1
|
Hsieh MJ, Tsai PH, Chiang PH, Kao ZK, Zhuang ZQ, Hsieh AR, Ho HL, Chiou SH, Liang KH, Chen YC. Genomic insights into mRNA COVID-19 vaccines efficacy: Linking genetic polymorphisms to waning immunity. Hum Vaccin Immunother 2024; 20:2399382. [PMID: 39254005 PMCID: PMC11404610 DOI: 10.1080/21645515.2024.2399382] [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: 06/25/2024] [Revised: 08/13/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024] Open
Abstract
Genetic polymorphisms have been linked to the differential waning of vaccine-induced immunity against COVID-19 following vaccination. Despite this, evidence on the mechanisms behind this waning and its implications for vaccination policy remains limited. We hypothesize that specific gene variants may modulate the development of vaccine-initiated immunity, leading to impaired immune function. This study investigates genetic determinants influencing the sustainability of immunity post-mRNA vaccination through a genome-wide association study (GWAS). Utilizing a hospital-based, test negative case-control design, we enrolled 1,119 participants from the Taiwan Precision Medicine Initiative (TPMI) cohort, all of whom completed a full mRNA COVID-19 vaccination regimen and underwent PCR testing during the Omicron outbreak. Participants were classified into breakthrough and protected groups based on PCR results. Genetic samples were analyzed using SNP arrays with rigorous quality control. Cox regression identified significant single nucleotide polymorphisms (SNPs) associated with breakthrough infections, affecting 743 genes involved in processes such as antigenic protein translation, B cell activation, and T cell function. Key genes identified include CD247, TRPV1, MYH9, CCL16, and RPTOR, which are vital for immune responses. Polygenic risk score (PRS) analysis revealed that individuals with higher PRS are at greater risk of breakthrough infections post-vaccination, demonstrating a high predictability (AUC = 0.787) in validating population. This finding confirms the significant influence of genetic variations on the durability of immune responses and vaccine effectiveness. This study highlights the importance of considering genetic polymorphisms in evaluating vaccine-induced immunity and proposes potential personalized vaccination strategies by tailoring regimens to individual genetic profiles.
Collapse
Affiliation(s)
- Min-Jia Hsieh
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ping-Hsing Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Pin-Hsuan Chiang
- Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Zih-Kai Kao
- Department of Information Management, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Zi-Qing Zhuang
- Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ai-Ru Hsieh
- Department of Statistics, Tamkang University, New Taipei, Taiwan
| | - Hsiang-Ling Ho
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- School of medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kung-Hao Liang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Biosafety level 3 laboratory, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chun Chen
- Department of Family Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Big Data Center, Taipei Veterans General Hospital, Taipei, Taiwan
- School of medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Family Medicine, Taipei Veterans General Hospital Yuli Branch, Hualien, Taiwan
| |
Collapse
|
2
|
Takahashi Y, Tanaka H, Taniguchi C, Ogata T. Efficiency of indirect protection of COVID-19 vaccination and interactions between indirect and direct protection on household transmission. Vaccine 2024; 42:126110. [PMID: 39019654 DOI: 10.1016/j.vaccine.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/20/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND There is little information on relationships between indirect and direct protection by COVID-19 vaccination on close contacts of the vaccinees. Here, we assessed effect modification of direct-indirect action influencing the protective effects of vaccination. METHODS Secondary attack rates (SARs) in household contacts (n = 2422) depending on vaccination status of the index cases (n = 1112) with known vaccination history during the delta variant-dominant period (August 2-November 2, 2021) in two public health jurisdictions were calculated using multivariable logistic regression analysis to assess indirect protection by COVID-19 vaccination as adjusted odds ratios (aORs) for SARs. The impact of the time of index case vaccination on indirect-direct protective effects was also assessed. FINDINGS Contacts of index cases receiving 2× COVID-19 vaccinations showed significantly lower SARs than contacts of unvaccinated index cases (aOR:0.48, 95 %CI = 0.32-0.74). Relative to contacts where neither index cases nor contacts themselves were vaccinated (0,0), those with (2,0), (0,2) and (2,2) had lower SARs (0.45, 95 %CI = 0.24-0.82, 0.24, 95 %CI = 0.17-0.032, 0.11, 95 %CI = 0.06-0.20, respectively. No significant interactions on the SARs regarding times of vaccination between index cases and household contacts were observed, indicating additive but not synergistic protection. INTERPRETATION The indirect protective effects of COVID-19 vaccination were attributed to an additive effect together with the direct effect on onward transmission in the household setting. These findings emphasize the importance of herd immunity by COVID-19 vaccination not only for unvaccinated but also vaccinated individuals.
Collapse
Affiliation(s)
- Yuki Takahashi
- Health and Medical General Affairs Division of Osaka Prefectural Government, Japan.
| | - Hideo Tanaka
- Neyagawa City Public Health Center, 28-3 Yasakacho, Neyagawa, Osaka, Japan.
| | - Chie Taniguchi
- College of Nursing, Aichi Medical University, 1-1 Yazakokarimata, Nagakute, Aichi, Japan.
| | - Tsuyoshi Ogata
- Itako Public Health Center of Ibaraki Prefectural Government, 1446-1 Osu, Itako, Ibaraki, Japan.
| |
Collapse
|
3
|
Richards NE, Ailsworth SM, Workman LJ, Bortz PS, Patel J, MacCallum M, Canderan G, Murphy D, Muehling LM, McGowan EC, Woodfolk JA, Kadl A, Platts-Mills TAE, Wilson JM. Mammalian Meat Allergy and IgE to Alpha-Gal in Central Virginia: Findings From a COVID-19 Vaccine and Patient Cohort. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:2817-2825.e2. [PMID: 38944197 DOI: 10.1016/j.jaip.2024.06.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/01/2024]
Abstract
BACKGROUND IgE to galactose-alpha-1,3-galactose (alpha-gal) is linked to tick bites and an important cause of anaphylaxis and urticarial reactions to mammalian meat. The alpha-gal syndrome (AGS) is recognized as being common in the southeastern United States. However, prevalence studies are lacking and open questions remain about risk factors and clinical presentation of alpha-gal sensitization. OBJECTIVE Here we characterized the prevalence as well as the presentation and risk factors of AGS and alpha-gal IgE sensitization in adults in central Virginia recruited without regard to the history of allergic disease. METHODS Adults in central Virginia, primarily University of Virginia Health employees, were recruited as part of a COVID-19 vaccine study. Subjects provided at least one blood sample and answered questionnaires about medical and dietary history. We used ImmunoCAP for IgE assays and assessed the ABO blood group by reverse typing using stored serum. We also investigated biobanked serum from COVID-19 patients. RESULTS Median age of the 267 enrollees was 42 years, 76% were female, and 43 (16%) were sensitized to alpha-gal (cutoff of 0.1 IU/mL), of which mammalian meat allergy was reported by seven (2.6%). Sensitized subjects (1) were older, (2) had higher total IgE levels but a similar frequency of IgE to common respiratory allergens, and (3) were more likely to report tick bites than were nonsensitized subjects. Among those who were sensitized, alpha-gal IgE levels were higher among meat-allergic than nonallergic subjects (geometric mean, 9.0 vs 0.5 IU/mL; P < .001). Mammalian meat and dairy consumption was common in individuals with low-level sensitization. CONCLUSION In central Virginia, AGS is a dominant cause of adult food allergy with a prevalence approaching or exceeding 2%.
Collapse
Affiliation(s)
- Nathan E Richards
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Va
| | - Samuel M Ailsworth
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Va
| | - Lisa J Workman
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Va
| | - Pamela Schoppee Bortz
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Va
| | - Jaimin Patel
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Va
| | - Matthew MacCallum
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Va
| | - Glenda Canderan
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Va
| | - Deborah Murphy
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Va
| | - Lyndsey M Muehling
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Va
| | - Emily C McGowan
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Va
| | - Judith A Woodfolk
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Va
| | - Alexandra Kadl
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Virginia, Charlottesville, Va; Department of Pharmacology, University of Virginia, Charlottesville, Va
| | - Thomas A E Platts-Mills
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Va
| | - Jeffrey M Wilson
- Division of Allergy and Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Va.
| |
Collapse
|
4
|
Martens B, Van Caeseele P, Bullard J, Loeppky C, Wei Y, Reimer J, McKinnon LR, Shaw SY, Kindrachuk J, Stein DR. A Population-Based Study of SARS-CoV-2 IgG Antibody Responses to Vaccination in Manitoba. Vaccines (Basel) 2024; 12:1095. [PMID: 39460263 PMCID: PMC11511381 DOI: 10.3390/vaccines12101095] [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: 04/29/2024] [Revised: 07/10/2024] [Accepted: 07/22/2024] [Indexed: 10/28/2024] Open
Abstract
Understanding variables that influence antibody responses to COVID-19 vaccination within a population can provide valuable information on future vaccination strategies. In this population-based study, we examined the antibody responses to COVID-19 vaccination in Manitoba using residual serum specimens collected between January 2021 and March 2022 (n = 20,365). Samples were tested for spike and nucleocapsid IgG against SARS-CoV-2 using clinically validated assays. We assessed the impacts of multiple factors on post-vaccination antibody titres including type of vaccine, age, sex, geographic location, number of doses received, and timing of vaccination. Our investigation demonstrated that vaccination with one dose of Moderna mRNA-1273 elicited higher anti-spike IgG titres overall compared to Pfizer BNT162b2 vaccination, while one dose of Pfizer BNT162b2 followed by a second dose of Moderna mRNA-1273 exhibited higher titres than two doses of Pfizer BNT162b2 or Moderna mRNA-1273, irrespective of age. Age and time post-vaccination had considerable effects on antibody responses, with older age groups exhibiting lower anti-spike IgG titres than younger ages, and titres of those vaccinated with Pfizer BNT162b2 waning faster than those vaccinated with Moderna mRNA-1273 or a combination of Pfizer BNT162b2 and Moderna mRNA-1273. Antibody titres did not appear to be affected by sex or geographic location. Our results identify how factors such as age and type of vaccine can influence antibody responses to vaccination, and how antibody titres wane over time. This information highlights the importance of tailoring vaccine regimens to specific populations, especially those at increased risk of severe COVID-19 and can be used to inform future vaccination strategies, scheduling of booster doses, and public health measures.
Collapse
Affiliation(s)
- Brielle Martens
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Paul Van Caeseele
- Department of Pediatrics and Child Health, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3A 1S1, Canada;
- Cadham Provincial Laboratory, Winnipeg, MB R3E 3J7, Canada
| | - Jared Bullard
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Cadham Provincial Laboratory, Winnipeg, MB R3E 3J7, Canada
| | - Carla Loeppky
- Epidemiology & Surveillance, Manitoba Health, Seniors and Active Living, Winnipeg, MB R3B 3M9, Canada
| | - Yichun Wei
- Epidemiology & Surveillance, Manitoba Health, Seniors and Active Living, Winnipeg, MB R3B 3M9, Canada
| | - Joss Reimer
- Winnipeg Regional Health Authority, Winnipeg, MB R3B 1E2, Canada
| | - Lyle R. McKinnon
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban 4001, South Africa
- Department of Medical Microbiology and Immunology, University of Nairobi, Nairobi P.O. Box 19676 00202, Kenya
| | - Souradet Y. Shaw
- Department of Community Health Sciences, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
| | - Jason Kindrachuk
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3E 0W2, Canada
- Manitoba Centre for Proteomics and Systems Biology, Health Science Centre, Winnipeg, Manitoba R3E 3P4, Canada
| | - Derek R. Stein
- Department of Medical Microbiology & Infectious Diseases, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Cadham Provincial Laboratory, Winnipeg, MB R3E 3J7, Canada
| |
Collapse
|
5
|
Bullock JL, Hickey TE, Kemp TJ, Metz J, Loftus S, Haynesworth K, Castro N, Luke BT, Lowy DR, Pinto LA. Longitudinal Assessment of BNT162b2- and mRNA-1273-Induced Anti-SARS-CoV-2 Spike IgG Levels and Avidity Following Three Doses of Vaccination. Vaccines (Basel) 2024; 12:516. [PMID: 38793767 PMCID: PMC11125776 DOI: 10.3390/vaccines12050516] [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: 03/29/2024] [Revised: 04/24/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
SARS-CoV-2 vaccination-induced protection against infection is likely to be affected by functional antibody features. To understand the kinetics of antibody responses in healthy individuals after primary series and third vaccine doses, sera from the recipients of the two licensed SARS-CoV-2 mRNA vaccines were assessed for circulating anti-SARS-CoV-2 spike IgG levels and avidity for up to 6 months post-primary series and 9 months after the third dose. Following primary series vaccination, anti-SARS-CoV-2 spike IgG levels declined from months 1 to 6, while avidity increased through month 6, irrespective of the vaccine received. The third dose of either vaccine increased anti-SARS-CoV-2 spike IgG levels and avidity and appeared to enhance antibody level persistence-generating a slower rate of decline in the 3 months following the third dose compared to the decline seen after the primary series alone. The third dose of both vaccines induced significant avidity increases 1 month after vaccination compared to the avidity response 6 months post-primary series vaccination (p ≤ 0.001). A significant difference in avidity responses between the two vaccines was observed 6 months post-third dose, where the BNT162b2 recipients had higher antibody avidity levels compared to the mRNA-1273 recipients (p = 0.020).
