccinees. Furthermore, SARS-CoV-2 infection induced higher anti-RBD IgA and IgG levels in CoronaVac vaccinees.
Collapse
Affiliation(s)
- Letícia Carrijo Masson
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Carolina do Prado Servian
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Vitor Hugo Jardim
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Déborah Dos Anjos
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Miriam Leandro Dorta
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | | | - Ana Maria Moro
- Laboratório de Biofármacos, Instituto Butantan, São Paulo 05503-900, SP, Brazil
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (iii-INCT), Goiânia 74605-050, GO, Brazil
| | - Pedro Roosevelt Torres Romão
- Laboratório de Imunologia Celular e Molecular, Programa de Pós-Graduação em Ciências da Saúde, Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre 90050-170, RS, Brazil
| | - Menira Souza
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Fabiola Souza Fiaccadori
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
| | - Simone Gonçalves Fonseca
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia 74605-050, GO, Brazil
- Instituto de Investigação em Imunologia, Instituto Nacional de Ciência e Tecnologia (iii-INCT), Goiânia 74605-050, GO, Brazil
| |
Collapse
|
13
|
Ciabattini A, Pastore G, Lucchesi S, Montesi G, Costagli S, Polvere J, Fiorino F, Pettini E, Lippi A, Ancillotti L, Tumbarello M, Fabbiani M, Montagnani F, Medaglini D. Trajectory of Spike-Specific B Cells Elicited by Two Doses of BNT162b2 mRNA Vaccine. Cells 2023; 12:1706. [PMID: 37443740 PMCID: PMC10340653 DOI: 10.3390/cells12131706] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/14/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
The mRNA vaccines for SARS-CoV-2 have demonstrated efficacy and immunogenicity in the real-world setting. However, most of the research on vaccine immunogenicity has been centered on characterizing the antibody response, with limited exploration into the persistence of spike-specific memory B cells. Here we monitored the durability of the memory B cell response up to 9 months post-vaccination, and characterized the trajectory of spike-specific B cell phenotypes in healthy individuals who received two doses of the BNT162b2 vaccine. To profile the spike-specific B cell response, we applied the tSNE and Cytotree automated approaches. Spike-specific IgA+ and IgG+ plasmablasts and IgA+ activated cells were observed 7 days after the second dose and disappeared 3 months later, while subsets of spike-specific IgG+ resting memory B cells became predominant 9 months after vaccination, and they were capable of differentiating into spike-specific IgG secreting cells when restimulated in vitro. Other subsets of spike-specific B cells, such as IgM+ or unswitched IgM+IgD+ or IgG+ double negative/atypical cells, were also elicited by the BNT162b2 vaccine and persisted up to month 9. The analysis of circulating spike-specific IgG, IgA, and IgM was in line with the plasmablasts observed. The longitudinal analysis of the antigen-specific B cell response elicited by mRNA-based vaccines provides valuable insights into our understanding of the immunogenicity of this novel vaccine platform destined for future widespread use, and it can help in guiding future decisions and vaccination schedules.
Collapse
Affiliation(s)
- Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Gabiria Pastore
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Simone Lucchesi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Giorgio Montesi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Simone Costagli
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Jacopo Polvere
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
- Department of Medicine and Surgery, LUM University “Giuseppe Degennaro”, 70010 Casamassima, Italy
| | - Elena Pettini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| | - Arianna Lippi
- Infectious and Tropical Diseases Unit, Department of Medical Sciences, University Hospital of Siena, 53100 Siena, Italy; (A.L.); (L.A.); (M.T.); (M.F.); (F.M.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Leonardo Ancillotti
- Infectious and Tropical Diseases Unit, Department of Medical Sciences, University Hospital of Siena, 53100 Siena, Italy; (A.L.); (L.A.); (M.T.); (M.F.); (F.M.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Mario Tumbarello
- Infectious and Tropical Diseases Unit, Department of Medical Sciences, University Hospital of Siena, 53100 Siena, Italy; (A.L.); (L.A.); (M.T.); (M.F.); (F.M.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Massimiliano Fabbiani
- Infectious and Tropical Diseases Unit, Department of Medical Sciences, University Hospital of Siena, 53100 Siena, Italy; (A.L.); (L.A.); (M.T.); (M.F.); (F.M.)
| | - Francesca Montagnani
- Infectious and Tropical Diseases Unit, Department of Medical Sciences, University Hospital of Siena, 53100 Siena, Italy; (A.L.); (L.A.); (M.T.); (M.F.); (F.M.)
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy; (G.P.); (S.L.); (G.M.); (S.C.); (J.P.); (F.F.); (E.P.); (D.M.)
| |
Collapse
|
14
|
Brisotto G, Montico M, Turetta M, Zanussi S, Cozzi MR, Vettori R, Boschian Boschin R, Vinante L, Matrone F, Revelant A, Palazzari E, Innocente R, Fanetti G, Gerratana L, Garutti M, Lisanti C, Bolzonello S, Nicoloso MS, Steffan A, Muraro E. Integration of Cellular and Humoral Immune Responses as an Immunomonitoring Tool for SARS-CoV-2 Vaccination in Healthy and Fragile Subjects. Viruses 2023; 15:1276. [PMID: 37376576 DOI: 10.3390/v15061276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 05/26/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
Cellular and humoral immunity are both required for SARS-CoV-2 infection recovery and vaccine efficacy. The factors affecting mRNA vaccination-induced immune responses, in healthy and fragile subjects, are still under investigation. Thus, we monitored the vaccine-induced cellular and humoral immunity in healthy subjects and cancer patients after vaccination to define whether a different antibody titer reflected similar rates of cellular immune responses and if cancer has an impact on vaccination efficacy. We found that higher titers of antibodies were associated with a higher probability of positive cellular immunity and that this greater immune response was correlated with an increased number of vaccination side effects. Moreover, active T-cell immunity after vaccination was associated with reduced antibody decay. The vaccine-induced cellular immunity appeared more likely in healthy subjects rather than in cancer patients. Lastly, after boosting, we observed a cellular immune conversion in 20% of subjects, and a strong correlation between pre- and post-boosting IFN-γ levels, while antibody levels did not display a similar association. Finally, our data suggested that integrating humoral and cellular immune responses could allow the identification of SARS-CoV-2 vaccine responders and that T-cell responses seem more stable over time compared to antibodies, especially in cancer patients.
Collapse
Affiliation(s)
- Giulia Brisotto
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Marcella Montico
- Clinical Trial Office, Scientific Direction, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Matteo Turetta
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Stefania Zanussi
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Maria Rita Cozzi
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Roberto Vettori
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Romina Boschian Boschin
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Lorenzo Vinante
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Fabio Matrone
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Alberto Revelant
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Elisa Palazzari
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Roberto Innocente
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Giuseppe Fanetti
- Division of Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Lorenzo Gerratana
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Mattia Garutti
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Camilla Lisanti
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Silvia Bolzonello
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Milena Sabrina Nicoloso
- Molecular Oncology Unit, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Agostino Steffan
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Elena Muraro
- Immunopathology and Cancer Biomarkers Units, Department of Cancer Research and Advanced Diagnostics, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| |
Collapse
|
15
|
Papaioannidou P, Skoumpa K, Bostanitis C, Michailidou M, Stergiopoulou T, Bostanitis I, Tsalidou M. Age, Sex and BMI Relations with Anti-SARS-CoV-2-Spike IgG Antibodies after BNT162b2 COVID-19 Vaccine in Health Care Workers in Northern Greece. Microorganisms 2023; 11:1279. [PMID: 37317253 DOI: 10.3390/microorganisms11051279] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/26/2023] [Accepted: 05/09/2023] [Indexed: 06/16/2023] Open
Abstract
The aim of this work was to study age, sex, and BMI (Body Mass Index)-related differences in the development of anti-SARS-CoV-2-Spike IgG antibodies, after vaccination with the BNT162b2 COVID-19 vaccine, in health care workers of a General Hospital in a city in Northern Greece. Blood sampling was drawn two to four weeks following the second dose of the vaccine, and six months after the first blood sample collection. Measurement of serum IgG antibodies against the spike domain of SARS-CoV-2 was performed using the SARS-CoV-2 IgG II Quant assay. All participants had sufficient serum IgG titers in the first measurement. Women developed higher IgG titers than men. The IgG titers were inversely related to age in both sexes; there was also a small, insignificant tendency to be inversely related to BMI. Six months after the first measurement, the IgG titers decreased dramatically to values less than 5% of the initial. This decrease was observed in both men and women and was inversely related to age. Multivariate regression analysis showed that age and sex explained with statistical significance 9% of the variance in SARS-CoV-2 IgG titers in our study population; the role of BMI was limited and insignificant.
Collapse
Affiliation(s)
- Paraskevi Papaioannidou
- 1st Department of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Kalypso Skoumpa
- Microbiological Department, General Hospital of Katerini, 601 00 Katerini, Greece
| | - Christos Bostanitis
- 1st Department of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Maria Michailidou
- 1st Department of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Theodouli Stergiopoulou
- 1st Department of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Ioannis Bostanitis
- 1st Department of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
| | - Maria Tsalidou
- 1st Department of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece
- Microbiological Department, General Hospital of Katerini, 601 00 Katerini, Greece
| |
Collapse
|
16
|
Mansourabadi AH, Aghamajidi A, Dorfaki M, Keshavarz F, Shafeghat Z, Moazzeni A, Arab FL, Rajabian A, Roozbehani M, Falak R, Faraji F, Jafari R. B lymphocytes in COVID-19: a tale of harmony and discordance. Arch Virol 2023; 168:148. [PMID: 37119286 PMCID: PMC10147999 DOI: 10.1007/s00705-023-05773-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 03/29/2023] [Indexed: 05/01/2023]
Abstract
B lymphocytes play a vital role in the human defense against viral infections by producing specific antibodies. They are also critical for the prevention of infectious diseases by vaccination, and their activation influences the efficacy of the vaccination. Since the beginning of coronavirus disease 2019 (COVID-19), which became the main concern of the world health system, many efforts have been made to treat and prevent the disease. However, for the development of successful therapeutics and vaccines, it is necessary to understand the interplay between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, and the immune system. The innate immune system provides primary and nonspecific defense against the virus, but within several days after infection, a virus-specific immune response is provided first by antibody-producing B cells, which are converted after the resolution of disease to memory B cells, which provide long-term immunity. Although a failure in B cell activation or B cell dysfunction can cause a severe form of the disease and also lead to vaccination inefficiency, some individuals with B cell immunodeficiency have shown less production of the cytokine IL-6, resulting in a better disease outcome. In this review, we present the latest findings on the interaction between SARS-CoV-2 and B lymphocytes during COVID-19 infection.
Collapse
Affiliation(s)
- Amir Hossein Mansourabadi
- Department of Immunology, School of medicine, Tehran University of Medical Sciences, Tehran, Iran
- Immunogenetics Research Network (IgReN), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Azin Aghamajidi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Dorfaki
- Department of Microbiology and Immunology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Keshavarz
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Shafeghat
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Moazzeni
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization (IBTO), Tehran, Iran
| | - Fahimeh Lavi Arab
- Department of Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Immunology Research Center, School of Medicine, Bu-Ali Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arezoo Rajabian
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Roozbehani
- Vaccine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Faraji
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Antimicrobial Resistance Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran.
- Institue of Immunology and Infectious diseases, Hazrat-e Rasool General Hospital, Floor 3, Building no. 3, Niyayesh St, Sattar Khan St, 1445613131, Tehran, Iran.
| | - Reza Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.
- Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Shafa St., Ershad Blvd, P.O. Box: 1138, 57147, Urmia, Iran.
| |
Collapse
|
17
|
Pozdnyakova V, Weber B, Cheng S, Ebinger JE. Review of Immunologic Manifestations of COVID-19 Infection and Vaccination. Heart Fail Clin 2023; 19:177-184. [PMID: 36863809 PMCID: PMC9973544 DOI: 10.1016/j.hfc.2022.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
We herein summarize currently available and clinically relevant information regarding the human immune responses to SARS-CoV-2 infection and vaccination, in relation to COVID-19 outcomes with a focus on acute respiratory distress syndrome (ARDS) and myocarditis.
