101
|
Korosec CS, Farhang-Sardroodi S, Dick DW, Gholami S, Ghaemi MS, Moyles IR, Craig M, Ooi HK, Heffernan JM. Long-term durability of immune responses to the BNT162b2 and mRNA-1273 vaccines based on dosage, age and sex. Sci Rep 2022; 12:21232. [PMID: 36481777 PMCID: PMC9732004 DOI: 10.1038/s41598-022-25134-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
The lipid nanoparticle (LNP)-formulated mRNA vaccines BNT162b2 and mRNA-1273 are a widely adopted multi vaccination public health strategy to manage the COVID-19 pandemic. Clinical trial data has described the immunogenicity of the vaccine, albeit within a limited study time frame. Here, we use a within-host mathematical model for LNP-formulated mRNA vaccines, informed by available clinical trial data from 2020 to September 2021, to project a longer term understanding of immunity as a function of vaccine type, dosage amount, age, and sex. We estimate that two standard doses of either mRNA-1273 or BNT162b2, with dosage times separated by the company-mandated intervals, results in individuals losing more than 99% humoral immunity relative to peak immunity by 8 months following the second dose. We predict that within an 8 month period following dose two (corresponding to the original CDC time-frame for administration of a third dose), there exists a period of time longer than 1 month where an individual has lost more than 99% humoral immunity relative to peak immunity, regardless of which vaccine was administered. We further find that age has a strong influence in maintaining humoral immunity; by 8 months following dose two we predict that individuals aged 18-55 have a four-fold humoral advantage compared to aged 56-70 and 70+ individuals. We find that sex has little effect on the immune response and long-term IgG counts. Finally, we find that humoral immunity generated from two low doses of mRNA-1273 decays at a substantially slower rate relative to peak immunity gained compared to two standard doses of either mRNA-1273 or BNT162b2. Our predictions highlight the importance of the recommended third booster dose in order to maintain elevated levels of antibodies.
Collapse
Affiliation(s)
- Chapin S Korosec
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
| | - Suzan Farhang-Sardroodi
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Department of Mathematics, University of Manitoba, 186 Dysart Road, Winnipeg, MB, R3T 2N2, Canada
| | - David W Dick
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Sameneh Gholami
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Mohammad Sajjad Ghaemi
- Digital Technologies Research Centre, National Research Council Canada, 222 College Street, Toronto, ON, M5T 3J1, Canada
| | - Iain R Moyles
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada
| | - Morgan Craig
- Department of Mathematics and Statistics, Université de Montréal & Sainte-Justine University Hospital Research Centre, 3175, ch. Côte Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Hsu Kiang Ooi
- Digital Technologies Research Centre, National Research Council Canada, 222 College Street, Toronto, ON, M5T 3J1, Canada
| | - Jane M Heffernan
- Modelling Infection and Immunity Lab, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
- Centre for Disease Modelling, Mathematics and Statistics, York University, 4700 Keele St, Toronto, ON, M3J 1P3, Canada.
| |
Collapse
|
102
|
Abufares HI, Oyoun Alsoud L, Alqudah MAY, Shara M, Soares NC, Alzoubi KH, El-Huneidi W, Bustanji Y, Soliman SSM, Semreen MH. COVID-19 Vaccines, Effectiveness, and Immune Responses. Int J Mol Sci 2022; 23:15415. [PMID: 36499742 PMCID: PMC9737588 DOI: 10.3390/ijms232315415] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/24/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
The COVID-19 pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has captivated the globe's attention since its emergence in 2019. This highly infectious, spreadable, and dangerous pathogen has caused health, social, and economic crises. Therefore, a worldwide collaborative effort was made to find an efficient strategy to overcome and develop vaccines. The new vaccines provide an effective immune response that safeguards the community from the virus' severity. WHO has approved nine vaccines for emergency use based on safety and efficacy data collected from various conducted clinical trials. Herein, we review the safety and effectiveness of the WHO-approved COVID-19 vaccines and associated immune responses, and their impact on improving the public's health. Several immunological studies have demonstrated that vaccination dramatically enhances the immune response and reduces the likelihood of future infections in previously infected individuals. However, the type of vaccination and individual health status can significantly affect immune responses. Exposure of healthy individuals to adenovirus vectors or mRNA vaccines causes the early production of antibodies from B and T cells. On the other hand, unhealthy individuals were more likely to experience harmful events due to relapses in their existing conditions. Taken together, aligning with the proper vaccination to a patient's case can result in better outcomes.