Collapse
Affiliation(s)
- Jimmie L. Bullock
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (J.L.B.J.); (T.E.H.); (T.J.K.); (J.M.); (S.L.); (K.H.)
| | - Thomas E. Hickey
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (J.L.B.J.); (T.E.H.); (T.J.K.); (J.M.); (S.L.); (K.H.)
| | - Troy J. Kemp
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (J.L.B.J.); (T.E.H.); (T.J.K.); (J.M.); (S.L.); (K.H.)
| | - Jordan Metz
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (J.L.B.J.); (T.E.H.); (T.J.K.); (J.M.); (S.L.); (K.H.)
| | - Sarah Loftus
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (J.L.B.J.); (T.E.H.); (T.J.K.); (J.M.); (S.L.); (K.H.)
| | - Katarzyna Haynesworth
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (J.L.B.J.); (T.E.H.); (T.J.K.); (J.M.); (S.L.); (K.H.)
| | - Nicholas Castro
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (J.L.B.J.); (T.E.H.); (T.J.K.); (J.M.); (S.L.); (K.H.)
| | - Brian T. Luke
- Bioinformatics and Computational Science Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA;
| | - Douglas R. Lowy
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
| | - Ligia A. Pinto
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA; (J.L.B.J.); (T.E.H.); (T.J.K.); (J.M.); (S.L.); (K.H.)
| |
Collapse
|
6
|
Alkema M, Smit MJ, Marin-Mogollon C, Totté K, Teelen K, van Gemert GJ, van de Vegte-Bolmer M, Mordmüller BG, Reimer JM, Lövgren-Bengtsson KL, Sauerwein RW, Bousema T, Plieskatt J, Theisen M, Jore MM, McCall MBB. A Pfs48/45-based vaccine to block Plasmodium falciparum transmission: phase 1, open-label, clinical trial. BMC Med 2024; 22:170. [PMID: 38649867 PMCID: PMC11036667 DOI: 10.1186/s12916-024-03379-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/02/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND The stalling global progress in malaria control highlights the need for novel tools for malaria elimination, including transmission-blocking vaccines. Transmission-blocking vaccines aim to induce human antibodies that block parasite development in the mosquito and mosquitoes becoming infectious. The Pfs48/45 protein is a leading Plasmodium falciparum transmission-blocking vaccine candidate. The R0.6C fusion protein, consisting of Pfs48/45 domain 3 (6C) and the N-terminal region of P. falciparum glutamate-rich protein (R0), has previously been produced in Lactococcus lactis and elicited functional antibodies in rodents. Here, we assess the safety and transmission-reducing efficacy of R0.6C adsorbed to aluminium hydroxide with and without Matrix-M™ adjuvant in humans. METHODS In this first-in-human, open-label clinical trial, malaria-naïve adults, aged 18-55 years, were recruited at the Radboudumc in Nijmegen, the Netherlands. Participants received four intramuscular vaccinations on days 0, 28, 56 and 168 with either 30 µg or 100 µg of R0.6C and were randomised for the allocation of one of the two different adjuvant combinations: aluminium hydroxide alone, or aluminium hydroxide combined with Matrix-M1™ adjuvant. Adverse events were recorded from inclusion until 84 days after the fourth vaccination. Anti-R0.6C and anti-6C IgG titres were measured by enzyme-linked immunosorbent assay. Transmission-reducing activity of participants' serum and purified vaccine-specific immunoglobulin G was assessed by standard membrane feeding assays using laboratory-reared Anopheles stephensi mosquitoes and cultured P. falciparum gametocytes. RESULTS Thirty-one participants completed four vaccinations and were included in the analysis. Administration of all doses was safe and well-tolerated, with one related grade 3 adverse event (transient fever) and no serious adverse events occurring. Anti-R0.6C and anti-6C IgG titres were similar between the 30 and 100 µg R0.6C arms, but higher in Matrix-M1™ arms. Neat participant sera did not induce significant transmission-reducing activity in mosquito feeding experiments, but concentrated vaccine-specific IgGs purified from sera collected two weeks after the fourth vaccination achieved up to 99% transmission-reducing activity. CONCLUSIONS R0.6C/aluminium hydroxide with or without Matrix-M1™ is safe, immunogenic and induces functional Pfs48/45-specific transmission-blocking antibodies, albeit at insufficient serum concentrations to result in transmission reduction by neat serum. Future work should focus on identifying alternative vaccine formulations or regimens that enhance functional antibody responses. TRIAL REGISTRATION The trial is registered with ClinicalTrials.gov under identifier NCT04862416.
Collapse
Affiliation(s)
- M Alkema
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M J Smit
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - C Marin-Mogollon
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - K Totté
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - K Teelen
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - G J van Gemert
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M van de Vegte-Bolmer
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - B G Mordmüller
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | | | - R W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
- Present Address: TropIQ Health Sciences, Nijmegen, the Netherlands
| | - T Bousema
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - J Plieskatt
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - M Theisen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
- Centre for Medical Parasitology at Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - M M Jore
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - M B B McCall
- Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
7
|
Korosec CS, Dick DW, Moyles IR, Watmough J. SARS-CoV-2 booster vaccine dose significantly extends humoral immune response half-life beyond the primary series. Sci Rep 2024; 14:8426. [PMID: 38637521 PMCID: PMC11026522 DOI: 10.1038/s41598-024-58811-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/03/2024] [Indexed: 04/20/2024] Open
Abstract
SARS-CoV-2 lipid nanoparticle mRNA vaccines continue to be administered as the predominant prophylactic measure to reduce COVID-19 disease pathogenesis. Quantifying the kinetics of the secondary immune response from subsequent doses beyond the primary series and understanding how dose-dependent immune waning kinetics vary as a function of age, sex, and various comorbidities remains an important question. We study anti-spike IgG waning kinetics in 152 individuals who received an mRNA-based primary series (first two doses) and a subset of 137 individuals who then received an mRNA-based booster dose. We find the booster dose elicits a 71-84% increase in the median Anti-S half life over that of the primary series. We find the Anti-S half life for both primary series and booster doses decreases with age. However, we stress that although chronological age continues to be a good proxy for vaccine-induced humoral waning, immunosenescence is likely not the mechanism, rather, more likely the mechanism is related to the presence of noncommunicable diseases, which also accumulate with age, that affect immune regulation. We are able to independently reproduce recent observations that those with pre-existing asthma exhibit a stronger primary series humoral response to vaccination than compared to those that do not, and further, we find this result is sustained for the booster dose. Finally, via a single-variate Kruskal-Wallis test we find no difference between male and female humoral decay kinetics, however, a multivariate approach utilizing Least Absolute Shrinkage and Selection Operator (LASSO) regression for feature selection reveals a statistically significant (p < 1 × 10 - 3 ), albeit small, bias in favour of longer-lasting humoral immunity amongst males.
Collapse
Affiliation(s)
- Chapin S Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
| | - David W Dick
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada.
| | - Iain R Moyles
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, M3J 1P3, ON, Canada
| | - James Watmough
- Department of Mathematics and Statistics, University of New Brunswick, 3 Bailey Dr, Fredericton, E3B 5A3, NB, Canada
| |
Collapse
|
8
|
Echeverri Tribin F, Williams E, Testamarck V, Carreño JM, Bielak D, Yellin T, Krammer F, Hoffer M, Pallikkuth S, Pahwa S. Determinants of health as predictors for differential antibody responses following SARS-CoV-2 primary and booster vaccination in an at-risk, longitudinal cohort. PLoS One 2024; 19:e0292566. [PMID: 38564600 PMCID: PMC10987003 DOI: 10.1371/journal.pone.0292566] [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: 09/22/2023] [Accepted: 01/23/2024] [Indexed: 04/04/2024] Open
Abstract
Post vaccine immunity following COVID-19 mRNA vaccination may be driven by extrinsic, or controllable and intrinsic, or inherent health factors. Thus, we investigated the effects of extrinsic and intrinsic on the peak antibody response following COVID-19 primary vaccination and on the trajectory of peak antibody magnitude and durability over time. Participants in a longitudinal cohort attended visits every 3 months for up to 2 years following enrollment. At baseline, participants provided information on their demographics, recreational behaviors, and comorbid health conditions which guided our model selection process. Blood samples were collected for serum processing and spike antibody testing at each visit. Cross-sectional and longitudinal models (linear-mixed effects models) were generated to assess the relationship between selected intrinsic and extrinsic health factors on peak antibody following vaccination and to determine the influence of these predictors on antibody over time. Following cross-sectional analysis, we observed higher peak antibody titers after primary vaccination in females, those who reported recreational drug use, younger age, and prior COVID-19 history. Following booster vaccination, females and Hispanics had higher peak titers after the 3rd and 4th doses, respectively. Longitudinal models demonstrated that Moderna mRNA-1273 recipients, females, and those previously vaccinated had increased peak titers over time. Moreover, drug users and half-dose Moderna mRNA-1273 recipients had higher peak antibody titers over time following the first booster, while no predictive factors significantly affected post-second booster antibody responses. Overall, both intrinsic and extrinsic health factors play a significant role in shaping humoral immunogenicity after initial vaccination and the first booster. The absence of predictive factors for second booster immunogenicity suggests a more robust and consistent immune response after the second booster vaccine administration.
Collapse
Affiliation(s)
- Felipe Echeverri Tribin
- Department of Biomedical Engineering, University of Miami, Miami, Florida, United States of America
- University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Erin Williams
- Department of Biomedical Engineering, University of Miami, Miami, Florida, United States of America
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Valeska Testamarck
- Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Dominika Bielak
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Temima Yellin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States of America
| | - Michael Hoffer
- Department of Biomedical Engineering, University of Miami, Miami, Florida, United States of America
- Department of Neurological Surgery, University of Miami, Miller School of Medicine, Miami, Florida, United States of America
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Savita Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| |
Collapse
|
9
|
Inoue W, Kimura Y, Okamoto S, Nogimori T, Sakaguchi-Mikami A, Yamamoto T, Tsunetsugu-Yokota Y. SARS-CoV-2-Specific Immune Responses in Vaccination and Infection during the Pandemic in 2020-2022. Viruses 2024; 16:446. [PMID: 38543812 PMCID: PMC10974545 DOI: 10.3390/v16030446] [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: 02/01/2024] [Revised: 03/10/2024] [Accepted: 03/11/2024] [Indexed: 05/23/2024] Open
Abstract
To gain insight into how immunity develops against SARS-CoV-2 from 2020 to 2022, we analyzed the immune response of a small group of university staff and students who were either infected or vaccinated. We investigated the levels of receptor-binding domain (RBD)-specific and nucleocapsid (N)-specific IgG and IgA antibodies in serum and saliva samples taken early (around 10 days after infection or vaccination) and later (around 1 month later), as well as N-specific T-cell responses. One patient who had been infected in 2020 developed serum RBD and N-specific IgG antibodies, but declined eight months later, then mRNA vaccination in 2021 produced a higher level of anti-RBD IgG than natural infection. In the vaccination of naïve individuals, vaccines induced anti-RBD IgG, but it declined after six months. A third vaccination boosted the IgG level again, albeit to a lower level than after the second. In 2022, when the Omicron variant became dominant, familial transmission occurred among vaccinated people. In infected individuals, the levels of serum anti-RBD IgG antibodies increased later, while anti-N IgG peaked earlier. The N-specific activated T cells expressing IFN γ or CD107a were detected only early. Although SARS-CoV-2-specific salivary IgA was undetectable, two individuals showed a temporary peak in RBD- and N-specific IgA antibodies in their saliva on the second day after infection. Our study, despite having a small sample size, revealed that SARS-CoV-2 infection triggers the expected immune responses against acute viral infections. Moreover, our findings suggest that the temporary mucosal immune responses induced early during infection may provide better protection than the currently available intramuscular vaccines.
Collapse
Affiliation(s)
- Wakana Inoue
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
| | - Yuta Kimura
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
| | - Shion Okamoto
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
| | - Takuto Nogimori
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; (T.N.); (T.Y.)
| | - Akane Sakaguchi-Mikami
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
| | - Takuya Yamamoto
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; (T.N.); (T.Y.)