Collapse
Affiliation(s)
- Valeriya Pozdnyakova
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Boulevard, D4005, Los Angeles, CA 90048, USA
| | - Brittany Weber
- Carl J. and Ruth Shapiro Cardiovascular Center, Brigham and Women's Hospital, 70 Francis Street, Boston, MA 02115, USA
| | - Susan Cheng
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 South Vicente Boulevard, Suite A3100, Los Angeles, CA 90048, USA
| | - Joseph E Ebinger
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, 127 South Vicente Boulevard, Suite A3100, Los Angeles, CA 90048, USA.
| |
Collapse
|
18
|
Bisceglia H, Barrier J, Ruiz J, Pagnon A. A FluoroSpot B assay for the detection of IgA and IgG SARS-CoV-2 spike-specific memory B cells: Optimization and qualification for use in COVID-19 vaccine trials. J Immunol Methods 2023; 515:113457. [PMID: 36914088 PMCID: PMC10008040 DOI: 10.1016/j.jim.2023.113457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/23/2023] [Accepted: 03/08/2023] [Indexed: 03/13/2023]
Abstract
BACKGROUND The generation of antigen-specific memory B cells is crucial to the long-term effectiveness of vaccines. When the protective antibodies circulating in the blood wane, memory B cells (MBC) can be rapidly reactivated and differentiated into antibody-secreting cells during a new infection. Such MBC responses are considered to be key in providing long-term protection after infection or vaccination. Here, we describe the optimization and qualification of a FluoroSpot assay to measure MBCs directed against the SARS-CoV-2 spike protein in the peripheral blood, for use in COVID-19 vaccine trials. METHODS We developed a FluoroSpot assay enabling simultaneous enumeration of B cells secreting IgA or IgG spike-specific antibodies after polyclonal stimulation of peripheral blood mononuclear cells (PBMCs) with interleukin-2 and the toll-like receptor agonist R848 for 5 days. The antigen coating was optimized using a capture antibody directed against the spike subunit-2 glycoprotein of SARS-CoV-2 to immobilize recombinant trimeric spike protein onto the membrane. RESULTS Compared to a direct spike protein coating, the addition of a capture antibody increased the number and the quality of detected spots for both spike-specific IgA and IgG secreting cells in PBMCs from COVID-19 convalescents. The qualification showed good sensitivity of the dual-color IgA-IgG FluoroSpot assay, with lower limits of quantitation of 18 background-subtracted (BS) antibody-secreting cells (ASCs)/well for spike-specific IgA and IgG responses. Linearity was demonstrated at values ranging from 18 to 73 and from 18 to 607 BS ASCs/well for spike-specific IgA and IgG, respectively, as was precision, with intermediate precision (percentage geometric coefficients of variation) of 12% and 26% for the proportion of spike-specific IgA and IgG MBCs (ratio specific/total IgA or Ig). The assay was specific, since no spike-specific MBCs were detected in PBMCs from pre-pandemic samples; the results were below the limit of detection of 17 BS ASCs/well. CONCLUSIONS These results show that the dual-color IgA-IgG FluoroSpot provides a sensitive, specific, linear, and precise tool to detect spike-specific MBC responses. This MBC FluoroSpot assay is a method of choice for monitoring spike-specific IgA and IgG MBC responses induced by COVID-19 candidate vaccines in clinical trials.
Collapse
Affiliation(s)
- Hélène Bisceglia
- Research Global Immunology Department, Sanofi, Marcy l'Étoile, France
| | - Julie Barrier
- Research Global Immunology Department, Sanofi, Marcy l'Étoile, France
| | - Joseline Ruiz
- Translational and Early Development Biostatistics, Sanofi, Marcy l'Étoile, France
| | - Anke Pagnon
- Research Global Immunology Department, Sanofi, Marcy l'Étoile, France.
| |
Collapse
|
19
|
Newby ML, Fogarty CA, Allen JD, Butler J, Fadda E, Crispin M. Variations within the Glycan Shield of SARS-CoV-2 Impact Viral Spike Dynamics. J Mol Biol 2023; 435:167928. [PMID: 36565991 PMCID: PMC9769069 DOI: 10.1016/j.jmb.2022.167928] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/25/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
The emergence of SARS-CoV-2 variants alters the efficacy of existing immunity, whether arisen naturally or through vaccination. Understanding the structure of the viral spike assists in determining the impact of mutations on the antigenic surface. One class of mutation impacts glycosylation attachment sites, which have the capacity to influence the antigenic structure beyond the immediate site of attachment. Here, we compare the site-specific glycosylation of recombinant viral spike mimetics of B.1.351 (Beta), P.1 (Gamma), B.1.617.2 (Delta), B.1.1.529 (Omicron). The P.1 strain exhibits two additional N-linked glycan sites compared to the other variants analyzed and we investigate the impact of these glycans by molecular dynamics. The acquired N188 site is shown to exhibit very limited glycan maturation, consistent with limited enzyme accessibility. Structural modeling and molecular dynamics reveal that N188 is located within a cavity by the receptor binding domain, which influences the dynamics of these attachment domains. These observations suggest a mechanism whereby mutations affecting viral glycosylation sites have a structural impact across the protein surface.
Collapse
Affiliation(s)
- Maddy L Newby
- School of Biological Sciences, University of Southampton, Southampton, UK. https://twitter.com/Maddy_Newby
| | - Carl A Fogarty
- Department of Chemistry and Hamilton Institute, Maynooth University, Maynooth, Kildare, Ireland. https://twitter.com/2016Carl
| | - Joel D Allen
- School of Biological Sciences, University of Southampton, Southampton, UK. https://twitter.com/JoelDalllen
| | - John Butler
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Elisa Fadda
- Department of Chemistry and Hamilton Institute, Maynooth University, Maynooth, Kildare, Ireland.
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
20
|
Song Z, Luo Q, Wan L, Zhu Q, Liu R, Yin X, Lu X, Wei L, Xiang Z, Zou Y. Analysis of Antibodies Induced after SARS-CoV-2 Vaccination Using Antigen Coded Bead Array Luminex Technology. Vaccines (Basel) 2023; 11:442. [PMID: 36851319 PMCID: PMC9964277 DOI: 10.3390/vaccines11020442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/02/2023] [Accepted: 02/08/2023] [Indexed: 02/17/2023] Open
Abstract
Objectives. Since the outbreak of SARS-CoV-2 in late 2019, nearly 12.2 billion doses of the COVID-19 vaccine have been administered worldwide; however, the humoral immune responses induced by different types of vaccines are yet to be fully validated. Methods. We analyzed antibody levels in 100 serum samples after vaccination with different types of COVID-19 vaccines and their reactivity against the RBD antigen of Delta and Omicron variants using a bead-based microarray. Results. Elevated levels of anti-wild-type (WT)-RBD IgG and anti-WT-NP IgG were detected in participants who received two doses of the inactivated vaccines (CoronaVac or BBIBP-CorV) and three doses of the recombinant spike protein vaccine (ZF2001), indicating that antibody responses to SARS-CoV-2 were generated regardless of the vaccine administered. We found highly correlated levels of serum anti-RBD IgG and anti-NP IgG (r = 0.432, p < 0.001). We observed that the antibodies produced in vivo after COVID-19 vaccination still reacted with variants of SARS-CoV-2 (p < 0.0001). Conclusions. Our results show that high levels of specific antibodies can be produced after completion of COVID-19 vaccination (two doses of the inactivated vaccines or three doses of ZF2001), with some degree of cross-reactivity to the RBD antigen of Delta and Omicron variants, and provide an accessible and practical experimental method for post-vaccination antibody detection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yizhou Zou
- Department of Immunology, School of Basic Medical of Central South University, Changsha 410083, China
| |
Collapse
|
21
|
Brown B, Ojha V, Fricke I, Al-Sheboul SA, Imarogbe C, Gravier T, Green M, Peterson L, Koutsaroff IP, Demir A, Andrieu J, Leow CY, Leow CH. Innate and Adaptive Immunity during SARS-CoV-2 Infection: Biomolecular Cellular Markers and Mechanisms. Vaccines (Basel) 2023; 11:408. [PMID: 36851285 PMCID: PMC9962967 DOI: 10.3390/vaccines11020408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/16/2023] Open
Abstract
The coronavirus 2019 (COVID-19) pandemic was caused by a positive sense single-stranded RNA (ssRNA) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, other human coronaviruses (hCoVs) exist. Historical pandemics include smallpox and influenza, with efficacious therapeutics utilized to reduce overall disease burden through effectively targeting a competent host immune system response. The immune system is composed of primary/secondary lymphoid structures with initially eight types of immune cell types, and many other subtypes, traversing cell membranes utilizing cell signaling cascades that contribute towards clearance of pathogenic proteins. Other proteins discussed include cluster of differentiation (CD) markers, major histocompatibility complexes (MHC), pleiotropic interleukins (IL), and chemokines (CXC). The historical concepts of host immunity are the innate and adaptive immune systems. The adaptive immune system is represented by T cells, B cells, and antibodies. The innate immune system is represented by macrophages, neutrophils, dendritic cells, and the complement system. Other viruses can affect and regulate cell cycle progression for example, in cancers that include human papillomavirus (HPV: cervical carcinoma), Epstein-Barr virus (EBV: lymphoma), Hepatitis B and C (HB/HC: hepatocellular carcinoma) and human T cell Leukemia Virus-1 (T cell leukemia). Bacterial infections also increase the risk of developing cancer (e.g., Helicobacter pylori). Viral and bacterial factors can cause both morbidity and mortality alongside being transmitted within clinical and community settings through affecting a host immune response. Therefore, it is appropriate to contextualize advances in single cell sequencing in conjunction with other laboratory techniques allowing insights into immune cell characterization. These developments offer improved clarity and understanding that overlap with autoimmune conditions that could be affected by innate B cells (B1+ or marginal zone cells) or adaptive T cell responses to SARS-CoV-2 infection and other pathologies. Thus, this review starts with an introduction into host respiratory infection before examining invaluable cellular messenger proteins and then individual immune cell markers.
Collapse
Affiliation(s)
| | | | - Ingo Fricke
- Independent Immunologist and Researcher, 311995 Lamspringe, Germany
| | - Suhaila A Al-Sheboul
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
- Department of Medical Microbiology, International School of Medicine, Medipol University-Istanbul, Istanbul 34810, Turkey
| | | | - Tanya Gravier
- Independent Researcher, MPH, San Francisco, CA 94131, USA
| | | | | | | | - Ayça Demir
- Faculty of Medicine, Afyonkarahisar University, Istanbul 03030, Turkey
| | - Jonatane Andrieu
- Faculté de Médecine, Aix–Marseille University, 13005 Marseille, France
| | - Chiuan Yee Leow
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| | - Chiuan Herng Leow
- Institute for Research in Molecular Medicine, (INFORMM), Universiti Sains Malaysia, USM, Penang 11800, Malaysia
| |
Collapse
|
22
|
Garofalo E, Biamonte F, Palmieri C, Battaglia AM, Sacco A, Biamonte E, Neri G, Antico GC, Mancuso S, Foti G, Torti C, Costanzo FS, Longhini F, Bruni A. Severe and mild-moderate SARS-CoV-2 vaccinated patients show different frequencies of IFNγ-releasing cells: An exploratory study. PLoS One 2023; 18:e0281444. [PMID: 36757971 PMCID: PMC9910754 DOI: 10.1371/journal.pone.0281444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 01/23/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Despite an apparent effective vaccination, some patients are admitted to the hospital after SARS-CoV-2 infection. The role of adaptive immunity in COVID-19 is growing; nonetheless, differences in the spike-specific immune responses between patients requiring or not hospitalization for SARS-CoV-2 infection remains to be evaluated. In this study, we aim to evaluate the spike-specific immune response in patients with mild-moderate or severeSARS-CoV-2 infection, after breakthrough infection following two doses of BNT162b2 mRNA vaccine. METHODS We included three cohorts of 15 cases which received the two BNT162b2 vaccine doses in previous 4 to 7 months: 1) patients with severe COVID-19; 2) patients with mild-moderate COVID-19 and 3) vaccinated individuals with a negative SARS-CoV-2 molecular pharyngeal swab (healthy subjects). Anti-S1 and anti-S2 specific SARS-CoV-2 IgM and IgG titers were measured through a chemiluminescence immunoassay technology. In addition, the frequencies of IFNγ-releasing cells were measured by ELISpot. RESULTS The spike-specific IFNγ-releasing cells were significantly lower in severe patients (8 [0; 26] s.f.c.×106), as compared to mild-moderate patients (135 [64; 159] s.f.c.×106; p<0.001) and healthy subjects (103 [50; 188] s.f.c.×106; p<0.001). The anti-Spike protein IgG levels were similar among the three cohorts of cases (p = 0.098). All cases had an IgM titer below the analytic sensitivity of the test. The Receiver Operating Curve analysis indicated the rate of spike-specific IFNγ-releasing cells can discriminate correctly severe COVID-19 and mild-moderate patients (AUC: 0.9289; 95%CI: 0.8376-1.000; p< 0.0001), with a diagnostic specificity of 100% for s.f.c. > 81.2 x 106. CONCLUSIONS 2-doses vaccinated patients requiring hospitalization for severe COVID-19 show a cellular-mediated immune response lower than mild-moderate or healthy subjects, despite similar antibody titers.