Collapse
Affiliation(s)
- Haneen Imad Abufares
- College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Leen Oyoun Alsoud
- College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Mohammad A. Y. Alqudah
- College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Mohd Shara
- College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Nelson C. Soares
- College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Karem H. Alzoubi
- College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Waseem El-Huneidi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Yasser Bustanji
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- College of Medicine, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Sameh S. M. Soliman
- College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Mohammad H. Semreen
- College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| |
Collapse
|
103
|
Bessen C, Plaza-Sirvent C, Simsek A, Bhat J, Marheinecke C, Urlaub D, Bonowitz P, Busse S, Schumann S, Blanco EV, Skaletz-Rorowski A, Brockmeyer NH, Overheu O, Reinacher-Schick A, Faissner S, Watzl C, Pfaender S, Potthoff A, Schmitz I. Impact of SARS-CoV-2 vaccination on systemic immune responses in people living with HIV. Front Immunol 2022; 13:1049070. [PMID: 36532034 PMCID: PMC9755486 DOI: 10.3389/fimmu.2022.1049070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022] Open
Abstract
Despite the development of vaccines, which protect healthy people from severe and life-threatening Covid-19, the immunological responses of people with secondary immunodeficiencies to these vaccines remain incompletely understood. Here, we investigated the humoral and cellular immune responses elicited by mRNA-based SARS-CoV-2 vaccines in a cohort of people living with HIV (PLWH) receiving anti-retroviral therapy. While antibody responses in PLWH increased progressively after each vaccination, they were significantly reduced compared to the HIV-negative control group. This was particularly noteworthy for the Delta and Omicron variants. In contrast, CD4+ Th cell responses exhibited a vaccination-dependent increase, which was comparable in both groups. Interestingly, CD4+ T cell activation negatively correlated with the CD4 to CD8 ratio, indicating that low CD4+ T cell numbers do not necessarily interfere with cellular immune responses. Our data demonstrate that despite the lower CD4+ T cell counts SARS-CoV-2 vaccination results in potent cellular immune responses in PLWH. However, the reduced humoral response also provides strong evidence to consider PLWH as vulnerable group and suggests subsequent vaccinations being required to enhance their protection against COVID-19.
Collapse
Affiliation(s)
- Clara Bessen
- Department of Molecular Immunology, Ruhr University, Bochum, Germany
| | | | - Agit Simsek
- Department of Molecular Immunology, Ruhr University, Bochum, Germany
| | - Jaydeep Bhat
- Department of Molecular Immunology, Ruhr University, Bochum, Germany
| | - Corinna Marheinecke
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Doris Urlaub
- Department for Immunology, Leibniz Research Centre for Working Environment and Human Factors (IfADo) at TU Dortmund, Dortmund, Germany
| | - Petra Bonowitz
- Department of Molecular Immunology, Ruhr University, Bochum, Germany
| | - Sandra Busse
- Department of Molecular Immunology, Ruhr University, Bochum, Germany
| | - Sabrina Schumann
- Department of Molecular Immunology, Ruhr University, Bochum, Germany
| | - Elena Vidal Blanco
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Adriane Skaletz-Rorowski
- WIR - Walk In Ruhr, Center for Sexual Health and Medicine, Bochum, Germany,Department of Dermatology, Venereology and Allergology, Interdisciplinary Immunological Outpatient Clinic, Center for Sexual Health and Medicine, Ruhr-Universität Bochum, Bochum, Germany
| | - Norbert H. Brockmeyer
- WIR - Walk In Ruhr, Center for Sexual Health and Medicine, Bochum, Germany,Department of Dermatology, Venereology and Allergology, Interdisciplinary Immunological Outpatient Clinic, Center for Sexual Health and Medicine, Ruhr-Universität Bochum, Bochum, Germany
| | - Oliver Overheu
- Department of Hematology, Oncology with Palliative Care, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Anke Reinacher-Schick
- Department of Hematology, Oncology with Palliative Care, St. Josef Hospital, Ruhr University Bochum, Bochum, Germany
| | - Simon Faissner
- Department of Neurology, Ruhr-University Bochum, St. Josef Hospital, Bochum, Germany
| | - Carsten Watzl
- Department for Immunology, Leibniz Research Centre for Working Environment and Human Factors (IfADo) at TU Dortmund, Dortmund, Germany
| | - Stephanie Pfaender
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Anja Potthoff