- Laboratory of Aging and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka 565-0871, Japan
- Department of Virology and Immunology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | - Yasuko Tsunetsugu-Yokota
- Department of Medical Technology, School of Health Sciences and Graduate School of Medical Technology, Tokyo University of Technology, Tokyo 144-8535, Japan; (W.I.); (Y.K.); (S.O.); (A.S.-M.)
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka 567-0085, Japan; (T.N.); (T.Y.)
- Research Institute, The World New Prosperity (WNP), Tokyo 169-0075, Japan
| |
Collapse
|
10
|
Alonso R, Gil-Manso S, Catalán P, Sánchez-Arcilla I, Marzola M, Correa-Rocha R, Muñoz P, Pion M. Neutralizing antibody levels detected early after mRNA-based vaccination do not predict by themselves subsequent breakthrough infections of SARS-CoV-2. Front Immunol 2024; 15:1341313. [PMID: 38404583 PMCID: PMC10884961 DOI: 10.3389/fimmu.2024.1341313] [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: 11/20/2023] [Accepted: 01/26/2024] [Indexed: 02/27/2024] Open
Abstract
The development of mRNA vaccines represented a significant achievement in response to the global health crisis during the SARS-CoV-2 pandemic. Evaluating vaccine efficacy entails identifying different anti-SARS-CoV-2 antibodies, such as total antibodies against the Receptor Binding Domain (RBD) of the S-protein, or neutralizing antibodies (NAbs). This study utilized an innovative PETIA-based kit to measure NAb, and the investigation aimed to assess whether levels of anti-RBD IgG and NAb uniformly measured 30 days after vaccination could predict individuals at a higher risk of subsequent infection in the months following vaccination. Among a cohort of healthy vaccinated healthcare workers larger than 6,000, 12 mRNA-1273- and 115 BNT162b2-vaccinated individuals contracted infections after the first two doses. The main finding is that neither anti-RBD IgG nor NAb levels measured at day 30 post-vaccination can be used as predictors of breakthrough infections (BI). Therefore, the levels of anti-SARS-CoV-2 antibodies detected shortly after vaccination are not the pivotal factors involved in antiviral protection, and other characteristics must be considered in understanding protection against infection. Furthermore, the levels of anti-RBD and NAbs followed a very similar pattern, with a correlation coefficient of r = 0.96. This robust correlation would justify ceasing the quantification of NAbs, as the information provided by both determinations is highly similar. This optimization would help allocate resources more efficiently and speed up the determination of individuals' humoral immunity status.
Collapse
Affiliation(s)
- Roberto Alonso
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBER (Centro de Investigación Biomédicas en Red) de Enfermedades Respiratorias, CIBERES, Barcelona, Spain
- Department of Medicine, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Sergio Gil-Manso
- Advanced ImmunoRegulation Group, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Pilar Catalán
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBER (Centro de Investigación Biomédicas en Red) de Enfermedades Respiratorias, CIBERES, Barcelona, Spain
| | - Ignacio Sánchez-Arcilla
- Department of Labour Risks Prevention, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Marco Marzola
- Department of Labour Risks Prevention, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBER (Centro de Investigación Biomédicas en Red) de Enfermedades Respiratorias, CIBERES, Barcelona, Spain
- Department of Medicine, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Marjorie Pion
- Advanced ImmunoRegulation Group, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | | |
Collapse
|
11
|
Ciccone EJ, Zhu DR, Gunderson AK, Hawke S, Ajeen R, Lodge EK, Shook-Sa BE, Abernathy H, Garrett HE, King E, Alavian N, Reyes R, Taylor JL, Beatty C, Chung C, Mendoza CE, Weber DJ, Markmann AJ, Premkumar L, Juliano JJ, Boyce RM, Aiello AE. Magnitude and Durability of the Antibody Response to mRNA-Based Vaccination Among SARS-CoV-2 Seronegative and Seropositive Health Care Personnel. Open Forum Infect Dis 2024; 11:ofae009. [PMID: 38293246 PMCID: PMC10826795 DOI: 10.1093/ofid/ofae009] [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: 10/13/2023] [Accepted: 01/04/2024] [Indexed: 02/01/2024] Open
Abstract
Few studies have described changes in SARS-CoV-2 antibody levels in response to infection and vaccination at frequent intervals and over extended follow-up periods. The purpose of this study was to assess changes in SARS-CoV-2-specific antibody responses among a prospective cohort of health care personnel over 18 months with up to 22 samples per person. Antibody levels and live virus neutralization were measured before and after mRNA-based vaccination with results stratified by (1) SARS-CoV-2 infection status prior to initial vaccination and (2) SARS-CoV-2 infection at any point during follow-up. We found that the antibody response to the first dose was almost 2-fold higher in individuals who were seropositive prior to vaccination, although neutralization titers were more variable. The antibody response induced by vaccination appeared to wane over time but generally persisted for 8 to 9 months, and those who were infected at any point during the study had slightly higher antibody levels over time vs those who remained uninfected. These findings underscore the need to account for SARS-CoV-2 natural infection as a modifier of vaccine responses, and they highlight the importance of frequent testing of longitudinal antibody titers over time. Together, our results provide a clearer understanding of the trajectories of antibody response among vaccinated individuals with and without prior SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | - Deanna R Zhu
- Department of Epidemiology, Gillings School of Global Public Health
| | | | - Sam Hawke
- Department of Biostatistics, Gillings School of Global Public Health
| | - Rawan Ajeen
- Institute for Global Health and Infectious Diseases
| | - Evans K Lodge
- Department of Epidemiology, Gillings School of Global Public Health
| | - Bonnie E Shook-Sa
- Department of Biostatistics, Gillings School of Global Public Health
| | | | - Haley E Garrett
- Department of Epidemiology, Gillings School of Global Public Health
| | - Elise King
- Institute for Global Health and Infectious Diseases
| | - Naseem Alavian
- Division of Hospital Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Raquel Reyes
- Division of Hospital Medicine, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | | | - Cherese Beatty
- Department of Epidemiology and Robert N. Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, New York
| | | | - Carmen E Mendoza
- Department of Epidemiology, Gillings School of Global Public Health
| | - David J Weber
- Division of Infectious Diseases, School of Medicine
- Department of Epidemiology, Gillings School of Global Public Health
| | | | - Lakshmanane Premkumar
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Jonathan J Juliano
- Division of Infectious Diseases, School of Medicine
- Department of Epidemiology, Gillings School of Global Public Health
| | - Ross M Boyce
- Division of Infectious Diseases, School of Medicine
- Department of Epidemiology, Gillings School of Global Public Health
| | - Allison E Aiello
- Department of Epidemiology and Robert N. Butler Columbia Aging Center, Mailman School of Public Health, Columbia University, New York, New York
| |
Collapse
|
12
|
Ravindran R, Kang H, McReynolds C, Sanghar GK, Chang WLW, Ramasamy S, Kolloli A, Kumar R, Subbian S, Hammock BD, Hartigan-O’Connor DJ, Ikram A, Haczku A, Khan IH. Dynamics of temporal immune responses in nonhuman primates and humans immunized with COVID-19 vaccines. PLoS One 2023; 18:e0287377. [PMID: 37856429 PMCID: PMC10586671 DOI: 10.1371/journal.pone.0287377] [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: 01/10/2023] [Accepted: 06/04/2023] [Indexed: 10/21/2023] Open
Abstract
We assessed the humoral immune responses to a COVID-19 vaccine in a well-controlled rhesus macaque model compared to humans immunized with two mRNA vaccines over several months post-second dose. The plasma IgG levels against seven coronaviruses (including SARS-CoV-2) and antibody subtypes (IgG 1-4 and IgM) against SARS-CoV-2 were evaluated using multiplex assays. The neutralization capacity of plasma antibodies against the original SAR-CoV-2 isolate and nine variants was evaluated in vaccinated humans and non-human primates. Immunization of macaques and humans with SARS-CoV-2 vaccines induced a robust neutralizing antibody response. In non-SIV-infected adult macaques immunized with an adenoviral vector expressing S-RBD (n = 7) or N protein (n = 3), elevated levels of IgG and neutralizing antibodies were detected 2 weeks post-second dose. Immune responses to the S-RBD vaccine in SIV-infected adult macaques (n = 2) were similar to the non-SIV-infected animals. Adult humans immunized with Pfizer (n = 35) or Moderna (n = 18) vaccines developed IgG and neutralizing antibodies at 4 weeks post-second dose. In both vaccine groups, IgG 1 was the predominant subtype, followed by IgG 3. The IgG levels, including total and IgG 1,2,3 elicited by the Moderna vaccine, were significantly higher than the corresponding levels elicited by the Pfizer vaccine at 4 weeks post-second dose. A significant correlation was observed between the plasma total IgG antibody levels and neutralization titers in both macaques and humans. Furthermore, broad-spectrum neutralization antibodies against several variants of SARS-CoV-2 were detected in the plasma of both macaques and humans after two vaccinations.
Collapse
Affiliation(s)
- Resmi Ravindran
- Department of Pathology and Laboratory Medicine, University of California, Davis, Davis, California, United States of America
| | - Harsharonjit Kang
- Department of Pathology and Laboratory Medicine, University of California, Davis, Davis, California, United States of America
| | - Cindy McReynolds
- Department of Entomology and Nematology, University of California, Davis, Davis, California, United States of America
| | - Gursharan Kaur Sanghar
- Pulmonary, Critical Care and Sleep Medicine, University of California, Davis, Davis, California, United States of America
| | - W. L. William Chang
- California National Primate Research Center, University of California, Davis, Davis, California, United States of America
| | - Santhamani Ramasamy
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Afsal Kolloli
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Ranjeet Kumar
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, New Jersey, United States of America
| | - Bruce D. Hammock
- Department of Entomology and Nematology, University of California, Davis, Davis, California, United States of America
| | - Dennis J. Hartigan-O’Connor
- California National Primate Research Center, University of California, Davis, Davis, California, United States of America
| | - Aamer Ikram
- National Institutes of Health, Islamabad, Pakistan
| | - Angela Haczku
- Pulmonary, Critical Care and Sleep Medicine, University of California, Davis, Davis, California, United States of America
| | - Imran H. Khan
- Department of Pathology and Laboratory Medicine, University of California, Davis, Davis, California, United States of America
| |
Collapse
|
13
|
Arientová S, Matúšková K, Bartoš O, Holub M, Beran O. Specific immune responses after BNT162b2 mRNA vaccination and COVID-19 infection. Front Immunol 2023; 14:1271353. [PMID: 37920457 PMCID: PMC10619853 DOI: 10.3389/fimmu.2023.1271353] [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: 08/02/2023] [Accepted: 09/28/2023] [Indexed: 11/04/2023] Open
Abstract
Although vaccines against COVID-19 are effective tools in preventing severe disease, recent studies have shown enhanced protection after vaccine boosters. The aim of our study was to examine the dynamics and duration of both humoral and cellular immune responses following a three-dose regimen of the BNT162b2 mRNA vaccine. In a longitudinal prospective study we enrolled 86 adults who received the BNT162b2 vaccine, 35 unvaccinated individuals with a history of mild COVID-19 and a control group of 30 healthy SARS-CoV-2 seronegative persons. We assessed the SARS-CoV-2-specific T cell responses and IgG production up to 12 months post the third BNT162b2 dose in 24 subjects. The vaccinated group had significantly higher IgG antibody levels after two doses compared to the convalescent group (p<0.001). After the third dose, IgG levels surged beyond those detected after the second dose (p<0.001). Notably, these elevated IgG levels were maintained 12 months post the third dose. After two doses, specific T cell responses were detected in 87.5% of the vaccinated group. Additionally, there was a significant decrease before the third dose. However, post the third dose, specific T cell responses surged and remained stable up to the 12-month period. Our findings indicate that the BNT162b2 vaccine induces potent and enduring humoral and cellular responses, which are notably enhanced by the third dose and remain persistant without a significant decline a year after the booster. Further research is essential to understand the potential need for subsequent boosters.