Collapse
Affiliation(s)
- Eugenio Garofalo
- Department of Medical and Surgical Sciences, Anesthesia and Intensive Care Unit, University Hospital Mater Domini, Magna Graecia University, Catanzaro, Italy
| | - Flavia Biamonte
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
- Interdepartmental Center of Services (CIS), Molecular Genomics and Pathology, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Camillo Palmieri
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Anna Martina Battaglia
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Alessandro Sacco
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
| | - Eugenio Biamonte
- Department of Medical and Surgical Sciences, Anesthesia and Intensive Care Unit, University Hospital Mater Domini, Magna Graecia University, Catanzaro, Italy
| | - Giuseppe Neri
- Department of Medical and Surgical Sciences, Anesthesia and Intensive Care Unit, University Hospital Mater Domini, Magna Graecia University, Catanzaro, Italy
| | | | - Serafina Mancuso
- Unit of Biochimica Clinica, University Hospital Mater Domini, Catanzaro, Italy
| | - Giuseppe Foti
- Unit of Infectious Disease, Grand Metropolitan Hospital, Reggio Calabria, Italy
| | - Carlo Torti
- Department of Medical and Surgical Sciences, Unit of Infectious and Tropical Diseases, "Magna Graecia" University, Catanzaro, Italy
| | - Francesco Saverio Costanzo
- Department of Experimental and Clinical Medicine, "Magna Graecia" University of Catanzaro, Catanzaro, Italy
- Interdepartmental Center of Services (CIS), Molecular Genomics and Pathology, "Magna Græcia" University of Catanzaro, Catanzaro, Italy
| | - Federico Longhini
- Department of Medical and Surgical Sciences, Anesthesia and Intensive Care Unit, University Hospital Mater Domini, Magna Graecia University, Catanzaro, Italy
| | - Andrea Bruni
- Department of Medical and Surgical Sciences, Anesthesia and Intensive Care Unit, University Hospital Mater Domini, Magna Graecia University, Catanzaro, Italy
| |
Collapse
|
23
|
Lo Tartaro D, Paolini A, Mattioli M, Swatler J, Neroni A, Borella R, Santacroce E, Di Nella A, Gozzi L, Busani S, Cuccorese M, Trenti T, Meschiari M, Guaraldi G, Girardis M, Mussini C, Piwocka K, Gibellini L, Cossarizza A, De Biasi S. Detailed characterization of SARS-CoV-2-specific T and B cells after infection or heterologous vaccination. Front Immunol 2023; 14:1123724. [PMID: 36845156 PMCID: PMC9947839 DOI: 10.3389/fimmu.2023.1123724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
The formation of a robust long-term antigen (Ag)-specific memory, both humoral and cell-mediated, is created following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or vaccination. Here, by using polychromatic flow cytometry and complex data analyses, we deeply investigated the magnitude, phenotype, and functionality of SARS-CoV-2-specific immune memory in two groups of healthy subjects after heterologous vaccination compared to a group of subjects who recovered from SARS-CoV-2 infection. We find that coronavirus disease 2019 (COVID-19) recovered patients show different long-term immunological profiles compared to those of donors who had been vaccinated with three doses. Vaccinated individuals display a skewed T helper (Th)1 Ag-specific T cell polarization and a higher percentage of Ag-specific and activated memory B cells expressing immunoglobulin (Ig)G compared to those of patients who recovered from severe COVID-19. Different polyfunctional properties characterize the two groups: recovered individuals show higher percentages of CD4+ T cells producing one or two cytokines simultaneously, while the vaccinated are distinguished by highly polyfunctional populations able to release four molecules, namely, CD107a, interferon (IFN)-γ, tumor necrosis factor (TNF), and interleukin (IL)-2. These data suggest that functional and phenotypic properties of SARS-CoV-2 adaptive immunity differ in recovered COVID-19 individuals and vaccinated ones.
Collapse
Affiliation(s)
- Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Marco Mattioli
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Julian Swatler
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Anita Neroni
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Rebecca Borella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Elena Santacroce
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Alessia Di Nella
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Licia Gozzi
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
| | - Stefano Busani
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Anesthesia and Intensive Care, Azienda Ospedaliero-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Michela Cuccorese
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, Azienda Unità Sanitaria Locale AUSL/AOU Policlinico, Modena, Italy
| | - Tommaso Trenti
- Department of Laboratory Medicine and Pathology, Diagnostic Hematology and Clinical Genomics, Azienda Unità Sanitaria Locale AUSL/AOU Policlinico, Modena, Italy
| | - Marianna Meschiari
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
| | - Giovanni Guaraldi
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Girardis
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Anesthesia and Intensive Care, Azienda Ospedaliero-Universitaria (AOU) Policlinico and University of Modena and Reggio Emilia, Modena, Italy
| | - Cristina Mussini
- Infectious Diseases Clinics, Azienda Ospedaliero-Universitaria (AOU) Policlinico di Modena, Modena, Italy
- Department of Surgery, Medicine, Dentistry and Morphological Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Katarzyna Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
- National Institute for Cardiovascular Research, Bologna, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia School of Medicine, Modena, Italy
| |
Collapse
|
24
|
State of the art in epitope mapping and opportunities in COVID-19. Future Sci OA 2023; 16:FSO832. [PMID: 36897962 PMCID: PMC9987558 DOI: 10.2144/fsoa-2022-0048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
The understanding of any disease calls for studying specific biological structures called epitopes. One important tool recently drawing attention and proving efficiency in both diagnosis and vaccine development is epitope mapping. Several techniques have been developed with the urge to provide precise epitope mapping for use in designing sensitive diagnostic tools and developing rpitope-based vaccines (EBVs) as well as therapeutics. In this review, we will discuss the state of the art in epitope mapping with a special emphasis on accomplishments and opportunities in combating COVID-19. These comprise SARS-CoV-2 variant analysis versus the currently available immune-based diagnostic tools and vaccines, immunological profile-based patient stratification, and finally, exploring novel epitope targets for potential prophylactic, therapeutic or diagnostic agents for COVID-19.
Collapse
|
25
|
Durability of Vaccine-Induced and Natural Immunity Against COVID-19: A Narrative Review. Infect Dis Ther 2023; 12:367-387. [PMID: 36622633 PMCID: PMC9828372 DOI: 10.1007/s40121-022-00753-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 12/20/2022] [Indexed: 01/10/2023] Open
Abstract
Vaccines developed against SARS-CoV-2 have proven to be highly effective in preventing symptomatic infection. Similarly, prior infection with SARS-CoV-2 has been shown to provide substantial protection against reinfection. However, it has become apparent that the protection provided to an individual after either vaccination or infection wanes over time. Waning protection is driven by both waning immunity over time since vaccination or initial infection, and the evolution of new variants of SARS-CoV-2. Both antibody and T/B-cells levels have been investigated as potential correlates of protection post-vaccination or post-infection. The activity of antibodies and T/B-cells provide some potential insight into the underlying causes of waning protection. This review seeks to summarise what is currently known about the waning of protection provided by both vaccination and/or prior infection, as well as the current information on the respective antibody and T/B-cell responses.
Collapse
|
26
|
B cell response after SARS-CoV-2 mRNA vaccination in people living with HIV. COMMUNICATIONS MEDICINE 2023; 3:13. [PMID: 36717688 PMCID: PMC9886211 DOI: 10.1038/s43856-023-00245-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 01/16/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Limited longitudinal data are available on immune response to mRNA SARS-CoV-2 vaccination in people living with HIV (PLWHIV); therefore, new evidence on induction and persistence of spike-specific antibodies and B cells is needed. METHODS In this pilot study we investigated the spike-specific humoral and B cell responses up to six months after vaccination with two doses of mRNA vaccines in 84 PLWHIV under antiretroviral therapy compared to 79 healthy controls (HCs). RESULTS Spike-specific IgG persisted six months in PLWHIV with no significant differences compared to HCs, even though a significantly lower IgG response was observed in patients with CD4+ T cells < 350/mmc. The frequency of subjects with antibodies capable of inhibiting ACE2/RBD binding was comparable between PLWHIV and HCs a month after the second vaccine dose, then a higher drop was observed in PLWHIV. A comparable percentage of spike-specific memory B cells was observed at month six in PLWHIV and HCs. However, PLWHIV showed a higher frequency of spike-specific IgD- CD27- double-negative memory B cells and a significantly lower rate of IgD- CD27+ Ig-switched memory B cells compared to HCs, suggesting a reduced functionality of the antigen-specific memory B population. CONCLUSIONS The mRNA vaccination against SARS-CoV-2 elicits humoral and B cell responses quantitatively similar between PLWHIV and HCs, but there are important differences in terms of antibody functionality and phenotypes of memory B cells, reinforcing the notion that tailored vaccination policies should be considered for these patients.
Collapse
|
27
|
Disanto G, Galante A, Cantu' M, Sacco R, Mele F, Eisler JJ, Keller F, Bernasconi E, Sallusto F, Zecca C, Gobbi C. Longitudinal Postvaccine SARS-CoV-2 Immunoglobulin G Titers, Memory B-Cell Responses, and Risk of COVID-19 in Multiple Sclerosis Over 1 Year. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200043. [PMID: 36396447 PMCID: PMC9747147 DOI: 10.1212/nxi.0000000000200043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND AND OBJECTIVES Some disease-modifying treatments impair response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines in multiple sclerosis (MS), potentially increasing the risk of breakthrough infections. We aimed to investigate longitudinal SARS-CoV-2 antibody dynamics and memory B cells after 2 and 3 messenger RNA (mRNA) vaccine doses and their association with the risk of COVID-19 in patients with MS on different treatments over 1 year. METHODS Prospective observational cohort study in patients with MS undergoing SARS-CoV-2 mRNA vaccinations. Antispike (anti-S) immunoglobulin G (IgG) titers were measured by chemiluminescence microparticle immunoassay. Frequencies of spike-specific memory B cells were measured on polyclonal stimulation of peripheral blood mononuclear cells and screening of secreted antibodies by ELISA. RESULTS We recruited 120 patients with MS (58 on anti-CD20 antibodies, 9 on sphingosine 1-phosphate (S1P) receptor modulators, 15 on cladribine, 24 on teriflunomide (TFL), and 14 untreated) and collected 392 samples up to 10.8 months after 2 vaccine doses. When compared with untreated patients, anti-CD20 antibodies (β = -2.07, p < 0.001) and S1P modulators (β = -2.02, p < 0.001) were associated with lower anti-S IgG, while TFL and cladribine were not. Anti-S IgG decreased with months since vaccine (β = -0.14, p < 0.001), independently of treatments. Within anti-CD20 patients, anti-S IgG remained higher in those with greater baseline B-cell counts and were not influenced by postvaccine anti-CD20 infusions. Anti-S IgG increase after a 3rd vaccine was mild on anti-CD20 and S1P modulators. Spike-specific memory B-cell responses were weaker on S1P modulators and anti-CD20 than on TFL and influenced by postvaccine anti-CD20 infusions. The frequency of breakthrough infections was comparable between DMTs, but the risk of COVID-19 was predicted by the last measured anti-S IgG titer before infection (OR = 0.56, 95% CI = 0.37-0.86, p = 0.008). DISCUSSION Postvaccine anti-S IgG titers decrease over time regardless of MS treatment and are associated with breakthrough COVID-19. Both humoral and specific memory B-cell responses are diminished on S1P modulators. Within anti-CD20-treated patients, B-cell count at first vaccine determines anti-S IgG production, whereas postvaccine anti-CD20 infusions negatively affect spike-specific memory B cells.