- WIR - Walk In Ruhr, Center for Sexual Health and Medicine, Bochum, Germany,Department of Dermatology, Venereology and Allergology, Interdisciplinary Immunological Outpatient Clinic, Center for Sexual Health and Medicine, Ruhr-Universität Bochum, Bochum, Germany
| | - Ingo Schmitz
- Department of Molecular Immunology, Ruhr University, Bochum, Germany,*Correspondence: Ingo Schmitz,
| |
Collapse
|
104
|
Zhang GF, Meng W, Chen L, Ding L, Feng J, Perez J, Ali A, Sun S, Liu Z, Huang Y, Guo H, Gao S. Neutralizing antibodies to SARS-CoV-2 variants of concern including Delta and Omicron in subjects receiving mRNA-1273, BNT162b2, and Ad26.COV2.S vaccines. J Med Virol 2022; 94:5678-5690. [PMID: 35902378 PMCID: PMC9353285 DOI: 10.1002/jmv.28032] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 07/27/2022] [Indexed: 01/06/2023]
Abstract
SARS-CoV-2 vaccines have contributed to the control of COVID-19 in some parts of the world. However, the constant emergence of variants of concern (VOCs) challenges the effectiveness of SARS-CoV-2 vaccines over time. In particular, Omicron contains a high number of mutations in the spike (S) protein gene, on which most vaccines were developed. In this study, we quantitated neutralizing antibodies in vaccine recipients at various times postvaccination using S protein-based pseudoviruses derived from wild type (WT) SARS-CoV-2 and five VOCs including Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Omicron (B.1.1.529). We found that two-dose mRNA-1273 and BNT162b2 vaccines elicited robust neutralizing antibodies against WT, Alpha, Beta, Gamma, and Delta, but wanned after 6 months with a faster decline observed for BNT162b2. Both mRNA-1273 and BNT162b2 elicited weak neutralizing antibodies against Omicron. One dose of Ad26.COV2.S vaccine induced weaker neutralizing antibodies against WT and most VOCs than mRNA-1273 and BNT162b2 did but moderate neutralizing antibodies against Delta and Omicron, which lasted for 6 months. These results support current recommendations of the Centers for Disease Control and Prevention for a booster 5 months after full immunization with an mRNA-based vaccine and the use of an mRNA-based vaccine 2 months after Ad26.COV2.S vaccination.
Collapse
Affiliation(s)
- George Fei Zhang
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Wen Meng
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Luping Chen
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Ling Ding
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Jian Feng
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Joseph Perez
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Abid Ali
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Shenyu Sun
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Zhentao Liu
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Electrical and Computer EngineeringSwanson School and EngineeringPittsburghPennsylvaniaUSA
| | - Yufei Huang
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of MedicineUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Electrical and Computer EngineeringSwanson School and EngineeringPittsburghPennsylvaniaUSA
| | - Haitao Guo
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Shou‐Jiang Gao
- Cancer Virology Program, UPMC Hillman Cancer CenterUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of Microbiology and Molecular GeneticsUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
105
|
Abstract
The United States Food and Drug Administration recently issued emergency use authorization for 2 mRNA vaccines for preventing COVID-19 disease caused by SARS-CoV-2 virus infections. BNT162b2 from Pfizer-BioNTech and mRNA-1273 by Moderna are planned for use in mass-immunization programs to curb the pandemic. A brief overview of COVID-19 mRNA vaccines is provided, describing the SARS-CoV-2 RNA, how mRNA vaccines work and the advantages of mRNA over other vaccine platforms. The Pfizer-BioNTech collaboration journey to short-list mRNA vaccine candidates and finally selecting BNT162b2 based on safety data is outlined, followed by the Phase 3 study of BNT162b2 demonstrating 95% efficacy in preventing COVID-19 infections. Studies regarding mRNA-1273 (Moderna) are described, including extended immunogenicity data up to 119 days. The Phase 3 COVE study of mRNA-1273 eventually showed vaccine efficacy of 94.5%. Recommendations for future mRNA vaccine development are provided, including ongoing safety surveillance, evaluation in under-represented groups in previous studies and improving mRNA vaccine thermostability. Finally, further logistical considerations are required for manufacturing, storing, distribution and implementing mass vaccination programs to curb the pandemic.