Collapse
Affiliation(s)
- Simona Arientová
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czechia
| | - Kateřina Matúšková
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czechia
| | - Oldřich Bartoš
- Military Health Institute, Military Medical Agency, Prague, Czechia
| | - Michal Holub
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czechia
| | - Ondřej Beran
- Department of Infectious Diseases, First Faculty of Medicine, Charles University and Military University Hospital Prague, Prague, Czechia
| |
Collapse
|
14
|
Hou CW, Williams S, Taylor K, Boyle V, Bobbett B, Kouvetakis J, Nguyen K, McDonald A, Harris V, Nussle B, Scharf P, Jehn ML, Lant T, Magee M, Chung Y, LaBaer J, Murugan V. Serological survey to estimate SARS-CoV-2 infection and antibody seroprevalence at a large public university: A cross-sectional study. BMJ Open 2023; 13:e072627. [PMID: 37536960 PMCID: PMC10401225 DOI: 10.1136/bmjopen-2023-072627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/26/2023] [Indexed: 08/05/2023] Open
Abstract
OBJECTIVE This study investigated the seroprevalence of SARS-CoV-2 antibodies among adults over 18 years. DESIGN Prospective cohort study. SETTINGS A large public university. PARTICIPANTS This study took volunteers over 5 days and recruited 1064 adult participants. PRIMARY OUTCOME MEASURES Seroprevalence of SARS-CoV-2-specific antibodies due to previous exposure to SARS-CoV-2 and/or vaccination. RESULTS The seroprevalence of the antireceptor binding domain (RBD) antibody was 90% by a lateral flow assay and 88% by a semiquantitative chemiluminescent immunoassay. The seroprevalence for antinucleocapsid was 20%. In addition, individuals with previous natural COVID-19 infection plus vaccination had higher anti-RBD antibody levels compared with those who had vaccination only or infection only. Individuals who had a breakthrough infection had the highest anti-RBD antibody levels. CONCLUSION Accurate estimates of the cumulative incidence of SARS-CoV-2 infection can inform the development of university risk mitigation protocols such as encouraging booster shots, extending mask mandates or reverting to online classes. It could help us to have clear guidance to act at the first sign of the next surge as well, especially since there is a surge of COVID-19 subvariant infections.
Collapse
Affiliation(s)
- Ching-Wen Hou
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - Stacy Williams
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - Kylee Taylor
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - Veronica Boyle
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - Bradley Bobbett
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - Joseph Kouvetakis
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - Keana Nguyen
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - Aaron McDonald
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - Valerie Harris
- Office of VP Research Development, Arizona State University, Tempe, AZ, USA
| | - Benjamin Nussle
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - Phillip Scharf
- College of Liberal Arts and Sciences, Arizona State University, Tempe, AZ, USA
| | - Megan L Jehn
- School of Human Evolution and Social Change, Arizona State University, Tempe, AZ, USA
| | - Timothy Lant
- Office of VP Research Development, Arizona State University, Tempe, AZ, USA
| | - Mitchell Magee
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - Yunro Chung
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - Joshua LaBaer
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| | - Vel Murugan
- Virginia G. Piper Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, Tempe, Arizona, USA
| |
Collapse
|
15
|
La Civita E, Zannella C, Brusa S, Romano P, Schettino E, Salemi F, Carrano R, Gentile L, Punziano A, Lagnese G, Spadaro G, Franci G, Galdiero M, Terracciano D, Portella G, Loffredo S. BNT162b2 Elicited an Efficient Cell-Mediated Response against SARS-CoV-2 in Kidney Transplant Recipients and Common Variable Immunodeficiency Patients. Viruses 2023; 15:1659. [PMID: 37632002 PMCID: PMC10459971 DOI: 10.3390/v15081659] [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: 07/05/2023] [Revised: 07/27/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
SARS-CoV-2 vaccination is the standard of care for the prevention of COVID-19 disease. Although vaccination triggers both humoral and cellular immune response, COVID-19 vaccination efficacy is currently evaluated by measuring antibodies only, whereas adaptative cellular immunity is unexplored. Our aim is to test humoral and cell-mediated response after three doses of BNT162b vaccine in two cohorts of fragile patients: Common Variable Immunodeficiency (CVID) patients and Kidney Transplant Recipients (KTR) patients compared to healthy donors. We enrolled 10 healthy controls (HCs), 19 CVID patients and 17 KTR patients. HC BNT162b third dose had successfully mounted humoral immune response. A positive correlation between Anti-Spike Trimeric IgG concentration and neutralizing antibody titer was also observed. CVID and KTR groups showed a lower humoral immune response compared to HCs. IFN-γ release induced by epitopes of the Spike protein in stimulated CD4+ and CD8+ T cells was similar among vaccinated HC, CVID and KTR. Patients vaccinated and infected showed a more efficient humoral and cell-mediated response compared to only vaccinated patients. In conclusion, CVID and KTR patients had an efficient cell-mediated but not humoral response to SARS-CoV-2 vaccine, suggesting that the evaluation of T cell responses could be a more sensitive marker of immunization in these subjects.
Collapse
Affiliation(s)
- Evelina La Civita
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (S.B.); (A.P.); (G.L.); (G.S.); (D.T.); (S.L.)
| | - Carla Zannella
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (C.Z.); (M.G.)
- UOC of Virology and Microbiology, University Hospital of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Stefano Brusa
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (S.B.); (A.P.); (G.L.); (G.S.); (D.T.); (S.L.)
| | - Paolo Romano
- Department of Public Health, Section of Nephrology, University of Naples “Federico II”, 80131 Naples, Italy; (P.R.); (E.S.); (F.S.); (R.C.)
| | - Elisa Schettino
- Department of Public Health, Section of Nephrology, University of Naples “Federico II”, 80131 Naples, Italy; (P.R.); (E.S.); (F.S.); (R.C.)
| | - Fabrizio Salemi
- Department of Public Health, Section of Nephrology, University of Naples “Federico II”, 80131 Naples, Italy; (P.R.); (E.S.); (F.S.); (R.C.)
| | - Rosa Carrano
- Department of Public Health, Section of Nephrology, University of Naples “Federico II”, 80131 Naples, Italy; (P.R.); (E.S.); (F.S.); (R.C.)
| | - Luca Gentile
- Integrated Department of Laboratory and Trasfusion Medicine, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Alessandra Punziano
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (S.B.); (A.P.); (G.L.); (G.S.); (D.T.); (S.L.)
| | - Gianluca Lagnese
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (S.B.); (A.P.); (G.L.); (G.S.); (D.T.); (S.L.)
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (S.B.); (A.P.); (G.L.); (G.S.); (D.T.); (S.L.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples “Federico II”, 80131 Naples, Italy
| | - Gianluigi Franci
- Department of Medicine, Surgery and Dentistry “ScholaMedicaSalernitana”, University of Salerno, 84081 Baronissi, Italy;
- Clinical Pathology and Microbiology Unit, San Giovanni di Dio e Ruggi D’Aragona University Hospital, 84125 Salerno, Italy
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (C.Z.); (M.G.)
- UOC of Virology and Microbiology, University Hospital of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (S.B.); (A.P.); (G.L.); (G.S.); (D.T.); (S.L.)
| | - Giuseppe Portella
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (S.B.); (A.P.); (G.L.); (G.S.); (D.T.); (S.L.)
| | - Stefania Loffredo
- Department of Translational Medical Sciences, University of Naples “Federico II”, 80131 Naples, Italy; (E.L.C.); (S.B.); (A.P.); (G.L.); (G.S.); (D.T.); (S.L.)
- Center for Basic and Clinical Immunology Research (CISI), University of Naples “Federico II”, 80131 Naples, Italy
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy
| |
Collapse
|
16
|
Ayers E, Canderan G, Williams ME, Keshavarz B, Portell CA, Wilson JM, Woodfolk JA. In-depth cellular and humoral dynamics of the response to COVID-19 vaccine booster in patients with chronic B-cell neoplasms. Blood Cancer J 2023; 13:114. [PMID: 37495573 PMCID: PMC10372059 DOI: 10.1038/s41408-023-00884-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 06/22/2023] [Accepted: 07/11/2023] [Indexed: 07/28/2023] Open
Affiliation(s)
- Emily Ayers
- Division of Hematology and Oncology and Comprehensive Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, USA.
| | - Glenda Canderan
- Division of Asthma, Allergy and Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA.
| | - Michael E Williams
- Division of Hematology and Oncology and Comprehensive Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Behnam Keshavarz
- Division of Asthma, Allergy and Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Craig A Portell
- Division of Hematology and Oncology and Comprehensive Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jeffrey M Wilson
- Division of Asthma, Allergy and Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Judith A Woodfolk
- Division of Asthma, Allergy and Immunology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
17
|
Takemoto Y, Tanimine N, Yoshinaka H, Tanaka Y, Takafuta T, Sugiyama A, Tanaka J, Ohdan H. Multi-phasic gene profiling using candidate gene approach predict the capacity of specific antibody production and maintenance following COVID-19 vaccination in Japanese population. Front Immunol 2023; 14:1217206. [PMID: 37564647 PMCID: PMC10411726 DOI: 10.3389/fimmu.2023.1217206] [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: 05/05/2023] [Accepted: 07/07/2023] [Indexed: 08/12/2023] Open
Abstract
Background Vaccination against severe acute respiratory syndrome coronavirus type 2 is highly effective in preventing infection and reducing the severity of coronavirus disease (COVID-19). However, acquired humoral immunity wanes within six months. Focusing on the different tempo of acquisition and attenuation of specific antibody titers in individuals, we investigated the impact of genetic polymorphisms on antibody production after COVID-19 vaccination. Methods In total 236 healthcare workers from a Japanese municipal hospital, who received two doses of the vaccine were recruited. We employed a candidate gene approach to identify the target genetic polymorphisms affecting antibody production after vaccination. DNA samples from the study populations were genotyped for 33 polymorphisms in 15 distinct candidate genes encoding proteins involved in antigen-presenting cell activation, T cell activation, T-B interaction, and B cell survival. We measured total anti-SARS-Cov2 spike IgG antibody titers and analyzed the association with genetic polymorphisms at several time points after vaccination using an unbiased statistical method, and stepwise logistic regression following multivariate regression. Results Significant associations were observed between seven SNPs in NLRP3, OAS1, IL12B, CTLA4, and IL4, and antibody titers at 3 weeks after the first vaccination as an initial response. Six SNPs in NLRP3, TNF, OAS1, IL12B, and CTLA4 were associated with high responders with serum antibody titer > 4000 BAU/ml as boosting effect at 3 weeks after the second vaccination. Analysis of long-term maintenance showed the significance of the three SNPs in IL12B, IL7R, and MIF for the maintenance of antibody titers and that in BAFF for attenuation of neutralizing antibodies. Finally, we proposed a predictive model composed of gene profiles to identify the individuals with rapid antibody attenuation by receiver operating characteristic (ROC) analysis (area under the curve (AUC)= 0.76, sensitivity = 82.5%, specificity=67.8%). Conclusions The candidate gene approach successfully showed shifting responsible gene profiles and initial and boosting effect mainly related to the priming phase into antibody maintenance including B cell survival, which traces the phase of immune reactions. These gene profiles provide valuable information for further investigation of humoral immunity against COVID-19 and for building a strategy for personalized vaccine schedules.
Collapse
Affiliation(s)
- Yuki Takemoto
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Naoki Tanimine
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hisaaki Yoshinaka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuka Tanaka
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Toshiro Takafuta
- Department of Internal Medicine, Hiroshima City Funairi Citizens Hospital, Hiroshima, Japan
| | - Aya Sugiyama
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Junko Tanaka
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
18
|
Harthaller T, Falkensammer B, Bante D, Huber M, Schmitt M, Benainouna H, Rössler A, Fleischer V, von Laer D, Kimpel J, Würzner R, Borena W. Retained avidity despite reduced cross-binding and cross-neutralizing antibody levels to Omicron after SARS-COV-2 wild-type infection or mRNA double vaccination. Front Immunol 2023; 14:1196988. [PMID: 37545492 PMCID: PMC10401431 DOI: 10.3389/fimmu.2023.1196988] [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: 03/30/2023] [Accepted: 06/29/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction The rapid evolution of SARS-CoV-2 has posed a challenge to long-lasting immunity against the novel virus. Apart from neutralizing function, binding antibodies induced by vaccination or infection play an important role in containing the infection. Methods To determine the proportion of wild-type (WT)-generated antibodies recognizant of more recent variants, plasma samples from either SARS-CoV-2 WT-infected (n = 336) or double-mRNA (Comirnaty)-vaccinated individuals (n = 354, age and sex matched to the convalescent group) were analyzed for binding antibody capacity against the S1 protein of the BA.1 omicron variant. Results Overall, 38.59% (95% CI, 37.01- 40.20) of WT-generated antibodies recognized Omicron BA.1 S1 protein [28.83% (95% CI, 26.73-30.91) after infection and 43.46% (95% CI, 41.61-45.31) after vaccination; p < 0.001]. Although the proportion of WT-generated binding and neutralizing antibodies also binding to BA.1 is substantially reduced, the avidity of the remaining antibodies against the Omicron variant was non-inferior to that of the ancestral virus: Omicron: 39.7% (95% CI: 38.1-41.3) as compared to the avidity to WT: 27.0% (95% CI, 25.5-28.4), respectively (p < 0.001). Furthermore, we noticed a modestly yet statistically significant higher avidity toward the Omicron epitopes among the vaccinated group (42.2%; 95% CI, 40.51-43.94) as compared to the convalescent counterparts (36.4%; 95% CI, 33.42-38.76) (p = 0.003), even after adjusting for antibody concentration. Discussion Our results suggest that an aspect of functional immunity against the novel strain was considerably retained after WT contact, speculatively counteracting the impact of immune evasion toward neutralization of the strain. Higher antibody levels and cross-binding capacity among vaccinated individuals suggest an advantage of repeated exposure in generating robust immunity.