Collapse
Affiliation(s)
- Giulio Disanto
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Alice Galante
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Marco Cantu'
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Rosaria Sacco
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Federico Mele
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Jennifer Jessica Eisler
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Franco Keller
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Enos Bernasconi
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Federica Sallusto
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Chiara Zecca
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland
| | - Claudio Gobbi
- From the Multiple Sclerosis Center (G.D., R.S., J.J.E., C.Z., C.G.), Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano; Institute for Research in Biomedicine (A.G., F.M., F.S.), Università Della Svizzera Italiana, Bellinzona; Institute of Laboratory Medicine (M.C., F.K.), Ente Ospedaliero Cantonale, Bellinzona; Department of Medicine (E.B.), Ente Ospedaliero Cantonale, Lugano; Faculty of Biomedical Sciences (E.B., F.S., C.Z., C.G.), Università Della Svizzera Italiana, Lugano; and Institute of Microbiology (F.S.), ETH Zurich, Switzerland.
| |
Collapse
|
28
|
Perico L, Todeschini M, Casiraghi F, Mister M, Pezzotta A, Peracchi T, Tomasoni S, Trionfini P, Benigni A, Remuzzi G. Long-term adaptive response in COVID-19 vaccine recipients and the effect of a booster dose. Front Immunol 2023; 14:1123158. [PMID: 36926327 PMCID: PMC10011096 DOI: 10.3389/fimmu.2023.1123158] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/16/2023] [Indexed: 03/08/2023] Open
Abstract
We examined the immune response in subjects previously infected with SARS-CoV2 and infection-naïve 9 months after primary 2-dose COVID-19 mRNA vaccination and 3 months after the booster dose in a longitudinal cohort of healthcare workers. Nine months after primary vaccination, previously infected subjects exhibited higher residual antibody levels, with significant neutralizing activity against distinct variants compared to infection-naïve subjects. The higher humoral response was associated with higher levels of receptor binding domain (RBD)-specific IgG+ and IgA+ memory B cells. The booster dose increased neither neutralizing activity, nor the B and T cell frequencies. Conversely, infection-naïve subjects needed the booster to achieve comparable levels of neutralizing antibodies as those found in previously infected subjects after primary vaccination. The neutralizing titer correlated with anti-RBD IFNγ producing T cells, in the face of sustained B cell response. Notably, pre-pandemic samples showed high Omicron cross-reactivity. These data show the importance of the booster dose in reinforcing immunological memory and increasing circulating antibodies in infection-naïve subjects.
Collapse
Affiliation(s)
- Luca Perico
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Marta Todeschini
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Federica Casiraghi
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Marilena Mister
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Anna Pezzotta
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Tobia Peracchi
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Susanna Tomasoni
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Piera Trionfini
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Ariela Benigni
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Department of Molecular Medicine, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
29
|
An Immunological Review of SARS-CoV-2 Infection and Vaccine Serology: Innate and Adaptive Responses to mRNA, Adenovirus, Inactivated and Protein Subunit Vaccines. Vaccines (Basel) 2022; 11:vaccines11010051. [PMID: 36679897 PMCID: PMC9865970 DOI: 10.3390/vaccines11010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus, which is defined by its positive-sense single-stranded RNA (ssRNA) structure. It is in the order Nidovirales, suborder Coronaviridae, genus Betacoronavirus, and sub-genus Sarbecovirus (lineage B), together with two bat-derived strains with a 96% genomic homology with other bat coronaviruses (BatCoVand RaTG13). Thus far, two Alphacoronavirus strains, HCoV-229E and HCoV-NL63, along with five Betacoronaviruses, HCoV-HKU1, HCoV-OC43, SARS-CoV, MERS-CoV, and SARS-CoV-2, have been recognized as human coronaviruses (HCoVs). SARS-CoV-2 has resulted in more than six million deaths worldwide since late 2019. The appearance of this novel virus is defined by its high and variable transmission rate (RT) and coexisting asymptomatic and symptomatic propagation within and across animal populations, which has a longer-lasting impact. Most current therapeutic methods aim to reduce the severity of COVID-19 hospitalization and virus symptoms, preventing the infection from progressing from acute to chronic in vulnerable populations. Now, pharmacological interventions including vaccines and others exist, with research ongoing. The only ethical approach to developing herd immunity is to develop and provide vaccines and therapeutics that can potentially improve on the innate and adaptive system responses at the same time. Therefore, several vaccines have been developed to provide acquired immunity to SARS-CoV-2 induced COVID-19-disease. The initial evaluations of the COVID-19 vaccines began in around 2020, followed by clinical trials carried out during the pandemic with ongoing population adverse effect monitoring by respective regulatory agencies. Therefore, durability and immunity provided by current vaccines requires further characterization with more extensive available data, as is presented in this paper. When utilized globally, these vaccines may create an unidentified pattern of antibody responses or memory B and T cell responses that need to be further researched, some of which can now be compared within laboratory and population studies here. Several COVID-19 vaccine immunogens have been presented in clinical trials to assess their safety and efficacy, inducing cellular antibody production through cellular B and T cell interactions that protect against infection. This response is defined by virus-specific antibodies (anti-N or anti-S antibodies), with B and T cell characterization undergoing extensive research. In this article, we review four types of contemporary COVID-19 vaccines, comparing their antibody profiles and cellular aspects involved in coronavirus immunology across several population studies.
Collapse
|
30
|
Barnwal A, Basu B, Tripathi A, Soni N, Mishra D, Banerjee A, Kumar R, Vrati S, Bhattacharyya J. SARS-CoV-2 Spike Protein-Activated Dendritic Cell-Derived Extracellular Vesicles Induce Antiviral Immunity in Mice. ACS Biomater Sci Eng 2022; 8:5338-5348. [PMID: 36445062 PMCID: PMC9717688 DOI: 10.1021/acsbiomaterials.2c01094] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/17/2022] [Indexed: 11/30/2022]
Abstract
The onset and spread of the SARS-CoV-2 virus have created an unprecedented universal crisis. Although vaccines have been developed against the parental SARS-CoV-2, outbreaks of the disease still occur through the appearance of different variants, suggesting a continuous need for improved and effective therapeutic strategies. Therefore, we developed a novel nanovesicle presenting Spike protein on the surface of the dendritic cell-derived extracellular vesicles (DEVs) for use as a potential vaccine platform against SARS-CoV-2. DEVs express peptide/MHC-I (pMHC-I) complexes, CCR-7, on their surface. The immunogenicity and efficacy of the Spike-activated DEVs were tested in mice and compared with free Spike protein. A 1/10 Spike equivalent dose of DEVs showed a superior potency in inducing anti-Spike IgG titers in blood of mice when compared to dendritic cells or free Spike protein treatment. Moreover, DEV-induced sera effectively reduced viral infection by 55-60% within 15 days of booster dose administration. Furthermore, a 1/10 Spike equivalent dose of DEV-treated mice was found to be equally effective in inducing CD19+CD38+ T-cells in the spleen and lymph node; CD8 cells in the bone marrow, spleen, and lymph node; and CD4+CD25+ T-cells in the spleen and lymph node after 90 days of treatment. Thus, our results support the immunogenic nature of DEVs, demonstrating that a low dose of DEVs induces antibodies to inhibit SARS-CoV-2 infection in vitro, therefore warranting further investigations.
Collapse
Affiliation(s)
- Anjali Barnwal
- Centre for Biomedical
Engineering, Indian Institute of Technology
Delhi, New Delhi 110016, India
- Department
of Biomedical Engineering, All India Institute
of Medical Science, New Delhi 110029, India
| | - Brohmomoy Basu
- Laboratory
of Virology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Aarti Tripathi
- Laboratory
of Virology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Naina Soni
- Laboratory
of Virology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Debasish Mishra
- Laboratory
of Virology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Arup Banerjee
- Laboratory
of Virology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Rajesh Kumar
- Translational
Health Science & Technology Institute, Faridabad 121001, Haryana, India
| | - Sudhanshu Vrati
- Laboratory
of Virology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Jayanta Bhattacharyya
- Centre for Biomedical
Engineering, Indian Institute of Technology
Delhi, New Delhi 110016, India
- Department
of Biomedical Engineering, All India Institute
of Medical Science, New Delhi 110029, India
| |
Collapse
|
31
|
Pettini E, Medaglini D, Ciabattini A. Profiling the B cell immune response elicited by vaccination against the respiratory virus SARS-CoV-2. Front Immunol 2022; 13:1058748. [PMID: 36505416 PMCID: PMC9729280 DOI: 10.3389/fimmu.2022.1058748] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
B cells play a fundamental role in host defenses against viral infections. Profiling the B cell response elicited by SARS-CoV-2 vaccination, including the generation and persistence of antigen-specific memory B cells, is essential for improving the knowledge of vaccine immune responsiveness, beyond the antibody response. mRNA-based vaccines have shown to induce a robust class-switched memory B cell response that persists overtime and is boosted by further vaccine administration, suggesting that memory B cells are critical in driving a recall response upon re-exposure to SARS-CoV-2 antigens. Here, we focus on the role of the B cell response in the context of SARS-CoV-2 vaccination, offering an overview of the different technologies that can be used to identify spike-specific B cells, characterize their phenotype using machine learning approaches, measure their capacity to reactivate following antigen encounter, and tracking the maturation of the B cell receptor antigenic affinity.
Collapse
Affiliation(s)
| | | | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
32
|
Differential Kinetics of Effector and Memory Responses Induced by Three Doses of SARS-CoV-2 mRNA Vaccine in a Cohort of Healthcare Workers. Vaccines (Basel) 2022; 10:vaccines10111809. [DOI: 10.3390/vaccines10111809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
We reported the long-term kinetics of immune response after vaccination and evaluated the immunogenicity after a third dose of mRNA vaccine in 86 healthcare workers. Humoral response was analyzed by measuring anti-spike IgG and SARS-CoV-2 NTAbs titer; cell-mediated response was measured as frequency of IFN-γ producing T-cells and cell proliferation. Memory B cells secreting SARS-CoV-2 RBD-IgG were measured by B-spot assay. At three weeks after the third dose (T4), the frequency of subjects showing NT-Abs titer at the upper detection limit (≥640) was significantly higher than that observed at three weeks after the second dose (26/77; 33.7% vs. 9/77; 11.6%; p = 0.0018). Additionally, at T4, all the subjects reached positive levels of T-cell mediated response (median 110 SFU/106 PBMC, IQR 73-231). While the number of IFNγ-producing T-cells decreased between second and third dose administration, the T-cell proliferative response did not decrease but was sustained during the follow-up. Among T-cell subsets, a higher proliferative response was observed in CD4+ than in CD8+ population. Moreover, even if a decline in antibody response was observed between the second and third dose, a sustained persistence of memory B cells was observed. Subsequently, the third dose did not affect the frequency of memory B cells, while it restored or increased the peak antibody levels detected after the second dose.