Collapse
Affiliation(s)
- Shyh Poh Teo
- Department of Internal Medicine, Raja Isteri Pengiran Anak Saleha (RIPAS) Hospital, Bandar Seri Begawan, Brunei Darussalam
| |
Collapse
|
106
|
Wu Q, Shen Y, Xie L, Zhang J, Qiu Z, Tang M, Chen L. Low acceptance rate of COVID-19 vaccination and reduced quality of life among heart transplant recipients during the COVID-19 pandemic. J Card Surg 2022; 37:4975-4981. [PMID: 36444896 PMCID: PMC9877912 DOI: 10.1111/jocs.17205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 10/27/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To investigate the impact of the current coronavirus disease 2019 (COVID-19) pandemic on the quality of life (QoL) and status of COVID-19 vaccination in heart transplant recipients (HTRs). METHODS Patients who underwent allogeneic heart transplants between June 2006 and December 2019, who survived were selected from a follow-up registration form at our center. Data were collected using questionnaires in 2021, the QoL survey was conducted using the MOS 36-item Short-Form Health Survey (SF-36) and compared to the same time frame in 2019. The patients were divided into two groups: post-epidemic (A) and pre-epidemic (B) groups. We also recorded whether the participants had been vaccinated against COVID-19 (Beijing Sinovac COVID-19 vaccine). All the data obtained were analyzed. RESULTS There were 88 patients who participated in the study. Only 12 (13.6%) were vaccinated. In terms of SF-36 scale assessments, after the outbreak of the COVID-19 pandemic, Group A scored lower in vitality [52.5(49.0, 58.0) vs. 75.0(69.0, 79.0), p < .001], social functioning [54.0(50.5, 58.0) vs. 74.0(67.5, 78.0), p < .001], role emotional [58.5(55.0, 62.0) vs. 67.0(63.0, 71.0), p < .001], and mental health [58.5(55.0, 62.0) vs. 76.0(72.0, 79.0), p < .001]. In Group A the mental component summary (MCS) significantly decreased [222.0(214.5, 230.0) vs. 289.0(277.5, 299.5), p < .001]. The PCS and MCS of HTRs who had been vaccinated against COVID-19 were significantly higher than those who had not [PCS: 283.5(280.0, 287.0) vs. 276.0(271.0, 279.0), p < .001; MCS: 245.0(141.5, 254.0) vs. 220.0(213.5, 226.5), p < .001]. CONCLUSION Low acceptance levels of COVID-19 vaccination were observed in the HTRs. The QoL of the HTRs decreased after the COVID-19 pandemic.