Collapse
Affiliation(s)
- Teresa Harthaller
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - Barbara Falkensammer
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - David Bante
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - Maria Huber
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - Melanie Schmitt
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - Habib Benainouna
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - Annika Rössler
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - Verena Fleischer
- Department of Hygiene, Microbiology and Public Health, Institute of Hygiene and Medical Microbiology, Innsbruck Medical University, Innsbruck, Austria
| | - Dorothee von Laer
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - Janine Kimpel
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Innsbruck Medical University, Innsbruck, Austria
| | - Reinhard Würzner
- Department of Hygiene, Microbiology and Public Health, Institute of Hygiene and Medical Microbiology, Innsbruck Medical University, Innsbruck, Austria
| | - Wegene Borena
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
19
|
Radziejewska J, Arkowski J, Susło R, Kędzierski K, Wawrzyńska M. Analysis of COVID-19 Incidence and Protective Potential of Persisting IgG Class Antibodies against SARS-CoV-2 Infection in Hospital Staff in Poland. Vaccines (Basel) 2023; 11:1198. [PMID: 37515014 PMCID: PMC10383555 DOI: 10.3390/vaccines11071198] [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: 05/04/2023] [Revised: 06/07/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
The immune responses to both SARS-CoV-2 infection and vaccines are of key importance in prevention efforts. In April and May 2020, 703 study participants tested for COVID-19 by PCR tests were registered. In June and July 2020, they were examined for the presence of SARS-CoV-2 S1/S2 IgG. From October 2020 to January 2021, those among the study population with COVID-19 confirmed by PCR tests were registered, and the same group of participants was invited to be examined again for the presence of SARS-CoV-2 antibodies. In June 2020, antibodies were detected in only 88% of those who had PCR-confirmed COVID-19 in April-May 2020, which suggests that a significant proportion of persons in the Polish population do not produce antibodies after contact with SARS-CoV-2 antigens or rapidly lose them and reach levels below the lab detection limit. The levels of IgG class anti-SARS-CoV-2 antibodies were significantly lower among people who previously had COVID-19 than for those who had received COVID-19 vaccination, which confirms the high immunogenicity of the vaccines against COVID-19 in the Polish population. The study confirms that a detectable level of IgG class anti-SARS-CoV-2 antibodies cannot be considered a reliable marker of the presence and strength of COVID-19 immunity preventing individuals from acquiring SARS-CoV-2 infection.
Collapse
Affiliation(s)
| | - Jacek Arkowski
- Centre for Preclinical Studies, Wrocław Medical University, Ul. Marcinkowskiego 1, 53-220 Wroclaw, Poland
| | - Robert Susło
- Epidemiology and Health Education Unit, Population Health Department, Wroclaw Medical University, Ul. Bujwida 44, 50-345 Wroclaw, Poland
| | - Kamil Kędzierski
- Department of Medical Emergencies, Wroclaw Medical University, Ul. Parkowa 34, 51-616 Wroclaw, Poland
| | - Magdalena Wawrzyńska
- Centre for Preclinical Studies, Wrocław Medical University, Ul. Marcinkowskiego 1, 53-220 Wroclaw, Poland
| |
Collapse
|
20
|
Braeye T, Catteau L, Brondeel R, van Loenhout JAF, Proesmans K, Cornelissen L, Van Oyen H, Stouten V, Hubin P, Billuart M, Djiena A, Mahieu R, Hammami N, Van Cauteren D, Wyndham-Thomas C. Vaccine effectiveness against transmission of alpha, delta and omicron SARS-COV-2-infection, Belgian contact tracing, 2021-2022. Vaccine 2023; 41:3292-3300. [PMID: 37085456 PMCID: PMC10073587 DOI: 10.1016/j.vaccine.2023.03.069] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 03/31/2023] [Accepted: 03/31/2023] [Indexed: 04/08/2023]
Abstract
OBJECTIVES Vaccine effectiveness against transmission (VET) of SARS-CoV-2-infection can be estimated from secondary attack rates observed during contact tracing. We estimated VET, the vaccine-effect on infectiousness of the index case and susceptibility of the high-risk exposure contact (HREC). METHODS We fitted RT-PCR-test results from HREC to immunity status (vaccine schedule, prior infection, time since last immunity-conferring event), age, sex, calendar week of sampling, household, background positivity rate and dominant VOC using a multilevel Bayesian regression-model. We included Belgian data collected between January 2021 and January 2022. RESULTS For primary BNT162b2-vaccination we estimated initial VET at 96% (95%CI 95-97) against Alpha, 87% (95%CI 84-88) against Delta and 31% (95%CI 25-37) against Omicron. Initial VET of booster-vaccination (mRNA primary and booster-vaccination) was 87% (95%CI 86-89) against Delta and 68% (95%CI 65-70) against Omicron. The VET-estimate against Delta and Omicron decreased to 71% (95%CI 64-78) and 55% (95%CI 46-62) respectively, 150-200 days after booster-vaccination. Hybrid immunity, defined as vaccination and documented prior infection, was associated with durable and higher or comparable (by number of antigen exposures) protection against transmission. CONCLUSIONS While we observed VOC-specific immune-escape, especially by Omicron, and waning over time since immunization, vaccination remained associated with a reduced risk of SARS-CoV-2-transmission.
Collapse
Affiliation(s)
- Toon Braeye
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium.
| | - Lucy Catteau
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Ruben Brondeel
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Joris A F van Loenhout
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Kristiaan Proesmans
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Laura Cornelissen
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Herman Van Oyen
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium; Department of Public Health and Primary Care, Ghent University, Corneel Heymanslaan 10, 9000 Gent, Belgium
| | - Veerle Stouten
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Pierre Hubin
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Matthieu Billuart
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Achille Djiena
- Agence pour une Vie de Qualité, Rue de la Rivelaine 11, 6061 Charleroi, Belgium
| | - Romain Mahieu
- Common Community Commission Brussels, Rue Belliard 71/1, 1040 Brussels, Belgium
| | - Naima Hammami
- Agency for Care and Health, Infection Prevention and Control, Flemish Community, Koningin Maria Hendrikaplein 70 bus 55, 9000 Gent, Belgium
| | - Dieter Van Cauteren
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| | - Chloé Wyndham-Thomas
- Department of Epidemiology and public health, Sciensano, Juliette Wytsmansstraat 14, 1000 Brussel, Belgium
| |
Collapse
|
21
|
Böröcz K, Kinyó Á, Simon D, Erdő-Bonyár S, Németh P, Berki T. Complexity of the Immune Response Elicited by Different COVID-19 Vaccines, in the Light of Natural Autoantibodies and Immunomodulatory Therapies. Int J Mol Sci 2023; 24:ijms24076439. [PMID: 37047412 PMCID: PMC10094397 DOI: 10.3390/ijms24076439] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/14/2023] Open
Abstract
Despite the abundance of data on the COVID-19 vaccine-induced immune activation, the impact of natural autoantibodies (nAAbs) on these processes is less well defined. Therefore, we investigated potential connections between vaccine efficacy and nAAb levels. We were also interested in the impact of immunomodulatory therapies on vaccine efficacy. Clinical residual samples were used for the assessment of the COVID-19 vaccine-elicited immune response (IR) (n=255), as well as for the investigation of the immunization-associated expansion of the nAAb pool (n=185). In order to study the potential interaction between immunomodulatory therapies and the vaccine-induced IR, untreated, healthy individuals and patients receiving anti-TNFα or anti-IL-17 therapies were compared (n total =45). In-house ELISAs (anticitrate synthase, anti-HSP60 and-70) and commercial ELISAs (anti-SARS-CoV-2 ELISAs IgG, IgA, NeutraLISA and IFN-γ release assay 'IGRA') were applied. We found significant differences in the IR given to different vaccines. Moreover, nAAb levels showed plasticity in response to anti-COVID-19 immunization. We conclude that our findings may support the theorem about the non-specific beneficial 'side effects' of vaccination, including the broadening of the nAAb repertoire. Considering immunomodulation, we suggest that anti-TNFα and anti-IL17 treatments may interfere negatively with MALT-associated IR, manifested as decreased IgA titers; however, the modest sample numbers of the herein presented model might be a limiting factor of reaching a more comprehensive conclusion.
Collapse
Affiliation(s)
- Katalin Böröcz
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Ágnes Kinyó
- Department of Dermatology, Venereology and Oncodermatology, Clinical Center, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Diana Simon
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Szabina Erdő-Bonyár
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Péter Németh
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, 7624 Pécs, Hungary
| | - Timea Berki
- Department of Immunology and Biotechnology, Clinical Center, University of Pécs Medical School, 7624 Pécs, Hungary
| |
Collapse
|
22
|
Sieghart D, Hana CA, Haslacher H, Perkmann T, Heinz LX, Fedrizzi C, Anderle K, Wiedermann U, Condur I, Drapalik S, Steinbrecher H, Mrak D, Mucher P, Hasenoehrl T, Zrdavkovic A, Wagner B, Palma S, Jordakieva G, Jorda A, Firbas C, Wagner A, Haiden N, Bergmann F, Crevenna R, Zeitlinger M, Bonelli M, Aletaha D, Radner H. Multiparametric Prediction Models for Coronavirus Disease 2019 Vaccine Selection: Results of a Comparative Population-Based Cohort Study. Clin Infect Dis 2023; 76:816-823. [PMID: 36328594 DOI: 10.1093/cid/ciac840] [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: 04/11/2022] [Revised: 10/12/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND An understanding vaccine-dependent effects on protective and sustained humoral immune response is crucial to planning future vaccination strategies against coronavirus disease 2019 (COVID-19). METHODS In this multicenter, population-based, cohort study including 4601 individuals after primary vaccination against COVID-19 ≥ 4 months earlier we compared factors associated with residual antibody levels against severe acute respiratory syndrome coronavirus-2 receptor-binding domain (RBD) across different vaccination strategies (BNT162b2, mRNA-1273, or ChAdOx1). RESULTS Our main model including 3787 individuals (2 × BNT162b2, n = 2271; 2 × mRNA-1273, n = 251; 2 × ChAdOx1, n = 1265), predicted significantly lower levels of anti-RBD antibodies after 6 months in individuals vaccinated with ChAdOx1 (392.7 binding antibody units per milliliter [BAU/mL]) compared with those vaccinated with BNT162b2 (1179.5 BAU/mL) or mRNA-1273 (2098.2 BAU/mL). Vaccine-dependent association of antibody levels was found for age with a significant predicted difference in BAU/ml per year for BNT162b2 (-21.5; 95% confidence interval [CI], -24.7 to -18.3) and no significant association for mRNA-1273 (-4.0; 95% CI, -20.0 to 12.1) or ChAdOx1 (1.7; 95% CI, .2 to 3.1). The predicted decrease over time since full immunization was highest in mRNA-1273 (-23.4; 95% CI, -31.4 to -15.4) compared with BNT162b2 (-5.9; 95% CI, -7 to -4.8). CONCLUSIONS Our study revealed population-based evidence of vaccine-dependent effects of age and time since full immunization on humoral immune response. Findings underline the importance of individualized vaccine selection, especially in elderly individuals.