Collapse
|
33
|
Fiorino F, Ciabattini A, Sicuranza A, Pastore G, Santoni A, Simoncelli M, Polvere J, Galimberti S, Baratè C, Sammartano V, Montagnani F, Bocchia M, Medaglini D. The third dose of mRNA SARS-CoV-2 vaccines enhances the spike-specific antibody and memory B cell response in myelofibrosis patients. Front Immunol 2022; 13:1017863. [PMID: 36248803 PMCID: PMC9556722 DOI: 10.3389/fimmu.2022.1017863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Accepted: 09/06/2022] [Indexed: 11/13/2022] Open
Abstract
Vaccination against SARS-CoV-2 using mRNA-based vaccines has been highly recommended for fragile subjects, including myelofibrosis patients (MF). Available data on the immune responsiveness of MF patients to mRNA SARS-CoV-2 vaccination, and the impact of the therapy with the JAK inhibitor ruxolitinib, are still fragmented. Here, we profile the spike-specific IgG and memory B-cell response in MF patients, treated or not with ruxolitinib, after the second and the third dose of SARS-CoV-2 BNT162b2 (BioNTech) and mRNA-1273 (Moderna) vaccines. Plasma and peripheral blood mononuclear cells samples were collected before vaccination, post the second and the third doses and tested for spike-specific antibodies, ACE2/RBD antibody inhibition binding activity and spike-specific B cells. The third vaccine dose significantly increased the spike-specific IgG titers in both ruxolitinib-treated and untreated patients, and strongly enhanced the percentage of subjects with antibodies capable of in vitro blocking ACE2/RBD interaction, from 50% up to 80%. While a very low frequency of spike-specific B cells was measured in blood 7 days after the second vaccination dose, a strong and significant increase was elicited by the third dose administration, generating a B cell response similar to the one detected in healthy controls. Despite the overall positive impact of the third dose in MF patients, two patients that were under active concomitant immunosuppressive treatment at the time of vaccination, and a patient that received lymphodepleting therapies in the past, remained low responders. The third mRNA vaccine dose strongly increases the SARS-CoV-2 specific humoral and B cell responses in MF patients, promoting a reactivation of the immune response similar to the one observed in healthy controls.
Collapse
Affiliation(s)
- Fabio Fiorino
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Anna Sicuranza
- Hematology Unit, Department of Medical Science, Surgery and Neuroscience, Azienda Ospedaliero Universitaria Senese, University of Siena, Siena, Italy
| | - Gabiria Pastore
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Adele Santoni
- Hematology Unit, Department of Medical Science, Surgery and Neuroscience, Azienda Ospedaliero Universitaria Senese, University of Siena, Siena, Italy
| | - Martina Simoncelli
- Hematology Unit, Department of Medical Science, Surgery and Neuroscience, Azienda Ospedaliero Universitaria Senese, University of Siena, Siena, Italy
| | - Jacopo Polvere
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Sara Galimberti
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Claudia Baratè
- Section of Hematology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Vincenzo Sammartano
- Hematology Unit, Department of Medical Science, Surgery and Neuroscience, Azienda Ospedaliero Universitaria Senese, University of Siena, Siena, Italy
| | - Francesca Montagnani
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, Azienda Ospedaliero Universitaria Senese, University Hospital of Siena, Siena, Italy
| | - Monica Bocchia
- Hematology Unit, Department of Medical Science, Surgery and Neuroscience, Azienda Ospedaliero Universitaria Senese, University of Siena, Siena, Italy
- *Correspondence: Donata Medaglini, ; Monica Bocchia,
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy
- *Correspondence: Donata Medaglini, ; Monica Bocchia,
| |
Collapse
|
34
|
Bhuiyan TR, Al Banna H, Kaisar MH, Karmakar PC, Hakim A, Akter A, Ahmed T, Tauheed I, Islam S, Hasnat MA, Sumon MA, Rashed A, Ghosh S, Clemens JD, Banu S, Shirin T, Weiskopf D, Sette A, Chowdhury F, Qadri F. Correlation of antigen-specific immune response with disease severity among COVID-19 patients in Bangladesh. Front Immunol 2022; 13:929849. [PMID: 36248882 PMCID: PMC9554593 DOI: 10.3389/fimmu.2022.929849] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 09/05/2022] [Indexed: 12/03/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a protean disease causing different degrees of clinical severity including fatality. In addition to humoral immunity, antigen-specific T cells may play a critical role in defining the protective immune response against SARS-CoV-2, the virus that causes this disease. As a part of a longitudinal cohort study in Bangladesh to investigate B and T cell-specific immune responses, we sought to evaluate the activation-induced marker (AIM) and the status of different immune cell subsets during a COVID-19 infection. We analyzed a total of 115 participants, which included participants with asymptomatic, mild, moderate, and severe clinical symptoms. We observed decreased mucosal-associated invariant T (MAIT) cell frequency on the initial days of the COVID-19 infection in symptomatic patients compared to asymptomatic patients. However, natural killer (NK) cells were found to be elevated in symptomatic patients just after the onset of the disease compared to both asymptomatic patients and healthy individuals. Moreover, we found a significant increase of AIM+ (both OX40+CD137+ and OX40+CD40L+) CD4+ T cells in moderate and severe COVID-19 patients in response to SARS-CoV-2 peptides (especially spike peptides) compared to pre-pandemic controls who are unexposed to SARS-CoV-2. Notably, we did not observe any significant difference in the CD8+ AIMs (CD137+CD69+), which indicates the exhaustion of CD8+ T cells during a COVID-19 infection. These findings suggest that patients who recovered from moderate and severe COVID-19 were able to mount a strong CD4+ T-cell response against shared viral determinants that ultimately induced T cells to mount further immune responses to SARS-CoV-2.
Collapse
Affiliation(s)
- Taufiqur Rahman Bhuiyan
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Hasan Al Banna
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - M. Hasanul Kaisar
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Polash Chandra Karmakar
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Al Hakim
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
- Department of Genetic Engineering and Biotechnology, Jagannath University, Dhaka, Bangladesh
| | - Afroza Akter
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Tasnuva Ahmed
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Imam Tauheed
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Shaumik Islam
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Mohammad Abul Hasnat
- Department of Cardiology, Department of Oncology, Kurmitola General Hospital, Dhaka, Bangladesh
| | - Mostafa Aziz Sumon
- Department of Cardiology, Department of Oncology, Kurmitola General Hospital, Dhaka, Bangladesh
| | - Asif Rashed
- Department of Microbiology, Department of Medicine, Mugda Medical College and Hospital, Dhaka, Bangladesh
| | - Shuvro Ghosh
- Department of Microbiology, Department of Medicine, Mugda Medical College and Hospital, Dhaka, Bangladesh
| | - John D. Clemens
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
- Department of Epidemiology, University of California Los Angeles (UCLA) Fielding School of Public Health, Los Angeles, CA, United States
- International Vaccine Institute, Seoul, South Korea
| | - Sayera Banu
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Tahmina Shirin
- Institute of Epidemiology, Disease Control and Research (IEDCR), Dhaka, Bangladesh
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, United States
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, United States
| | - Fahima Chowdhury
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
| | - Firdausi Qadri
- Infectious Diseases Division, International Centre for Diarrhoeal Disease Research Bangladesh (ICDDRB), Dhaka, Bangladesh
- *Correspondence: Firdausi Qadri,
| |
Collapse
|
35
|
Hartley GE, Edwards ESJ, O’Hehir RE, van Zelm MC. New insights into human immune memory from SARS-CoV-2 infection and vaccination. Allergy 2022; 77:3553-3566. [PMID: 36048132 PMCID: PMC9538469 DOI: 10.1111/all.15502] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/13/2022] [Accepted: 08/29/2022] [Indexed: 01/28/2023]
Abstract
Since early 2020, the world has been embroiled in an ongoing viral pandemic with SARS-CoV-2 and emerging variants resulting in mass morbidity and an estimated 6 million deaths globally. The scientific community pivoted rapidly, providing unique and innovative means to identify infected individuals, technologies to evaluate immune responses to infection and vaccination, and new therapeutic strategies to treat infected individuals. Never before has immunology been so critically at the forefront of combatting a global pandemic. It has now become evident that not just antibody responses, but formation and durability of immune memory cells following vaccination are associated with protection against severe disease from SARS-CoV-2 infection. Furthermore, the emergence of variants of concern (VoC) highlight the need for immunological markers to quantify the protective capacity of Wuhan-based vaccines. Thus, harnessing and modulating the immune response is key to successful vaccination and treatment of disease. We here review the latest knowledge about immune memory generation and durability following natural infection and vaccination, and provide insights into the attributes of immune memory that may protect from emerging variants.
Collapse
Affiliation(s)
- Gemma E. Hartley
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Emily S. J. Edwards
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Robyn E. O’Hehir
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia,Allergy, Asthma and Clinical Immunology ServiceAlfred HospitalMelbourneVictoriaAustralia
| | - Menno C. van Zelm
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia,Allergy, Asthma and Clinical Immunology ServiceAlfred HospitalMelbourneVictoriaAustralia
| |
Collapse
|
36
|
Mrak D, Sieghart D, Simader E, Tobudic S, Radner H, Mandl P, Göschl L, Koblischke M, Hommer N, Wagner A, Mayer M, Schubert L, Hartl L, Kozbial K, Hofer P, Kartnig F, Hummel T, Kerschbaumer A, Deimel T, Puchner A, Gudipati V, Thalhammer R, Munda P, Uyanik-Ünal K, Zuckermann A, Novacek G, Reiberger T, Garner-Spitzer E, Reindl-Schwaighofer R, Kain R, Winkler S, Smolen JS, Stiasny K, Fischer GF, Perkmann T, Haslacher H, Zeitlinger M, Wiedermann U, Aberle JH, Aletaha D, Heinz LX, Bonelli M. Heterologous vector versus homologous mRNA COVID-19 booster vaccination in non-seroconverted immunosuppressed patients: a randomized controlled trial. Nat Commun 2022; 13:5362. [PMID: 36097029 PMCID: PMC9467419 DOI: 10.1038/s41467-022-33036-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 08/30/2022] [Indexed: 11/09/2022] Open
Abstract
Impaired response to COVID-19 vaccination is of particular concern in immunosuppressed patients. To determine the best vaccination strategy for this vulnerable group we performed a single center, 1:1 randomized blinded clinical trial. Patients who failed to seroconvert upon two mRNA vaccinations (BNT162b2 or mRNA-1273) are randomized to receive either a third dose of the same mRNA or the vector vaccine ChAdOx1 nCoV-19. Primary endpoint is the difference in SARS-CoV-2 spike antibody seroconversion rate between vector and mRNA vaccinated patients four weeks after the third dose. Secondary outcomes include cellular immune responses. Seroconversion rates at week four are significantly higher in the mRNA (homologous vaccination, 15/24, 63%) as compared to the vector vaccine group (heterologous vaccination, 4/22, 18%). SARS-CoV-2-specific T-cell responses are reduced but could be increased after a third dose of either vector or mRNA vaccine. In a multivariable logistic regression analysis, patient age and vaccine type are associated with seroconversion. No serious adverse event is attributed to COVID-19 booster vaccination. Efficacy and safety data underline the importance of a booster vaccination and support the use of a homologous mRNA booster vaccination in immunosuppressed patients.Trial registration: EudraCT No.: 2021-002693-10.