Collapse
Affiliation(s)
- Qingsong Wu
- Department of Cardiovascular Surgery, Union HospitalFujian Medical UniversityFujianFuzhouP. R. China,Key Laboratory of Cardio‐Thoracic Surgery (Fujian Medical University)Fujian Province UniversityFuzhouFujianP. R. China,Fujian Provincial Special Reserve Talents LaboratoryFujianFuzhouP. R. China
| | - Yue Shen
- Department of Cardiovascular Surgery, Union HospitalFujian Medical UniversityFujianFuzhouP. R. China,Key Laboratory of Cardio‐Thoracic Surgery (Fujian Medical University)Fujian Province UniversityFuzhouFujianP. R. China,Fujian Provincial Special Reserve Talents LaboratoryFujianFuzhouP. R. China
| | - Linfeng Xie
- College of Clinical Medicine, Union HospitalFujian Medical UniversityFujianFuzhouP. R. China
| | - Jiaxin Zhang
- Department of Cardiovascular Surgery, Union HospitalFujian Medical UniversityFujianFuzhouP. R. China
| | - Zhihuang Qiu
- Department of Cardiovascular Surgery, Union HospitalFujian Medical UniversityFujianFuzhouP. R. China,Key Laboratory of Cardio‐Thoracic Surgery (Fujian Medical University)Fujian Province UniversityFuzhouFujianP. R. China,Fujian Provincial Special Reserve Talents LaboratoryFujianFuzhouP. R. China
| | - Mirong Tang
- Department of Cardiovascular Surgery, Union HospitalFujian Medical UniversityFujianFuzhouP. R. China
| | - Liangwan Chen
- Department of Cardiovascular Surgery, Union HospitalFujian Medical UniversityFujianFuzhouP. R. China,Key Laboratory of Cardio‐Thoracic Surgery (Fujian Medical University)Fujian Province UniversityFuzhouFujianP. R. China,Fujian Provincial Special Reserve Talents LaboratoryFujianFuzhouP. R. China
| |
Collapse
|
107
|
Yamanaka Y, Yokota I, Yasumoto A, Morishita E, Horiuchi H. Time of Day of Vaccination Does Not Associate With SARS-CoV-2 Antibody Titer Following First Dose of mRNA COVID-19 Vaccine. J Biol Rhythms 2022; 37:700-706. [PMID: 36154515 PMCID: PMC9726636 DOI: 10.1177/07487304221124661] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The immune system exhibits circadian rhythms, and its response to viral infection is influenced by the circadian clock system. Previous studies have reported associations between the time of day of vaccination against COVID-19 and production of anti-SARS-CoV-2 antibody titer. We examined the effect of vaccination time of day on anti-SARS-CoV-2 antibody titer after the first dose of vaccination with the mRNA-1273 (Moderna) COVID-19 vaccine in an adult population. A total of 332 Japanese adults participated in the present study. All participants were not infected with SARS-CoV-2 and had already received the first dose of mRNA-1273 2 to 4 weeks prior to participating in the study. The participants were asked to provide basic demographic characteristics (age, sex, medical history, allergy, medication, and mean sleep duration), the number of days after the first dose of vaccination, and the time of day of vaccination. Blood was collected from the participants, and SARS-CoV-2 antibody titers were measured. Ordinary least square regression was used for assessing the relationship between basic demographic characteristics, number of days after vaccination, time of day of vaccination, and the log10-transformed normalized antibody titer. The least square mean of antibody titers was not associated with the vaccination time and sleep durations. The least square means of antibody titers was associated with age; the antibody titers decreased in people aged 50 to 59 years and 60 to 64 years. The present findings demonstrate that the vaccination time with mRNA-1273 was not associated with the SARS-CoV-2 antibody titer in an adult population, suggesting that these results do not support restricting vaccination to a particular time of day. The present findings may be useful in optimizing SARS-CoV-2 vaccination strategies.