Collapse
Affiliation(s)
- Daniela Sieghart
- Department of Internal Medicine III, Division of Rheumatology, Medical University Vienna, Vienna, Austria
| | - Claudia A Hana
- Department of Internal Medicine III, Division of Rheumatology, Medical University Vienna, Vienna, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Thomas Perkmann
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Leonhard X Heinz
- Department of Internal Medicine III, Division of Rheumatology, Medical University Vienna, Vienna, Austria
| | - Clemens Fedrizzi
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Karolina Anderle
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Ursula Wiedermann
- Center of Pathophysiology, Infectiology & Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Irina Condur
- Department of Internal Medicine III, Division of Rheumatology, Medical University Vienna, Vienna, Austria
| | | | | | - Daniel Mrak
- Department of Internal Medicine III, Division of Rheumatology, Medical University Vienna, Vienna, Austria
| | - Patrick Mucher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Timothy Hasenoehrl
- Department of Physical Medicine, Rehabilitation and Occupational Medicine, Medical University Vienna, Vienna, Austria
| | - Andrej Zrdavkovic
- Department of Physical Medicine, Rehabilitation and Occupational Medicine, Medical University Vienna, Vienna, Austria
| | - Barbara Wagner
- Department of Physical Medicine, Rehabilitation and Occupational Medicine, Medical University Vienna, Vienna, Austria
| | - Stefano Palma
- Department of Physical Medicine, Rehabilitation and Occupational Medicine, Medical University Vienna, Vienna, Austria
| | - Galateja Jordakieva
- Department of Physical Medicine, Rehabilitation and Occupational Medicine, Medical University Vienna, Vienna, Austria
| | - Anselm Jorda
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Christa Firbas
- Department of Physical Medicine, Rehabilitation and Occupational Medicine, Medical University Vienna, Vienna, Austria
| | - Angelika Wagner
- Center of Pathophysiology, Infectiology & Immunology, Institute of Specific Prophylaxis and Tropical Medicine, Medical University of Vienna, Vienna, Austria
| | - Nadja Haiden
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Felix Bergmann
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Richard Crevenna
- Department of Physical Medicine, Rehabilitation and Occupational Medicine, Medical University Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Michael Bonelli
- Department of Internal Medicine III, Division of Rheumatology, Medical University Vienna, Vienna, Austria
| | - Daniel Aletaha
- Department of Internal Medicine III, Division of Rheumatology, Medical University Vienna, Vienna, Austria
| | - Helga Radner
- Department of Internal Medicine III, Division of Rheumatology, Medical University Vienna, Vienna, Austria
| |
Collapse
|
23
|
Townsend JP, Hassler HB, Dornburg A. Infection by SARS-CoV-2 with alternate frequencies of mRNA vaccine boosting. J Med Virol 2023; 95:e28461. [PMID: 36602045 DOI: 10.1002/jmv.28461] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 12/16/2022] [Accepted: 12/26/2022] [Indexed: 01/06/2023]
Abstract
One of the most consequential unknowns of the COVID-19 pandemic is the frequency at which vaccine boosting provides sufficient protection from infection. We quantified the statistical likelihood of breakthrough infections over time following different boosting schedules with messenger RNA (mRNA)-1273 (Moderna) and BNT162b2 (Pfizer-BioNTech). We integrated anti-Spike IgG antibody optical densities with profiles of the waning of antibodies and corresponding probabilities of infection associated with coronavirus endemic transmission. Projecting antibody levels over time given boosting every 6 months, 1, 1.5, 2, or 3 years yielded respective probabilities of fending off infection over a 6-year span of >93%, 75%, 55%, 40%, and 24% (mRNA-1273) and >89%, 69%, 49%, 36%, and 23% (BNT162b2). Delaying the administration of updated boosters has bleak repercussions. It increases the probability of individual infection by SARS-CoV-2, and correspondingly, ongoing disease spread, prevalence, morbidity, hospitalization, and mortality. Instituting regular, population-wide booster vaccination updated to predominant variants has the potential to substantially forestall-and with global, widespread uptake, eliminate-COVID-19.
Collapse
Affiliation(s)
- Jeffrey P Townsend
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA.,Department of Ecology and Evolutionary Biology, Yale University, New Haven, Connecticut, USA.,Program in Computational Biology and Bioinformatics, Yale University, New Haven, Connecticut, USA.,Program in Microbiology, Yale University, New Haven, Connecticut, USA
| | - Hayley B Hassler
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut, USA
| | - Alex Dornburg
- Department of Bioinformatics and Genomics, University of North Carolina, Charlotte, North Carolina, USA
| |
Collapse
|
24
|
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
|
25
|
Onifade AA, Fowotade A, Rahamon SK, Edem VF, Yaqub SA, Akande OK, Arinola OG. Seroprevalence of anti-SARS-CoV-2 specific antibodies in vaccinated and vaccine naïve adult Nigerians. PLoS One 2023; 18:e0280276. [PMID: 36689402 PMCID: PMC9870169 DOI: 10.1371/journal.pone.0280276] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 12/26/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Reports on the evaluation of immune responses to different COVID-19 vaccines are limited. Similarly, effects of age and gender have not been well explored as variables that could impact on the vaccine-induced antibody response. Therefore, seroprevalence of anti-SARS-CoV-2 specific antibodies in vaccinated and vaccine naïve adult Nigerians was determined in this study. METHODOLOGY A total of 141 adults were enrolled into this study. Presence or absence of SARS-CoV-2 infection was confirmed by real-time reverse-transcriptase polymerase-chain reaction (RT-PCR) assay on nasopharyngeal and oropharyngeal swab specimens. Anti-SARS-CoV-2 Specific IgG and IgM antibodies were qualitatively detected using a Rapid Diagnostic Test kit. RESULTS Pre-vaccination, 77% of the study participants had never had PCR-confirmed COVID-19 test yet 66.7% of them were seropositive for SARS-CoV-2 antibodies. Of 111 COVID-19 vaccinated participants, 69.2% and 73.8% of them had SARS-CoV-2 specific IgG post-first and second doses of COVID-19 vaccine respectively. However, 23.1% and 21.4% of the participants who have had first and second doses respectively had no detectable anti-SARS-CoV-2 antibodies. The proportion of participants with SARS-CoV-2 specific IgG was insignificantly higher in those between the ages of 18-40 years and 41-59 years compared with individuals aged ≥60 years. No significant association was observed between gender and seropositivity for SARS-CoV-2 antibodies. CONCLUSION There is high SARS-CoV-2 antibody seroprevalence among Nigerian adults who never had PCR-confirmed COVID-19. Also, there is the need for anti-SARS-CoV-2 antibodies screening post vaccination as this could be essential in achieving herd immunity. Age and gender do not seem to have significant association with seropositivity.
Collapse
Affiliation(s)
| | - Adeola Fowotade
- Biorepository Clinical Virology Laboratory, College of Medicine, University of Ibadan, Ibadan, Nigeria
- Department of Medical Microbiology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Sheu Kadiri Rahamon
- Department of Immunology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Victory Fabian Edem
- Department of Immunology, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | | - Olatunji Kadri Akande
- Biorepository Clinical Virology Laboratory, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | | |
Collapse
|
26
|
Antibody Response Over Time to SARS-CoV-2 Vaccination in US Veterans Living With Human Immunodeficiency Virus. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2023. [DOI: 10.1097/ipc.0000000000001209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
27
|
Ailsworth SM, Keshavarz B, Richards NE, Workman LJ, Murphy DD, Nelson MR, Platts-Mills TAE, Wilson JM. Enhanced SARS-CoV-2 IgG durability following COVID-19 mRNA booster vaccination and comparison of BNT162b2 with mRNA-1273. Ann Allergy Asthma Immunol 2023; 130:67-73. [PMID: 36241020 PMCID: PMC9553965 DOI: 10.1016/j.anai.2022.10.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/19/2022] [Accepted: 10/03/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND BNT162b2 (Pfizer/BioNTech, Comirnaty) and mRNA-1273 (Moderna, Spikevax) are messenger RNA (mRNA) vaccines that elicit antibodies against the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike receptor-binding domain (S-RBD) and have been approved by the US Food and Drug Administration to combat the coronavirus disease 2019 (COVID-19) pandemic. Because vaccine efficacy and antibody levels waned over time after the 2-shot primary series, the US Food and Drug Administration authorized a booster (third) dose for both mRNA vaccines to adults in the fall of 2021. OBJECTIVE To evaluate the magnitude and durability of S-RBD immunoglobulin (Ig)G after the booster mRNA vaccine dose in comparison to the primary series. We also compared S-RBD IgG levels after BNT162b2 and mRNA-1273 boosters and explored effects of age and prior infection. METHODS Surrounding receipt of the second and third homologous mRNA vaccine doses, adults in an employee-based cohort provided serum and completed questionnaires, including information about previous COVID-19 infection. The IgG to S-RBD was measured using an ImmunoCAP-based system. A subset of samples were assayed for IgG to SARS-CoV-2 nucleocapsid by commercial assay. RESULTS There were 228 subjects who had samples collected between 7 and 150 days after their primary series vaccine and 117 subjects who had samples collected in the same time frame after their boost. Antibody levels from 7 to 31 days after the primary series and booster were similar, but S-RBD IgG was more durable over time after the boost, regardless of prior infection status. In addition, mRNA-1273 post-boost antibody levels exceeded BNT162b2 out to 5 months. CONCLUSION The COVID-19 mRNA vaccine boosters increase antibody durability, suggesting enhanced long-term clinical protection from SARS-CoV-2 infection compared with the 2-shot regimen.
Collapse
Affiliation(s)
- Samuel M Ailsworth
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Behnam Keshavarz
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Nathan E Richards
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Lisa J Workman
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Deborah D Murphy
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Michael R Nelson
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Thomas A E Platts-Mills
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Virginia
| | - Jeffrey M Wilson
- Division of Allergy & Clinical Immunology, Department of Medicine, University of Virginia, Charlottesville, Virginia.
| |
Collapse
|
28
|
Bjarnarson SP, Brynjolfsson SF. The role of antigen availability during B cell induction and its effect on sustained memory and antibody production after infection and vaccination-lessons learned from the SARS-CoV-2 pandemic. Clin Exp Immunol 2022; 210:273-282. [PMID: 36480298 PMCID: PMC9985164 DOI: 10.1093/cei/uxac113] [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: 07/01/2022] [Revised: 10/05/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
The importance of antibodies, particularly neutralizing antibodies, has been known for decades. When examining the immune responses against a pathogen after a vaccination or infection it is easier to measure the levels of antigen-specific antibodies than the T-cell response, but it does not give the whole picture. The levels of neutralizing antibodies are harder to determine but give a better indication of the quality of the antibody response. The induction of long-lived antibody-secreting plasma cells is crucial for a persistent humoral immune response, which has been shown for example after vaccination with the vaccinia vaccine, where antibody levels have been shown to persist for decades. With the SARS-CoV-2 pandemic ravaging the world for the past years and the monumental effort in designing and releasing novel vaccines against the virus, much effort has been put into analysing the quantity, quality, and persistence of antibody responses.
Collapse
Affiliation(s)
- Stefania P Bjarnarson
- Department of Immunology, Landspitali—The National University Hospital of Iceland, Reykjavik, Iceland
- Faculty of Medicine, Biomedical Center, University of Iceland, Reykjavik, Iceland
| | - Siggeir F Brynjolfsson
- Correspondence: Department of Immunology, Landspitali—The National University Hospital of Iceland, Reykjavik, Iceland.
| |
Collapse
|
29
|
Gil-Manso S, Alonso R, Catalán P, Sánchez-Arcilla I, Marzola M, Correa-Rocha R, Pion M, Muñoz P. IgG anti-RBD levels during 8-month follow-up post-vaccination with BNT162b2 and mRNA-1273 vaccines in healthcare workers: A one-center study. Front Cell Infect Microbiol 2022; 12:1035155. [PMID: 36530428 PMCID: PMC9748346 DOI: 10.3389/fcimb.2022.1035155] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 11/01/2022] [Indexed: 12/02/2022] Open
Abstract
Introduction Since the COVID-19 outbreak, specific mRNA-based anti-SARS-CoV-2 vaccines have been developed and distributed worldwide. Because this is the first time that mRNA vaccines have been used, there are several questions regarding their capacity to confer immunity and the durability of the specific anti-SARS-CoV-2 response. Therefore, the objective of this study was to recruit a large cohort of healthcare workers from the Gregorio Marañón Hospital vaccinated with the mRNA-1273 or BNT126b2 vaccines and to follow-up on IgG anti-RBD levels at 8 months post-vaccination. Methods We recruited 4,970 volunteers and measured IgG anti-RBD antibodies on days 30 and 240 post-vaccination. Results We observed that both vaccines induced high levels of antibodies on day 30, while a drastic wane was observed on day 240, where mRNA-1273 vaccinated induced higher levels than BNT162b2. Stratifying by vaccine type, age, gender, and comorbidities, we identified that older mRNA-1273-vaccinated volunteers had higher antibody levels than the younger volunteers, contrary to what was observed in the BNT162b2-vaccinated volunteers. Discussion In conclusion, we observed that mRNA-1273 has a higher capacity to induce a humoral response than BNT162b2 and that age is a factor in the specific response.