Collapse
Affiliation(s)
- Daniel Mrak
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Daniela Sieghart
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Simader
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Selma Tobudic
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Helga Radner
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Peter Mandl
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Lisa Göschl
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | | | - Nikolaus Hommer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Angelika Wagner
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, Vienna, Austria
| | - Margareta Mayer
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Lorenz Schubert
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Lukas Hartl
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Karin Kozbial
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Philipp Hofer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Felix Kartnig
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Hummel
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Andreas Kerschbaumer
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Deimel
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Antonia Puchner
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Venugopal Gudipati
- Institute of Hygiene and Applied Immunology, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Renate Thalhammer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Petra Munda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Keziban Uyanik-Ünal
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Andreas Zuckermann
- Department of Cardiac Surgery, Medical University of Vienna, Vienna, Austria
| | - Gottfried Novacek
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Erika Garner-Spitzer
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, Vienna, Austria
| | - Roman Reindl-Schwaighofer
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Renate Kain
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Stefan Winkler
- Division of Infectious Diseases and Tropical Medicine, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
| | - Josef S Smolen
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Gottfried F Fischer
- Department of Blood Group Serology and Transfusion Medicine, Medical University of Vienna, Vienna, Austria
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center of Pathophysiology, Infectiology and Immunology, Medical University Vienna, Vienna, Austria
| | - Judith H Aberle
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Daniel Aletaha
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Leonhard X Heinz
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| | - Michael Bonelli
- Division of Rheumatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
37
|
Fan Z, Duan S, Liu F, Shi W, Yang Z, Bai R, Li T, Chen J, Xie H, Li J, Tang Y. SARS-CoV-2 vaccination in androgen sensitive phenotypes – A study on associated factors for SARS-CoV-2 vaccination and its adverse effects among androgenetic alopecia and benign prostate hyperplasia patients. Front Immunol 2022; 13:919958. [PMID: 36119091 PMCID: PMC9478654 DOI: 10.3389/fimmu.2022.919958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundAndrogen sensitivity, which was established as the leading etiology of androgenetic alopecia (AGA) and benign prostatic hyperplasia (BPH), plays an important role in SARS-CoV-2 infection. Vaccination is essential for AGA and BPH patients in view of the high risk from SARS-CoV-2 infection.PurposeWe aimed to investigate the associated factors for SARS-CoV-2 vaccination and its side effects in populations with AGA and BPH.MethodWe collected the data on SARS-CoV-2 vaccination and adverse reactions of male AGA and BPH patients visited the outpatient of Xiangya hospital by telephone and web-based questionnaires. Vaccination rate and adverse reactions were compared by different vaccine types and use of anti-androgen therapy.ResultA total of 457 AGA patients and 397 BPH patients were recruited in this study. Among which, 92.8% AGA patients and 61.0% BPH patients had at least the first dose of SARS-CoV-2 vaccination (p < 0.001). Having comorbidities and use of anti-androgen therapy increased the risk of un-vaccination among AGA by 2.875 and 3.729 times, respectively (p < 0.001). Around 31.1% AGA patients and 9.5% BPH patients presented adverse reactions, which were mostly mild. Anti-androgen therapy increased the inclination of injection site pain after vaccination (18.7% vs 11.9%; OR: 1.708, 95% CI: 1.088-2.683, p = 0.019).ConclusionCo-existence of other systemic diseases and anti-androgen therapy were the limiting factors for SARS-CoV-2 unvaccination, especially in AGA patients. The importance of SARS-CoV-2 vaccines should be strengthened and popularized in androgen sensitive phenotypes.
Collapse
Affiliation(s)
- Zhihua Fan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Shixin Duan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Fangfen Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Shi
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ziye Yang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Ruiyang Bai
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Tao Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
| | - Jingxian Chen
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yan Tang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yan Tang,
| |
Collapse
|
38
|
Almendro-Vázquez P, Chivite-Lacaba M, Utrero-Rico A, González-Cuadrado C, Laguna-Goya R, Moreno-Batanero M, Sánchez-Paz L, Luczkowiak J, Labiod N, Folgueira MD, Delgado R, Paz-Artal E. Cellular and humoral immune responses and breakthrough infections after three SARS-CoV-2 mRNA vaccine doses. Front Immunol 2022; 13:981350. [PMID: 36059485 PMCID: PMC9428395 DOI: 10.3389/fimmu.2022.981350] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Background SARS-CoV-2 vaccination has proven the most effective measure to control the COVID-19 pandemic. Booster doses are being administered with limited knowledge on their need and effect on immunity. Objective To determine the duration of specific T cells, antibodies and neutralization after 2-dose vaccination, to assess the effect of a third dose on adaptive immunity and to explore correlates of protection against breakthrough infection. Methods 12-month longitudinal assessment of SARS-CoV-2-specific T cells, IgG and neutralizing antibodies triggered by 2 BNT162b2 doses followed by a third mRNA-1273 dose in a cohort of 77 healthcare workers: 17 with SARS-CoV-2 infection prior to vaccination (recovered) and 60 naïve. Results Peak levels of cellular and humoral response were achieved 2 weeks after the second dose. Antibodies declined thereafter while T cells reached a plateau 3 months after vaccination. The decline in neutralization was specially marked in naïve individuals and it was this group who benefited most from the third dose, which resulted in a 20.9-fold increase in neutralization. Overall, recovered individuals maintained higher levels of T cells, antibodies and neutralization 1 to 6 months post-vaccination than naïve. Seventeen asymptomatic or mild SARS-CoV-2 breakthrough infections were reported during follow-up, only in naïve individuals. This viral exposure boosted adaptive immunity. High peak levels of T cells and neutralizing antibodies 15 days post-vaccination associated with protection from breakthrough infections. Conclusion Booster vaccination in naïve individuals and the inclusion of viral antigens other than spike in future vaccine formulations could be useful strategies to prevent SARS-CoV-2 breakthrough infections.
Collapse
Affiliation(s)
- Patricia Almendro-Vázquez
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- *Correspondence: Patricia Almendro-Vázquez,
| | - Marta Chivite-Lacaba
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Alberto Utrero-Rico
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | | | - Rocio Laguna-Goya
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas (CIBERINFEC – Instituto de Salud Carlos III), Madrid, Spain
| | | | - Laura Sánchez-Paz
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Joanna Luczkowiak
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Nuria Labiod
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - María Dolores Folgueira
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Microbiology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Rafael Delgado
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas (CIBERINFEC – Instituto de Salud Carlos III), Madrid, Spain
- Department of Microbiology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Department of Medicine, Medical School, Universidad Complutense de Madrid, Madrid, Spain
| | - Estela Paz-Artal
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas (CIBERINFEC – Instituto de Salud Carlos III), Madrid, Spain
- Department of Immunology, Ophthalmology and ENT, Medical School, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
39
|
Guerrini G, Gioria S, Sauer AV, Lucchesi S, Montagnani F, Pastore G, Ciabattini A, Medaglini D, Calzolai L. Monitoring Anti-PEG Antibodies Level upon Repeated Lipid Nanoparticle-Based COVID-19 Vaccine Administration. Int J Mol Sci 2022; 23:8838. [PMID: 36012103 PMCID: PMC9408675 DOI: 10.3390/ijms23168838] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/01/2022] [Accepted: 08/05/2022] [Indexed: 12/04/2022] Open
Abstract
PEGylated lipids are one of the four constituents of lipid nanoparticle mRNA COVID-19 vaccines. Therefore, various concerns have been raised on the generation of anti-PEG antibodies and their potential role in inducing hypersensitivity reactions following vaccination or in reducing vaccine efficacy due to anti-carrier immunity. Here, we assess the prevalence of anti-PEG antibodies, in a cohort of vaccinated individuals, and give an overview of their time evolution after repeated vaccine administrations. Results indicate that, in our cohort, the presence of PEG in the formulation did not influence the level of anti-Spike antibodies generated upon vaccination and was not related to any reported, serious adverse effects. The time-course analysis of anti-PEG IgG showed no significant booster effect after each dose, whereas for IgM a significant increase in antibody levels was detected after the first and third dose. Data suggest that the presence of PEG in the formulation does not affect safety or efficacy of lipid-nanoparticle-based COVID-19 vaccines.
Collapse
Affiliation(s)
| | - Sabrina Gioria
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Aisha V. Sauer
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Simone Lucchesi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Francesca Montagnani
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
- Department of Medical Sciences, Infectious and Tropical Diseases Unit, University Hospital of Siena, 53100 Siena, Italy
| | - Gabiria Pastore
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Annalisa Ciabattini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Luigi Calzolai
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| |
Collapse
|
40
|
Renia L, Goh YS, Rouers A, Le Bert N, Chia WN, Chavatte JM, Fong SW, Chang ZW, Zhuo NZ, Tay MZ, Chan YH, Tan CW, Yeo NKW, Amrun SN, Huang Y, Wong JXE, Hor PX, Loh CY, Wang B, Ngoh EZX, Salleh SNM, Carissimo G, Dowla S, Lim AJ, Zhang J, Lim JME, Wang CI, Ding Y, Pada S, Sun LJ, Somani J, Lee ES, Ong DLS, Leo YS, MacAry PA, Lin RTP, Wang LF, Ren EC, Lye DC, Bertoletti A, Young BE, Ng LFP. Lower vaccine-acquired immunity in the elderly population following two-dose BNT162b2 vaccination is alleviated by a third vaccine dose. Nat Commun 2022; 13:4615. [PMID: 35941158 PMCID: PMC9358634 DOI: 10.1038/s41467-022-32312-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 07/23/2022] [Indexed: 12/12/2022] Open
Abstract
Understanding the impact of age on vaccinations is essential for the design and delivery of vaccines against SARS-CoV-2. Here, we present findings from a comprehensive analysis of multiple compartments of the memory immune response in 312 individuals vaccinated with the BNT162b2 SARS-CoV-2 mRNA vaccine. Two vaccine doses induce high antibody and T cell responses in most individuals. However, antibody recognition of the Spike protein of the Delta and Omicron variants is less efficient than that of the ancestral Wuhan strain. Age-stratified analyses identify a group of low antibody responders where individuals ≥60 years are overrepresented. Waning of the antibody and cellular responses is observed in 30% of the vaccinees after 6 months. However, age does not influence the waning of these responses. Taken together, while individuals ≥60 years old take longer to acquire vaccine-induced immunity, they develop more sustained acquired immunity at 6 months post-vaccination. A third dose strongly boosts the low antibody responses in the older individuals against the ancestral Wuhan strain, Delta and Omicron variants.
Collapse
Affiliation(s)
- Laurent Renia
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| | - Yun Shan Goh
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Angeline Rouers
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Nina Le Bert
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Wan Ni Chia
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Jean-Marc Chavatte
- National Public Health Laboratory, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Siew-Wai Fong
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Zi Wei Chang
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Nicole Ziyi Zhuo
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Matthew Zirui Tay
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yi-Hao Chan
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Chee Wah Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Nicholas Kim-Wah Yeo
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Siti Naqiah Amrun
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yuling Huang
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Joel Xu En Wong
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Pei Xiang Hor
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Chiew Yee Loh
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Bei Wang
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Eve Zi Xian Ngoh
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Siti Nazihah Mohd Salleh
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Guillaume Carissimo
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Samanzer Dowla
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Alicia Jieling Lim
- National Public Health Laboratory, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Jinyan Zhang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Joey Ming Er Lim
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Cheng-I Wang
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Ying Ding
- National Centre for Infectious Diseases, Singapore, Singapore
| | | | | | - Jyoti Somani
- Division of Infectious Diseases, Department of Medicine, National University Hospital, National University Health System, Singapore, Singapore
| | - Eng Sing Lee
- National healthcare group polyclinic, Jurong, Singapore
| | - Desmond Luan Seng Ong
- National University Polyclinics, National University of Singapore, Singapore, Singapore
| | - Yee-Sin Leo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
- Tan Tock Seng Hospital, Singapore, Singapore
| | - Paul A MacAry
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Life Sciences Institute, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
| | - Raymond Tzer Pin Lin
- National Public Health Laboratory, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
| | - Ee Chee Ren
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - David C Lye
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Tan Tock Seng Hospital, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Antonio Bertoletti
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Barnaby Edward Young
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- National Centre for Infectious Diseases, Singapore, Singapore
- Tan Tock Seng Hospital, Singapore, Singapore
| | - Lisa F P Ng
- A*STAR Infectious Diseases Labs (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- National Institute of Health Research, Health Protection Research Unit in Emerging and Zoonotic Infections, University of Liverpool, Liverpool, UK
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| |
Collapse
|
41
|
Sheikh‐Mohamed S, Sanders EC, Gommerman JL, Tal MC. Guardians of the oral and nasopharyngeal galaxy: IgA and protection against SARS-CoV-2 infection. Immunol Rev 2022; 309:75-85. [PMID: 35815463 PMCID: PMC9349649 DOI: 10.1111/imr.13118] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In early 2020, a global emergency was upon us in the form of the coronavirus disease 2019 (COVID-19) pandemic. While horrific in its health, social and economic devastation, one silver lining to this crisis has been a rapid mobilization of cross-institute, and even cross-country teams that shared common goals of learning as much as we could as quickly as possible about the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and how the immune system would respond to both the virus and COVID-19 vaccines. Many of these teams were formed by women who quickly realized that the classical model of "publish first at all costs" was maladaptive for the circumstances and needed to be supplanted by a more collaborative solution-focused approach. This review is an example of a collaboration that unfolded in separate countries, first Canada and the United States, and then also Israel. Not only did the collaboration allow us to cross-validate our results using different hands/techniques/samples, but it also took advantage of different vaccine types and schedules that were rolled out in our respective home countries. The result of this collaboration was a new understanding of how mucosal immunity to SARS-CoV-2 infection vs COVID-19 vaccination can be measured using saliva as a biofluid, what types of vaccines are best able to induce (limited) mucosal immunity, and what are potential correlates of protection against breakthrough infection. In this review, we will share what we have learned about the mucosal immune response to SARS-CoV-2 and to COVID-19 vaccines and provide a perspective on what may be required for next-generation pan-sarbecoronavirus vaccine approaches.