Collapse
Affiliation(s)
- Yujiro Yamanaka
- Laboratory of Life and Health Sciences, Graduate School of Education and Faculty of Education, Hokkaido University, Sapporo, Japan,Research and Education Center for Brain Science, Hokkaido University, Sapporo, Japan,Yujiro Yamanaka, Laboratory of Life and Health Sciences, Graduate School of Education and Faculty of Education, Hokkaido University, North-11, West-7, Kita-Ku, Sapporo 060-0811, Japan; e-mail:
| | - Isao Yokota
- Department of Biostatistics, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Atsushi Yasumoto
- Division of Laboratory and Transfusion Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Eriko Morishita
- Department of Hematology, Kanazawa University Hospital, Kanazawa, Japan
| | - Hisanori Horiuchi
- Department of Molecular and Cellular Biology, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| |
Collapse
|
108
|
Nanishi E, Levy O, Ozonoff A. Waning effectiveness of SARS-CoV-2 mRNA vaccines in older adults: a rapid review. Hum Vaccin Immunother 2022; 18:2045857. [PMID: 35240940 PMCID: PMC9196671 DOI: 10.1080/21645515.2022.2045857] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/07/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
The U.S. Centers for Disease Control and Prevention (CDC) and other health agencies have recently recommended a booster dose of COVID-19 vaccines for specific vulnerable groups including adults 65 years and older. There is limited evidence whether vaccine effectiveness (VE) in older adults decreases over time, especially against severe COVID-19. We performed a rapid review of published studies available through 4 November 2021 that provide effectiveness data on messenger RNA (mRNA) vaccines approved/licensed in the United States and identified eight eligible studies which evaluated VE in older adults. There is evidence of a decline in VE against both severe acute respiratory syndrome coronavirus 2 infection and severe COVID-19 in older adults among studies which analyzed data up to July-October 2021. Our findings suggest that VE diminishes in older adults, which supports the current recommendation for a booster dose in this population.
Collapse
Affiliation(s)
- Etsuro Nanishi
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| | - Al Ozonoff
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT & Harvard, Cambridge, MA, USA
| |
Collapse
|
109
|
Mohseni Afshar Z, Barary M, Hosseinzadeh R, Alijanpour A, Hosseinzadeh D, Ebrahimpour S, Nazary K, Sio TT, Sullman MJM, Carson-Chahhoud K, Babazadeh A. Breakthrough SARS-CoV-2 infections after vaccination: a critical review. Hum Vaccin Immunother 2022; 18:2051412. [PMID: 35302905 PMCID: PMC9115792 DOI: 10.1080/21645515.2022.2051412] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 03/06/2022] [Indexed: 12/28/2022] Open
Abstract
At the beginning of the current pandemic, it was believed that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection would induce lifelong immunity and that reinfections would be unlikely. However, after several cases of reinfection were documented in previously infected patients, this was understood to be a false assumption, and this waning humoral immunity has raised significant concerns. Accordingly, long-term and durable vaccine-induced antibody protection against infection have also become a challenge, as several breakthroughs of COVID-19 infection have been identified in individuals who were fully vaccinated. This review discusses the current evidence on breakthrough COVID-19 infections occurring after vaccination.
Collapse
Affiliation(s)
- Zeinab Mohseni Afshar
- Clinical Research Development Center, Imam Reza Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Barary
- Student Research Committee, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Students’ Scientific Research Center (SSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Rezvan Hosseinzadeh
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | | | | | - Soheil Ebrahimpour
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Kosar Nazary
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Terence T. Sio
- Department of Radiation Oncology, Mayo Clinic, Phoenix, Arizona, USA
| | - Mark J. M. Sullman
- Department of Social Sciences, University of Nicosia, Nicosia, Cyprus
- Department of Life and Health Sciences, University of Nicosia, Nicosia, Cyprus
| | | | - Arefeh Babazadeh
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
110
|
Omma A, Batirel A, Aydin M, Yilmaz Karadag F, Erden A, Kucuksahin O, Armagan B, Güven SC, Karakas O, Gokdemir S, Altunal LN, Buber AA, Gemcioglu E, Zengin O, Inan O, Sahiner ES, Korukluoglu G, Sezer Z, Ozdarendeli A, Kara A, Ates I. Safety and immunogenicity of inactive vaccines as booster doses for COVID-19 in Türkiye: A randomized trial. Hum Vaccin Immunother 2022; 18:2122503. [PMID: 36315843 PMCID: PMC9746394 DOI: 10.1080/21645515.2022.2122503] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Protective neutralizing antibody titers reduce in time after COVID-19 vaccinations, as in individuals who have had COVID-19. This study aimed to evaluate the safety and immunogenicity of CoronaVac and TURKOVAC vaccines used as a booster dose after CoronaVac primary vaccination. This double-blind, randomized, controlled, phase II, multicenter study included healthy male and female adults (18-60 years) who were vaccinated with two doses of CoronaVac vaccine and did not exceed the duration of at least 90 days and a maximum of 270 days from the second dose of vaccination. Among 236 eligible volunteers, 222 were recruited for randomization between July 12, 2021 and September 10, 2021; 108 and 114 were randomized to the TURKOVAC and CoronaVac arms, respectively. The primary endpoint was adverse events (AEs) (ClinicalTrials.gov; Identifier: NCT04979949). On day 28, at the neutralizing antibody threshold of 1/6, the positivity rate reached 100% from 46.2% to 98.2% from 52.6% in the TURKOVAC and CoronaVac arms, respectively, against the Wuhan variant and the positivity rate reached 80.6% from 8.7% in the TURKOVAC arm vs. 71.9% from 14.0% in the CoronaVac arm against the Delta variant. IgG spike antibody positivity rate increased from 57.3% to 98.1% and from 57.9% to 97.4% in the TURKOVAC and CoronaVac arms, respectively. The TURKOVAC and CoronaVac arms were comparable regarding the frequency of overall AEs. Both vaccines administered as booster yielded higher antibody titers with acceptable safety profiles.