Collapse
Affiliation(s)
- Sergio Gil-Manso
- Advanced ImmunoRegulation Group, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain,*Correspondence: Sergio Gil-Manso, ; Roberto Alonso,
| | - Roberto Alonso
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain,Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain,CIBER (Centro de Investigación Biomédicas en Red) de Enfermedades Respiratorias, CIBERES, Barcelona, Spain,Department of Medicine, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain,*Correspondence: Sergio Gil-Manso, ; Roberto Alonso,
| | - Pilar Catalán
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain,Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain,CIBER (Centro de Investigación Biomédicas en Red) de Enfermedades Respiratorias, CIBERES, Barcelona, Spain
| | - Ignacio Sánchez-Arcilla
- Department of Labour Risks Prevention, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Marco Marzola
- Department of Labour Risks Prevention, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Marjorie Pion
- Advanced ImmunoRegulation Group, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain,Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, Madrid, Spain,CIBER (Centro de Investigación Biomédicas en Red) de Enfermedades Respiratorias, CIBERES, Barcelona, Spain,Department of Medicine, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | | |
Collapse
|
30
|
Babouee Flury B, Güsewell S, Egger T, Leal O, Brucher A, Lemmenmeier E, Meier Kleeb D, Möller JC, Rieder P, Rütti M, Schmid HR, Stocker R, Vuichard-Gysin D, Wiggli B, Besold U, McGeer A, Risch L, Friedl A, Schlegel M, Kuster SP, Kahlert CR, Kohler P. Risk and symptoms of COVID-19 in health professionals according to baseline immune status and booster vaccination during the Delta and Omicron waves in Switzerland-A multicentre cohort study. PLoS Med 2022; 19:e1004125. [PMID: 36342956 PMCID: PMC9678290 DOI: 10.1371/journal.pmed.1004125] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 11/21/2022] [Accepted: 10/14/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Knowledge about protection conferred by previous Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection and/or vaccination against emerging viral variants allows clinicians, epidemiologists, and health authorities to predict and reduce the future Coronavirus Disease 2019 (COVID-19) burden. We investigated the risk and symptoms of SARS-CoV-2 (re)infection and vaccine breakthrough infection during the Delta and Omicron waves, depending on baseline immune status and subsequent vaccinations. METHODS AND FINDINGS In this prospective, multicentre cohort performed between August 2020 and March 2022, we recruited hospital employees from ten acute/nonacute healthcare networks in Eastern/Northern Switzerland. We determined immune status in September 2021 based on serology and previous SARS-CoV-2 infections/vaccinations: Group N (no immunity); Group V (twice vaccinated, uninfected); Group I (infected, unvaccinated); Group H (hybrid: infected and ≥1 vaccination). Date and symptoms of (re)infections and subsequent (booster) vaccinations were recorded until March 2022. We compared the time to positive SARS-CoV-2 swab and number of symptoms according to immune status, viral variant (i.e., Delta-dominant before December 27, 2021; Omicron-dominant on/after this date), and subsequent vaccinations, adjusting for exposure/behavior variables. Among 2,595 participants (median follow-up 171 days), we observed 764 (29%) (re)infections, thereof 591 during the Omicron period. Compared to group N, the hazard ratio (HR) for (re)infection was 0.33 (95% confidence interval [CI] 0.22 to 0.50, p < 0.001) for V, 0.25 (95% CI 0.11 to 0.57, p = 0.001) for I, and 0.04 (95% CI 0.02 to 0.10, p < 0.001) for H in the Delta period. HRs substantially increased during the Omicron period for all groups; in multivariable analyses, only belonging to group H was associated with protection (adjusted HR [aHR] 0.52, 95% CI 0.35 to 0.77, p = 0.001); booster vaccination was associated with reduction of breakthrough infection risk in groups V (aHR 0.68, 95% CI 0.54 to 0.85, p = 0.001) and H (aHR 0.67, 95% CI 0.45 to 1.00, p = 0.048), largely observed in the early Omicron period. Group H (versus N, risk ratio (RR) 0.80, 95% CI 0.66 to 0.97, p = 0.021) and participants with booster vaccination (versus nonboosted, RR 0.79, 95% CI 0.71 to 0.88, p < 0.001) reported less symptoms during infection. Important limitations are that SARS-CoV-2 swab results were self-reported and that results on viral variants were inferred from the predominating strain circulating in the community at that time, rather than sequencing. CONCLUSIONS Our data suggest that hybrid immunity and booster vaccination are associated with a reduced risk and reduced symptom number of SARS-CoV-2 infection during Delta- and Omicron-dominant periods. For previously noninfected individuals, booster vaccination might reduce the risk of symptomatic Omicron infection, although this benefit seems to wane over time.
Collapse
Affiliation(s)
- Baharak Babouee Flury
- Cantonal Hospital St Gallen, Division of Infectious Diseases and Hospital Epidemiology, St Gallen, Switzerland
| | - Sabine Güsewell
- Cantonal Hospital St Gallen, Division of Infectious Diseases and Hospital Epidemiology, St Gallen, Switzerland
| | - Thomas Egger
- Cantonal Hospital St Gallen, Division of Infectious Diseases and Hospital Epidemiology, St Gallen, Switzerland
| | - Onicio Leal
- Epitrack, Recife, Brazil
- Department of Economics, University of Zurich, Zurich, Switzerland
| | - Angela Brucher
- Psychiatry Services of the Canton of St. Gallen (South), St Gallen, Switzerland
| | - Eva Lemmenmeier
- Clienia Littenheid AG, Private Clinic for Psychiatry and Psychotherapy, Littenheid, Switzerland
| | | | | | | | - Markus Rütti
- Fuerstenland Toggenburg Hospital Group, Wil, Switzerland
| | | | | | - Danielle Vuichard-Gysin
- Thurgau Hospital Group, Division of Infectious Diseases and Hospital Epidemiology, Muensterlingen, Switzerland
| | - Benedikt Wiggli
- Kantonsspital Baden, Division of Infectious Diseases and Hospital Epidemiology, Baden, Switzerland
| | - Ulrike Besold
- Geriatric Clinic St. Gallen, St. Gallen, Switzerland
| | | | - Lorenz Risch
- Labormedizinisches Zentrum Dr Risch Ostschweiz AG, Buchs, Switzerland
- Private Universität im Fürstentum Liechtenstein, Triesen, Liechtenstein
- Center of Laboratory Medicine, University Institute of Clinical Chemistry, University of Bern, Inselspital, Bern, Switzerland
| | - Andrée Friedl
- Kantonsspital Baden, Division of Infectious Diseases and Hospital Epidemiology, Baden, Switzerland
| | - Matthias Schlegel
- Cantonal Hospital St Gallen, Division of Infectious Diseases and Hospital Epidemiology, St Gallen, Switzerland
| | - Stefan P. Kuster
- Cantonal Hospital St Gallen, Division of Infectious Diseases and Hospital Epidemiology, St Gallen, Switzerland
| | - Christian R. Kahlert
- Cantonal Hospital St Gallen, Division of Infectious Diseases and Hospital Epidemiology, St Gallen, Switzerland
- Children’s Hospital of Eastern Switzerland, Department of Infectious Diseases and Hospital Epidemiology, St. Gallen, Switzerland
- * E-mail: (CRK); (PK)
| | - Philipp Kohler
- Cantonal Hospital St Gallen, Division of Infectious Diseases and Hospital Epidemiology, St Gallen, Switzerland
- * E-mail: (CRK); (PK)
| | | |
Collapse
|
31
|
Avni Biron I, Maayan Y, Mishael T, Hadar E, Neeman M, Plitman Mayo R, Sela HY, Yagel S, Goldenberg R, Ben Ya’acov A, Grisaru Granovsky S, Ollech JE, Edelman-Klapper H, Rabinowitz KM, Pauker MH, Yanai H, Goren S, Cohen D, Dotan I, Bar-Gil Shitrit A. SARS-CoV-2 IgG Antibody Levels in Women with IBD Vaccinated during Pregnancy. Vaccines (Basel) 2022; 10:1833. [PMID: 36366342 PMCID: PMC9699106 DOI: 10.3390/vaccines10111833] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/21/2022] [Accepted: 10/24/2022] [Indexed: 10/10/2023] Open
Abstract
INTRODUCTION Regulatory agencies supported vaccination of pregnant women with SARS-CoV-2 mRNA vaccines, including patients with IBD. No data exist regarding these vaccines in IBD during pregnancy. AIM To assess the serologic response to two doses of the mRNA SARS-CoV-2 BNT162b2 vaccine in pregnant women with IBD vaccinated during pregnancy, compared to that of pregnant women without IBD, and non-pregnant women with IBD. METHODS Anti-spike antibody levels were assessed in all women and in cord blood of consenting women. RESULTS From December 2020 to December 2021, 139 women were assessed: pregnant with IBD-36, pregnant without IBD-61, and not pregnant with IBD-42. Antibodies were assessed in cords of two and nine newborns of women with and without IBD, respectively. Mean gestational ages at administration of the second vaccine doses were 22.0 weeks in IBD and 23.2 weeks in non-IBD, respectively. Mean (SD) duration from the second vaccine dose to serology analysis in pregnant women with IBD, without IBD, and in non-pregnant women with IBD was 10.6 (4.9), 16.4 (6.3), and 4.3 (1.0) weeks, respectively. All women mounted a serologic response. In multivariable analysis, no correlation was found between the specific group and antibody levels. In both pregnancy groups, an inverse correlation between antibody levels and the interval from the second vaccine dose was demonstrated. Cord blood antibody levels exceeded maternal levels in women with and without IBD. CONCLUSION All patients with IBD mounted a serologic response. The interval between vaccine administration to serology assessment was the most important factor determining antibody levels. A third vaccine dose should be considered in pregnant women with IBD vaccinated at early stages of pregnancy.
Collapse
Affiliation(s)
- Irit Avni Biron
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Rabin Medical Center, Petah-Tikva 4919001, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 9436008, Israel
| | - Yair Maayan
- IBD MOM Unit, Shaare Zedek Medical Center, Digestive Diseases Institute, Jerusalem 9436008, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9103102, Israel
- Department of Military Medicine and “Tzameret”, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel, and Medical Corps, Israel Defense Forces, Rehovot 7661041, Israel
| | - Tali Mishael
- IBD MOM Unit, Shaare Zedek Medical Center, Digestive Diseases Institute, Jerusalem 9436008, Israel
- Obstetrics and Gynecology Department, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| | - Eran Hadar
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 9436008, Israel
- Helen Schneider Hospital for Women, Rabin Medical Center, Petah-Tikva 49100, Israel
| | - Michal Neeman
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Romina Plitman Mayo
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Hen Y. Sela
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9103102, Israel
- Obstetrics and Gynecology Department, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| | - Simcha Yagel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9103102, Israel
- Obstetrics and Gynecology Department, Mount Scopus Hadassah Medical Center, Jerusalem 9103102, Israel
| | - Rosalind Goldenberg
- IBD MOM Unit, Shaare Zedek Medical Center, Digestive Diseases Institute, Jerusalem 9436008, Israel
| | - Ami Ben Ya’acov
- IBD MOM Unit, Shaare Zedek Medical Center, Digestive Diseases Institute, Jerusalem 9436008, Israel
| | - Sorina Grisaru Granovsky
- IBD MOM Unit, Shaare Zedek Medical Center, Digestive Diseases Institute, Jerusalem 9436008, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9103102, Israel
- Obstetrics and Gynecology Department, Shaare Zedek Medical Center, Jerusalem 9103102, Israel
| | - Jacob E. Ollech
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Rabin Medical Center, Petah-Tikva 4919001, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 9436008, Israel
| | - Hadar Edelman-Klapper
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Rabin Medical Center, Petah-Tikva 4919001, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 9436008, Israel
| | - Keren Masha Rabinowitz
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Rabin Medical Center, Petah-Tikva 4919001, Israel
- Felsenstein Medical Research Center, Sackler School of Medicine, Tel Aviv 69978, Israel
| | - Maor H. Pauker
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Rabin Medical Center, Petah-Tikva 4919001, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 9436008, Israel
| | - Henit Yanai
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Rabin Medical Center, Petah-Tikva 4919001, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 9436008, Israel
| | - Sophy Goren
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dani Cohen
- School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Iris Dotan
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Rabin Medical Center, Petah-Tikva 4919001, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 9436008, Israel
| | - Ariella Bar-Gil Shitrit
- IBD MOM Unit, Shaare Zedek Medical Center, Digestive Diseases Institute, Jerusalem 9436008, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9103102, Israel
| |
Collapse
|
32
|
Importance of the COVID-19 Vaccine Booster Dose in Protection and Immunity. Vaccines (Basel) 2022; 10:vaccines10101708. [PMID: 36298573 PMCID: PMC9610198 DOI: 10.3390/vaccines10101708] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/07/2022] [Accepted: 10/11/2022] [Indexed: 11/23/2022] Open
Abstract
Background: There is debate on the necessity of booster doses of COVID-19 vaccination, especially in countries with limited resources. Methods: This cross-sectional study was conducted in a referral laboratory in Tehran, Iran. The level of COVID-19 antibodies was measured and compared between individuals regarding the number of COVID-19 vaccine shots. Results: In this study, 176 individuals with a mean age of 36.3 (±11.7) years participated. A total of 112 individuals received two doses of the COVID-19 vaccine, and 64 individuals received three doses. Level of all antibodies was higher in those who received three doses than in those who received two doses of the COVID-19 vaccine. Considering the SARS-CoV-2 Spike IgG, the difference was not statistically significant but for the SARS-CoV-2 RBD IgG and SARS-CoV-2 NAB the difference was statistically significant. Regarding to the background variables, receiving influenza vaccine in the past year, history of autoimmune diseases and past medical history of chicken pox showed a significant association with the number of vaccine doses received. Their effects on the outcome variables assessed with multivariate logistic regression analysis. Conclusion: The results of our study show that a booster dose of the COVID-19 vaccine enhances the antibody response.