Collapse
Affiliation(s)
| | - Erin C. Sanders
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
| | | | - Michal Caspi Tal
- Department of Biological EngineeringMassachusetts Institute of TechnologyCambridgeMassachusettsUSA
- Institute for Stem Cell Biology and Regenerative Medicine and the Ludwig Cancer CenterStanford University School of MedicineStanfordCaliforniaUSA
| |
Collapse
|
42
|
Morales-Núñez JJ, García-Chagollán M, Muñoz-Valle JF, Díaz-Pérez SA, Torres-Hernández PC, Rodríguez-Reyes SC, Santoscoy-Ascencio G, Sierra García de Quevedo JJ, Hernández-Bello J. Differences in B-Cell Immunophenotypes and Neutralizing Antibodies Against SARS-CoV-2 After Administration of BNT162b2 (Pfizer-BioNTech) Vaccine in Individuals with and without Prior COVID-19 - A Prospective Cohort Study. J Inflamm Res 2022; 15:4449-4466. [PMID: 35958186 PMCID: PMC9361858 DOI: 10.2147/jir.s374304] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 07/30/2022] [Indexed: 12/15/2022] Open
Abstract
Purpose Understanding the humoral immune response dynamics carried out by B cells in COVID-19 vaccination is little explored; therefore, we analyze the changes induced in the different cellular subpopulations of B cells after vaccination with BNT162b2 (Pfizer-BioNTech). Methods This prospective cohort study evaluated thirty-nine immunized health workers (22 with prior COVID-19 and 17 without prior COVID-19) and ten subjects not vaccinated against SARS-CoV-2 (control group). B cell subpopulations (transitional, mature, naïve, memory, plasmablasts, early plasmablast, and double-negative B cells) and neutralizing antibody levels were analyzed and quantified by flow cytometry and ELISA, respectively. Results The dynamics of the B cells subpopulations after vaccination showed the following pattern: the percentage of transitional B cells was higher in the prior COVID-19 group (p < 0.05), whereas virgin B cells were more prevalent in the group without prior COVID-19 (p < 0.05), mature B cells predominated in both vaccinated groups (p < 0.01), and memory B cells, plasmablasts, early plasmablasts, and double-negative B cells were higher in the not vaccinated group (p < 0.05). Conclusion BNT162b2 vaccine induces changes in B cell subpopulations, especially generating plasma cells and producing neutralizing antibodies against SARS-CoV-2. However, the previous infection with SARS-CoV-2 does not significantly alter the dynamics of these subpopulations but induces more rapid and optimal antibody production.
Collapse
Affiliation(s)
- José Javier Morales-Núñez
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Mariel García-Chagollán
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - José Francisco Muñoz-Valle
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Saúl Alberto Díaz-Pérez
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | | | - Saraí Citlalic Rodríguez-Reyes
- Institute of Translational Nutrigenetics and Nutrigenomics, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
| | | | | | - Jorge Hernández-Bello
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, Mexico
- Correspondence: Jorge Hernández-Bello,s Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara, Jalisco, 44340, Mexico, Tel +52 3334509355, Email
| |
Collapse
|
43
|
Anti-TNFα Treatment Impairs Long-Term Immune Responses to COVID-19 mRNA Vaccine in Patients with Inflammatory Bowel Diseases. Vaccines (Basel) 2022; 10:vaccines10081186. [PMID: 35893835 PMCID: PMC9330864 DOI: 10.3390/vaccines10081186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 01/02/2023] Open
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 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.
Collapse
|
44
|
Moore T, Hossain R, Doores KJ, Shankar-Hari M, Fear DJ. SARS-CoV-2-Specific Memory B Cell Responses Are Maintained After Recovery from Natural Infection and Postvaccination. Viral Immunol 2022; 35:425-436. [PMID: 35857310 DOI: 10.1089/vim.2022.0013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory coronavirus 2 (SARS-CoV-2), has resulted in major worldwide disruption and loss of life over the last 2 years. Many research studies have shown waning serological SARS-CoV-2-specific IgG antibody titers over time, yet, it is unclear whether these changes are reflected in the potential functional reactivation of SARS-CoV-2 antigen-specific memory B cells (MBC) populations. This is especially true in the contexts of differing COVID-19 disease severity and after vaccination regimens. This study aimed to investigate these by polyclonal in vitro reactivation of MBC populations followed by analysis using SAR-CoV-2 antigen-specific B cell ELISpots and IgG antibody ELISAs. Natural disease-associated differences were investigated in 52 donors who have recovered from COVID-19 with varying disease severity, from asymptomatic to severe COVID-19 disease, accompanied by a longitudinal evaluation in a subset of donors. Overall, these data showed limited disease severity-associated differences between donor groups but did show that COVID-19 serologically positive donors had strong antigen-specific MBC-associated responses. MBC responses were better maintained 6 months after recovery from infection when compared to serological antigen-specific IgG antibody titers. A similar investigation after vaccination using 14 donors showed robust serological antigen-specific antibody responses against spike protein that waned over time. MBC-associated responses against spike protein were also observed but showed less waning over time, indicating maintenance of a protective response 6 months after vaccination. Further research is required to evaluate these putatively functional SARS-CoV-2-specific responses in the context of long-term protection mediated by vaccination against this pathogen.
Collapse
Affiliation(s)
- Tom Moore
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, United Kingdom
| | - Rojony Hossain
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, United Kingdom
| | - Katie J Doores
- Department of Infectious Diseases, School of Immunology & Microbial Sciences, King's College London, United Kingdom
| | - Manu Shankar-Hari
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, United Kingdom.,The Queen's Medical Research Institute, Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom.,Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, United Kingdom
| | - David J Fear
- MRC & Asthma UK Centre in Allergic Mechanisms of Asthma, Peter Gorer Department of Immunobiology, School of Immunology & Microbial Sciences, King's College London, United Kingdom
| |
Collapse
|
45
|
Torresi J, Edeling MA, Nolan T, Godfrey DI. A Complementary Union of SARS-CoV2 Natural and Vaccine Induced Immune Responses. Front Immunol 2022; 13:914167. [PMID: 35911696 PMCID: PMC9326230 DOI: 10.3389/fimmu.2022.914167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/13/2022] [Indexed: 12/27/2022] Open
Abstract
Our understanding of the immune responses that follow SARS-CoV-2 infection and vaccination has progressed considerably since the COVID-19 pandemic was first declared on the 11th of March in 2020. Recovery from infection is associated with the development of protective immune responses, although over time these become less effective against new emerging SARS-CoV-2 variants. Consequently, reinfection with SARS-CoV-2 variants is not infrequent and has contributed to the ongoing pandemic. COVID-19 vaccines have had a tremendous impact on reducing infection and particularly the number of deaths associated with SARS-CoV-2 infection. However, waning of vaccine induced immunity plus the emergence of new variants has necessitated the use of boosters to maintain the benefits of vaccination in reducing COVID-19 associated deaths. Boosting is also beneficial for individuals who have recovered from COVID-19 and developed natural immunity, also enhancing responses immune responses to SARS-CoV-2 variants. This review summarizes our understanding of the immune responses that follow SARS-CoV-2 infection and vaccination, the risks of reinfection with emerging variants and the very important protective role vaccine boosting plays in both vaccinated and previously infected individuals.
Collapse
Affiliation(s)
- Joseph Torresi
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Melissa A. Edeling
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| | - Terry Nolan
- Department of Infectious Diseases, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
- Murdoch Children’s Research Institute, Parkville, VIC, Australia
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
46
|
Notarte KI, Guerrero‐Arguero I, Velasco JV, Ver AT, Santos de Oliveira MH, Catahay JA, Khan MSR, Pastrana A, Juszczyk G, Torrelles JB, Lippi G, Martinez‐Sobrido L, Henry BM. Characterization of the significant decline in humoral immune response six months post-SARS-CoV-2 mRNA vaccination: A systematic review. J Med Virol 2022; 94:2939-2961. [PMID: 35229324 PMCID: PMC9088566 DOI: 10.1002/jmv.27688] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/16/2022] [Accepted: 02/27/2022] [Indexed: 11/11/2022]
Abstract
Accumulating evidence shows a progressive decline in the efficacy of coronavirus disease 2019 (COVID-19) (severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2]) messenger RNA (mRNA) vaccines such as Pfizer-BioNTech (mRNA BNT161b2) and Moderna (mRNA-1273) in preventing breakthrough infections due to diminishing humoral immunity over time. Thus, this review characterizes the kinetics of anti-SARS-CoV-2 antibodies after the second dose of a primary cycle of COVID-19 mRNA vaccination. A systematic search of the literature was performed and a total of 18 articles (N = 15 980 participants) were identified and reviewed. The percent difference of means of reported antibody titers was then calculated to determine the decline in humoral response after the peak levels postvaccination. Findings revealed that the peak humoral response was reached at 21-28 days after the second dose, after which serum levels progressively diminished at 4-6-month postvaccination. Additionally, results showed that regardless of age, sex, serostatus, and presence of comorbidities, longitudinal data reporting antibody measurement exhibited a decline of both anti-receptor binding domain immunoglobulin G (IgG) and anti-spike IgG, ranging from 94% to 95% at 90-180 days and 55%-85% at 140-160 days, respectively, after the peak antibody response. This suggests that the rate of antibody decline may be independent of patient-related factors and peak antibody titers but mainly a function of time and antibody class/molecular target. Hence, this study highlights the necessity of more efficient vaccination strategies to provide booster administration in attenuating the effects of waning immunity, especially in the appearance of new variants of concerns.
Collapse
Affiliation(s)
- Kin Israel Notarte
- Faculty of Medicine and SurgeryUniversity of Santo TomasManilaPhilippines
| | - Israel Guerrero‐Arguero
- Disease Intervention & Prevention and Population Health ProgramsTexas Biomedical Research InstituteSan AntonioTexasUSA
| | | | | | | | | | - Md. Siddiqur Rahman Khan
- Disease Intervention & Prevention and Population Health ProgramsTexas Biomedical Research InstituteSan AntonioTexasUSA
| | - Adriel Pastrana
- Faculty of Medicine and SurgeryUniversity of Santo TomasManilaPhilippines
| | | | - Jordi B. Torrelles
- Disease Intervention & Prevention and Population Health ProgramsTexas Biomedical Research InstituteSan AntonioTexasUSA
| | - Giuseppe Lippi
- Section of Clinical BiochemistryUniversity of VeronaVeronaItaly
| | - Luis Martinez‐Sobrido
- Disease Intervention & Prevention and Population Health ProgramsTexas Biomedical Research InstituteSan AntonioTexasUSA
| | - Brandon Michael Henry
- Disease Intervention & Prevention and Population Health ProgramsTexas Biomedical Research InstituteSan AntonioTexasUSA
| |
Collapse
|
47
|
Astakhova EA, Byazrova MG, Yusubalieva GM, Kulemzin SV, Kruglova NA, Prilipov AG, Baklaushev VP, Gorchakov AA, Taranin AV, Filatov AV. Functional Profiling of In Vitro Reactivated Memory B Cells Following Natural SARS-CoV-2 Infection and Gam-COVID-Vac Vaccination. Cells 2022; 11:1991. [PMID: 35805076 PMCID: PMC9265778 DOI: 10.3390/cells11131991] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
Both SARS-CoV-2 infection and vaccination have previously been demonstrated to elicit robust, yet somewhat limited immunity against the evolving variants of SARS-CoV-2. Nevertheless, reports performing side-by-side comparison of immune responses following infection vs. vaccination have been relatively scarce. The aim of this study was to compare B-cell response to adenovirus-vectored vaccination in SARS-CoV-2-naive individuals with that observed in the COVID-19 convalescent patients six months after the first encounter with the viral antigens. We set out to use a single analytical platform and performed comprehensive analysis of serum levels of receptor binding domain (RBD)-specific and virus-neutralizing antibodies, frequencies of RBD-binding circulating memory B cells (MBCs), MBC-derived antibody-secreting cells, as well as RBD-specific and virus-neutralizing activity of MBC-derived antibodies after Gam-COVID-Vac (Sputnik V) vaccination and/or natural SARS-CoV-2 infection. Overall, natural immunity was superior to Gam-COVID-Vac vaccination. The levels of neutralizing MBC-derived antibodies in the convalescent patients turned out to be significantly higher than those found following vaccination. Our results suggest that after six months, SARS-CoV-2-specific MBC immunity is more robust in COVID-19 convalescent patients than in Gam-COVID-Vac recipients. Collectively, our data unambiguously indicate that natural immunity outperforms Gam-COVID-Vac-induced immunity six months following recovery/vaccination, which should inform healthcare and vaccination decisions.
Collapse
Affiliation(s)
- Ekaterina A. Astakhova
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, 115522 Moscow, Russia; (E.A.A.); (M.G.B.); (A.G.P.)
- Department of Immunology, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| | - Maria G. Byazrova
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, 115522 Moscow, Russia; (E.A.A.); (M.G.B.); (A.G.P.)
- Department of Immunology, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
- Department of Immunology, Institute of Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Laboratory of Cell Technology, Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies of the FMBA of Russia, 115682 Moscow, Russia; (G.M.Y.); (V.P.B.)
| | - Sergey V. Kulemzin
- Laboratory of Immunogenetics, Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (S.V.K.); (A.A.G.); (A.V.T.)
| | - Natalia A. Kruglova
- Laboratory of Gene Therapy of Socially Significant Diseases, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology of the Russian Academy of Sciences, 119334 Moscow, Russia;
| | - Alexey G. Prilipov
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, 115522 Moscow, Russia; (E.A.A.); (M.G.B.); (A.G.P.)
- Laboratory of Molecular Genetics, N.F. Gamaleya National Research Center for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, 123098 Moscow, Russia
| | - Vladimir P. Baklaushev
- Laboratory of Cell Technology, Federal Research and Clinical Center for Specialized Types of Medical Care and Medical Technologies of the FMBA of Russia, 115682 Moscow, Russia; (G.M.Y.); (V.P.B.)
| | - Andrey A. Gorchakov
- Laboratory of Immunogenetics, Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (S.V.K.); (A.A.G.); (A.V.T.)
| | - Alexander V. Taranin
- Laboratory of Immunogenetics, Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia; (S.V.K.); (A.A.G.); (A.V.T.)
| | - Alexander V. Filatov
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, 115522 Moscow, Russia; (E.A.A.); (M.G.B.); (A.G.P.)
- Department of Immunology, Faculty of Biology, Lomonosov Moscow State University, 119234 Moscow, Russia
| |
Collapse
|
48
|
Zavaglio F, Cassaniti I, Sammartino JC, Tonello S, Sainaghi PP, Novelli V, Meloni F, Lilleri D, Baldanti F. mRNA BNT162b Vaccine Elicited Higher Antibody and CD4 + T-Cell Responses than Patients with Mild COVID-19. Microorganisms 2022; 10:microorganisms10061250. [PMID: 35744768 PMCID: PMC9228401 DOI: 10.3390/microorganisms10061250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/13/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022] Open
Abstract
We compared the development and persistence of antibody and T-cell responses elicited by the mRNA BNT162b2 vaccine or SARS-CoV-2 infection. We analysed 37 post-COVID-19 patients (15 with pneumonia and 22 with mild symptoms) and 20 vaccinated subjects. Anti-Spike IgG and neutralising antibodies were higher in vaccinated subjects and in patients with pneumonia than in patients with mild COVID-19, and persisted at higher levels in patients with pneumonia while declining in vaccinated subjects. However, the booster dose restored the initial antibody levels. The proliferative CD4+ T-cell response was similar in vaccinated subjects and patients with pneumonia, but was lower in mild COVID-19 patients and persisted in both vaccinated subjects and post-COVID patients. Instead, the proliferative CD8+ T-cell response was lower in vaccinated subjects than in patients with pneumonia, decreased six months after vaccination, and was not restored after the booster dose. The cytokine profile was mainly TH1 in both vaccinated subjects and post-COVID-19 patients. The mRNA BNT162b2 vaccine elicited higher levels of antibody and CD4+ T-cell responses than those observed in mild COVID-19 patients. While the antibody response declined after six months and required a booster dose to be restored at the initial levels, the proliferative CD4+ T-cell response persisted over time.
Collapse
Affiliation(s)
- Federica Zavaglio
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (F.Z.); (I.C.); (J.C.S.); (F.B.)
| | - Irene Cassaniti
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (F.Z.); (I.C.); (J.C.S.); (F.B.)
| | - Josè Camilla Sammartino
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (F.Z.); (I.C.); (J.C.S.); (F.B.)
| | - Stelvio Tonello
- Immunoreumatology Laboratory, Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (P.P.S.)
- Internal Medicine Laboratory, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Pier Paolo Sainaghi
- Immunoreumatology Laboratory, Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Piemonte Orientale, 28100 Novara, Italy; (S.T.); (P.P.S.)
- Internal Medicine Laboratory, Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
- Immunorheumatology Unit, Division of Internal Medicine, “Maggiore della Carità” Univerisity Hospital, 28100 Novara, Italy
| | - Viola Novelli
- Medical Direction, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Federica Meloni
- Research Laboratory of Lung Diseases, Section of Cell Biology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Daniele Lilleri
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (F.Z.); (I.C.); (J.C.S.); (F.B.)
- Correspondence: ; Tel.: +39-0382-501501
| | - Fausto Baldanti
- Microbiology and Virology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (F.Z.); (I.C.); (J.C.S.); (F.B.)
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
49
|
Calcoen B, Callewaert N, Vandenbulcke A, Kerstens W, Imbrechts M, Vercruysse T, Dallmeier K, Van Weyenbergh J, Maes P, Bossuyt X, Zapf D, Dieckmann K, Callebaut K, Thibaut HJ, Vanhoorelbeke K, De Meyer SF, Maes W, Geukens N. High Incidence of SARS-CoV-2 Variant of Concern Breakthrough Infections Despite Residual Humoral and Cellular Immunity Induced by BNT162b2 Vaccination in Healthcare Workers: A Long-Term Follow-Up Study in Belgium. Viruses 2022; 14:1257. [PMID: 35746728 PMCID: PMC9228150 DOI: 10.3390/v14061257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 11/26/2022] Open
Abstract
To mitigate the massive COVID-19 burden caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), several vaccination campaigns were initiated. We performed a single-center observational trial to monitor the mid- (3 months) and long-term (10 months) adaptive immune response and to document breakthrough infections (BTI) in healthcare workers (n = 84) upon BNT162b2 vaccination in a real-world setting. Firstly, serology was determined through immunoassays. Secondly, antibody functionality was analyzed via in vitro binding inhibition and pseudovirus neutralization and circulating receptor-binding domain (RBD)-specific B cells were assessed. Moreover, the induction of SARS-CoV-2-specific T cells was investigated by an interferon-γ release assay combined with flowcytometric profiling of activated CD4+ and CD8+ T cells. Within individuals that did not experience BTI (n = 62), vaccine-induced humoral and cellular immune responses were not correlated. Interestingly, waning over time was more pronounced within humoral compared to cellular immunity. In particular, 45 of these 62 subjects no longer displayed functional neutralization against the delta variant of concern (VoC) at long-term follow-up. Noteworthily, we reported a high incidence of symptomatic BTI cases (17.11%) caused by alpha and delta VoCs, although vaccine-induced immunity was only slightly reduced compared to subjects without BTI at mid-term follow-up.
Collapse
Affiliation(s)
- Bas Calcoen
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, 8500 Kortrijk, Belgium; (B.C.); (A.V.); (K.V.); (S.F.D.M.)
| | - Nico Callewaert
- AZ Groeninge Hospital, Department of Laboratory Medicine, 8500 Kortrijk, Belgium; (K.C.); (N.C.)
| | - Aline Vandenbulcke
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, 8500 Kortrijk, Belgium; (B.C.); (A.V.); (K.V.); (S.F.D.M.)
| | - Winnie Kerstens
- Laboratory of Virology and Chemotherapy, Translational Platform Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, KU Leuven Rega Institute, 3000 Leuven, Belgium; (W.K.); (T.V.); (H.J.T.)
| | - Maya Imbrechts
- PharmAbs, the KU Leuven Antibody Center, KU Leuven, 3000 Leuven, Belgium; (M.I.); (N.G.)
| | - Thomas Vercruysse
- Laboratory of Virology and Chemotherapy, Translational Platform Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, KU Leuven Rega Institute, 3000 Leuven, Belgium; (W.K.); (T.V.); (H.J.T.)
| | - Kai Dallmeier
- Laboratory of Virology, Molecular Vaccinology and Vaccine Discovery, Department of Microbiology, Immunology and Transplantation, KU Leuven Rega Institute, 3000 Leuven, Belgium;
| | - Johan Van Weyenbergh
- Laboratory for Clinical and Epidemiological Virology, KU Leuven Rega Institute, 3000 Leuven, Belgium; (J.V.W.); (P.M.)
| | - Piet Maes
- Laboratory for Clinical and Epidemiological Virology, KU Leuven Rega Institute, 3000 Leuven, Belgium; (J.V.W.); (P.M.)
| | - Xavier Bossuyt
- Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium;
- Department of Laboratory Medicine, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Dorinja Zapf
- Institut für Experimentelle Immunologie, EUROIMMUN Medizinische Labordiagnostika AG, 23552 Lübeck, Germany; (D.Z.); (K.D.)
| | - Kersten Dieckmann
- Institut für Experimentelle Immunologie, EUROIMMUN Medizinische Labordiagnostika AG, 23552 Lübeck, Germany; (D.Z.); (K.D.)
| | - Kim Callebaut
- AZ Groeninge Hospital, Department of Laboratory Medicine, 8500 Kortrijk, Belgium; (K.C.); (N.C.)
| | - Hendrik Jan Thibaut
- Laboratory of Virology and Chemotherapy, Translational Platform Virology and Chemotherapy, Department of Microbiology, Immunology and Transplantation, KU Leuven Rega Institute, 3000 Leuven, Belgium; (W.K.); (T.V.); (H.J.T.)
| | - Karen Vanhoorelbeke
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, 8500 Kortrijk, Belgium; (B.C.); (A.V.); (K.V.); (S.F.D.M.)
- PharmAbs, the KU Leuven Antibody Center, KU Leuven, 3000 Leuven, Belgium; (M.I.); (N.G.)
| | - Simon F. De Meyer
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, 8500 Kortrijk, Belgium; (B.C.); (A.V.); (K.V.); (S.F.D.M.)
| | - Wim Maes
- PharmAbs, the KU Leuven Antibody Center, KU Leuven, 3000 Leuven, Belgium; (M.I.); (N.G.)
| | - Nick Geukens
- PharmAbs, the KU Leuven Antibody Center, KU Leuven, 3000 Leuven, Belgium; (M.I.); (N.G.)
| |
Collapse
|
50
|
Guerrini G, Magrì D, Gioria S, Medaglini D, Calzolai L. Characterization of nanoparticles-based vaccines for COVID-19. NATURE NANOTECHNOLOGY 2022; 17:570-576. [PMID: 35710950 DOI: 10.1038/s41565-022-01129-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 03/23/2022] [Indexed: 06/15/2023]
Abstract
Several vaccines against COVID-19 use nanoparticles to protect the antigen cargo (either proteins or nucleic acids), increase the immunogenicity and ultimately the efficacy. The characterization of these nanomedicines is challenging due to their intrinsic complexity and requires the use of multidisciplinary techniques and competencies. The accurate characterization of nanovaccines can be conceptualized as a combination of physicochemical, immunological and toxicological assays. This will help to address key challenges in the preclinical characterization, will guide the rapid development of safe and effective vaccines for current and future health crises, and will streamline the regulatory process.
Collapse
Affiliation(s)
| | - Davide Magrì
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Sabrina Gioria
- European Commission, Joint Research Centre (JRC), Ispra, Italy
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Medical Biotechnologies, University of Siena, Siena, Italy.
| | - Luigi Calzolai
- European Commission, Joint Research Centre (JRC), Ispra, Italy.
| |
Collapse
|