Collapse
|
33
|
Sugiyama A, Kurisu A, Nagashima S, Hando K, Saipova K, Akhmedova S, Abe K, Imada H, Hussain MRA, Ouoba S, E B, Ko K, Akita T, Yamazaki S, Yokozaki M, Tanaka J. Seroepidemiological study of factors affecting anti-spike IgG antibody titers after a two-dose mRNA COVID-19 vaccination in 3744 healthy Japanese volunteers. Sci Rep 2022; 12:16294. [PMID: 36175506 PMCID: PMC9520958 DOI: 10.1038/s41598-022-20747-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/19/2022] [Indexed: 02/06/2023] Open
Abstract
Several factors related to anti-spike(S) IgG antibody titers after mRNA COVID-19 vaccination have been elucidated, but the magnitude of the effects of each factor has not been fully understood. This cross-sectional study assessed anti-S and anti-nucleocapsid (N) antibody titers on 3744 healthy volunteers (median age, 36 years; IQR, 24-49 years; females, 59.0%) who received two doses of mRNA-1273 or BNT162b2 vaccine and completed a survey questionnaire. Multiple regression was conducted to identify factors associated with antibody titers. All but one participant tested positive for anti-S antibodies (99.97%). The following factors were independently and significantly associated with high antibody titer: < 3 months from vaccination (ratio of means 4.41); mRNA-1273 vaccine (1.90, vs BNT162b2); anti-N antibody positivity (1.62); age (10's: 1.50, 20's: 1.37, 30's: 1.26, 40's: 1.16, 50's: 1.15, vs ≧60's); female (1.07); immunosuppressive therapy (0.54); current smoking (0.85); and current drinking (0.96). The largest impact on anti-S IgG antibody titers was found in elapsed time after vaccination, followed by vaccine brand, immunosuppressants, previous SARS-CoV-2 infection (anti-N antibody positive), and age. Although the influence of adverse reactions after the vaccine, gender, smoking, and drinking was relatively small, they were independently related factors.
Collapse
Affiliation(s)
- Aya Sugiyama
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Akemi Kurisu
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Shintaro Nagashima
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Kiyomi Hando
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Khilola Saipova
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan ,grid.444564.30000 0004 0402 7972Department of Clinical Radiology and Oncology, Andijan State Medical Institute, Andijan, Uzbekistan
| | - Sayyora Akhmedova
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan ,Department of Cardiorheumatology, Republican Specialized Scientific-Practical Medical Center of Pediatrics, Tashkent, Uzbekistan
| | - Kanon Abe
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Hirohito Imada
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Md Razeen Ashraf Hussain
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Serge Ouoba
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan ,grid.457337.10000 0004 0564 0509Unité de Recherche Clinique de Nanoro (URCN), Institut de Recherche en Science de La Santé (IRSS), Nanoro, Burkina Faso
| | - Bunthen E
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan ,grid.415732.6Payment Certification Agency (PCA), Ministry of Health, Phnom Penh, Cambodia
| | - Ko Ko
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Tomoyuki Akita
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| | - Shinichi Yamazaki
- grid.470097.d0000 0004 0618 7953Division of Clinical Laboratory Medicine, Hiroshima University Hospital, Hiroshima, Japan
| | - Michiya Yokozaki
- grid.470097.d0000 0004 0618 7953Division of Clinical Laboratory Medicine, Hiroshima University Hospital, Hiroshima, Japan
| | - Junko Tanaka
- grid.257022.00000 0000 8711 3200Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3, Kasumi, Minami-ku, Hiroshima, 734-8551 Japan
| |
Collapse
|
34
|
Di Giacomo AM, Giacobini G, Anichini G, Gandolfo C, D'alonzo V, Calabrò L, Lofiego MF, Cusi MG, Maio M. SARS-CoV-2 infection in cancer patients on active therapy after the booster dose of mRNA vaccines. Eur J Cancer 2022; 171:143-149. [PMID: 35717822 PMCID: PMC9130712 DOI: 10.1016/j.ejca.2022.05.018] [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: 03/31/2022] [Revised: 05/04/2022] [Accepted: 05/13/2022] [Indexed: 11/18/2022]
Abstract
INTRODUCTION The protective role against SARS-CoV-2 infection by the third booster dose of mRNA vaccines in cancer patients with solid malignancies is presently unknown. We prospectively investigated the occurrence of COVID-19 in cancer patients on active therapy after the booster vaccine dose. METHODS Cancer patients on treatment at the Center for Immuno-Oncology (CIO) of the University Hospital of Siena, Italy, and health care workers at CIO who had received a booster third dose of mRNA vaccine entered a systematic follow-up monitoring period to prospectively assess their potential risk of SARS-CoV-2 infection. Serological and microneutralization assay were utilized to assess levels of anti-spike IgG, and of neutralizing antibodies to the SARS-CoV-2 Wild Type, Delta and Omicron variants, respectively, after the booster dose and after negativization of the nasopharyngeal swab for those who had developed COVID-19. RESULTS Ninety cancer patients with solid tumors on active treatment (Cohort 1) and 30 health care workers (Cohort 2) underwent a booster third dose of mRNA vaccine. After the booster dose, the median value of anti-spike IgG was higher (p = 0.009) in patients than in healthy subjects. Remarkably, 11/90 (12%) patients and 11/30 (37%) healthy subjects tested positive to SARS-CoV-2 infection during the monitoring period. Similar levels of anti-spike IgG and of neutralizing antibodies against all the investigated variants, with geometric mean titers of neutralizing antibodies against the Omicron being the lowest were detected after the booster dose and after COVID-19 in both Cohorts. CONCLUSIONS The occurrence of SARS-CoV-2 infection we observed in a sizable proportion of booster-dosed cancer patients and in healthy subjects during the Omicron outbreak indicates that highly specific vaccines against SARS-CoV-2 variants are urgently required.
Collapse
Affiliation(s)
- Anna M Di Giacomo
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, Department of Oncology, University Hospital of Siena, Siena, Italy; University of Siena, Siena, Italy; Italian Network for Tumor Bio-Immunotherapy Foundation Onlus, Italy
| | - Gianluca Giacobini
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, Department of Oncology, University Hospital of Siena, Siena, Italy
| | - Gabriele Anichini
- Virology Unit, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Claudia Gandolfo
- Virology Unit, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Vincenzo D'alonzo
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, Department of Oncology, University Hospital of Siena, Siena, Italy; University of Siena, Siena, Italy
| | - Luana Calabrò
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, Department of Oncology, University Hospital of Siena, Siena, Italy
| | - Maria F Lofiego
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, Department of Oncology, University Hospital of Siena, Siena, Italy
| | - Maria G Cusi
- Virology Unit, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Michele Maio
- Center for Immuno-Oncology, Medical Oncology and Immunotherapy, Department of Oncology, University Hospital of Siena, Siena, Italy; University of Siena, Siena, Italy; Italian Network for Tumor Bio-Immunotherapy Foundation Onlus, Italy.
| |
Collapse
|
35
|
Gil-Manso S, Carbonell D, Pérez-Fernández VA, López-Esteban R, Alonso R, Muñoz P, Ochando J, Sánchez-Arcilla I, Bellón JM, Correa-Rocha R, Pion M. Cellular and Humoral Responses Follow-up for 8 Months after Vaccination with mRNA-Based Anti-SARS-CoV-2 Vaccines. Biomedicines 2022; 10:biomedicines10071676. [PMID: 35884980 PMCID: PMC9312914 DOI: 10.3390/biomedicines10071676] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/01/2022] [Accepted: 07/09/2022] [Indexed: 11/16/2022] Open
Abstract
Vaccination against SARS-CoV-2 has become the main method of reducing mortality and severity of COVID-19. This work aims to study the evolution of the cellular and humoral responses conferred by two mRNA vaccines after two doses against SARS-CoV-2. On days 30 and 240 after the second dose of both vaccines, the anti-S antibodies in plasma were evaluated from 82 volunteers vaccinated with BNT162b2 and 68 vaccinated with mRNA-1273. Peripheral blood was stimulated with peptides encompassing the entire SARS-CoV-2 Spike sequence. IgG Anti-S antibodies (humoral) were quantified on plasma, and inflammatory cytokines (cellular) were measured after stimulation. We observed a higher response (both humoral and cellular) with the mRNA-1273 vaccine. Stratifying by age and gender, differences between vaccines were observed, especially in women under 48 and men over 48 years old. Therefore, this work could help to set up a vaccination strategy that could be applied to confer maximum immunity.
Collapse
Affiliation(s)
- Sergio Gil-Manso
- Advanced Immunoregulation Group, Gregorio Marañón Health Research Institute (IiSGM), Gregorio Marañón University General Hospital, 28009 Madrid, Spain; (S.G.-M.); (D.C.); (V.A.P.-F.)
| | - Diego Carbonell
- Advanced Immunoregulation Group, Gregorio Marañón Health Research Institute (IiSGM), Gregorio Marañón University General Hospital, 28009 Madrid, Spain; (S.G.-M.); (D.C.); (V.A.P.-F.)
- Department of Hematology, Gregorio Marañón Health Research Institute (IiSGM), Gregorio Marañón University General Hospital, 28009 Madrid, Spain
| | - Verónica Astrid Pérez-Fernández
- Advanced Immunoregulation Group, Gregorio Marañón Health Research Institute (IiSGM), Gregorio Marañón University General Hospital, 28009 Madrid, Spain; (S.G.-M.); (D.C.); (V.A.P.-F.)
| | - Rocío López-Esteban
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IiSGM), Gregorio Marañón University General Hospital, 28009 Madrid, Spain; (R.L.-E.); (R.C.-R.)
| | - Roberto Alonso
- Department of Clinical Microbiology and Infectious Diseases, Gregorio Marañón University General Hospital, 28009 Madrid, Spain; (R.A.); (P.M.)
- School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Gregorio Marañón University General Hospital, 28009 Madrid, Spain; (R.A.); (P.M.)
- School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Jordi Ochando
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- National Centre of Microbiology, Carlos III Health Institute, 28222 Madrid, Spain
| | - Ignacio Sánchez-Arcilla
- Department of Occupational Risk Prevention, Gregorio Marañón University General Hospital, 28009 Madrid, Spain;
| | - Jose M Bellón
- Department of Biostatistics, Gregorio Marañón Health Research Institute (IiSGM), Gregorio Marañón University General Hospital, 28009 Madrid, Spain;
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IiSGM), Gregorio Marañón University General Hospital, 28009 Madrid, Spain; (R.L.-E.); (R.C.-R.)
| | - Marjorie Pion
- Advanced Immunoregulation Group, Gregorio Marañón Health Research Institute (IiSGM), Gregorio Marañón University General Hospital, 28009 Madrid, Spain; (S.G.-M.); (D.C.); (V.A.P.-F.)
- Correspondence: ; Tel.: +34-664-43-44-02
| |
Collapse
|
36
|
Matveeva O, Ershov A. Retrospective Cohort Study of the Effectiveness of the Sputnik V and EpiVacCorona Vaccines against the SARS-CoV-2 Delta Variant in Moscow (June-July 2021). Vaccines (Basel) 2022; 10:984. [PMID: 35891148 PMCID: PMC9320764 DOI: 10.3390/vaccines10070984] [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: 06/01/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 02/04/2023] Open
Abstract
The goal of this study was to evaluate the epidemiological effectiveness of the Sputnik V and EpiVacCorona vaccines against COVID-19. This work is a retrospective cohort study of COVID-19 patients. The cohort created by the Moscow Health Department included more than 300,000 infected people who sought medical care in June and July 2021. Analysis of data revealed a tendency for the increase in the Sputnik V vaccine effectiveness (VE) as the severity of the disease increased. Protection was the lowest for mild disease, and it was more pronounced for severe disease. We also observed a decrease in VE with increasing age. For the youngest group (18-50 years old), the estimated VE in preventing death in June 2021 was 95% (95% CI 64-100), and for the older group (50+ years old), it was 74% (95% CI 67-87). The estimated protection against a severe form of the disease in the 18-50-year-old group was above 81% (CI 95% 72-93), and in the 50+ years-old group, it was above 68% (CI 95% 65-82). According to our analysis, EpiVacCorona proved to be an ineffective vaccine and therefore cannot protect against COVID-19.
Collapse
Affiliation(s)
- Olga Matveeva
- Sendai Viralytics LLC, 23 Nylander Way, Acton, MA 01720, USA
| | - Alexander Ershov
- Medusa Project SIA, Krisjana Barona iela 5-2, LV-1050 Rīga, Latvia;
| |
Collapse
|