Collapse
Affiliation(s)
- Ahmet Omma
- Clinic of Rheumatology, Ankara City Hospital, Ankara, Türkiye,CONTACT Ahmet Omma Clinic of Rheumatology, Ankara City Hospital, Ankara06800, Turkey
| | - Ayse Batirel
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, International Medical School, Kartal Dr. Lutfi Kirdar City Hospital, Istanbul, Türkiye
| | - Mehtap Aydin
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Umraniye Training and Research Hospital, Istanbul, Türkiye
| | - Fatma Yilmaz Karadag
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Sancaktepe Sehit Prof. Dr. Ilhan Varank Training and Research Hospital, Istanbul, Türkiye
| | | | - Orhan Kucuksahin
- Department of Internal Medicine, Division of Rheumatology, Yıldırım Beyazıt University School of Medicine, Ankara, Türkiye
| | - Berkan Armagan
- Clinic of Rheumatology, Ankara City Hospital, Ankara, Türkiye
| | | | - Ozlem Karakas
- Clinic of Rheumatology, Ankara City Hospital, Ankara, Türkiye
| | - Selim Gokdemir
- Department of Clinical Pharmacology, University of Health Sciences, Kartal Dr. Lutfi Kirdar City Hospital, Istanbul, Türkiye
| | - Lutfiye Nilsun Altunal
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Umraniye Training and Research Hospital, Istanbul, Türkiye
| | - Aslihan Ayse Buber
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Sancaktepe Sehit Prof. Dr. Ilhan Varank Training and Research Hospital, Istanbul, Türkiye
| | - Emin Gemcioglu
- Department of Internal Medicine, University of Health Sciences Ankara City Hospital, Ankara, Türkiye
| | - Oguzhan Zengin
- Department of Internal Medicine, University of Health Sciences Ankara City Hospital, Ankara, Türkiye
| | - Osman Inan
- Department of Internal Medicine, University of Health Sciences Ankara City Hospital, Ankara, Türkiye
| | - Enes Seyda Sahiner
- Department of Internal Medicine, University of Health Sciences Ankara City Hospital, Ankara, Türkiye
| | - Gulay Korukluoglu
- National Virology Reference Laboratory, General Directorate of Public Health, Ankara, Türkiye
| | - Zafer Sezer
- Department of Pharmacology, Medical Faculty, Erciyes University, Kayseri, Türkiye
| | - Aykut Ozdarendeli
- Vaccine Research, Development and Application Center, Erciyes University, Kayseri, Türkiye,Department of Microbiology, Medical Faculty, Erciyes University, Kayseri, Türkiye
| | - Ates Kara
- Division of Pediatric Infectious Diseases, Department of Pediatrics, Hacettepe University, Ankara, Türkiye,Head of Türkiye Vaccine Institute, Ankara, Türkiye
| | - Ihsan Ates
- Department of Internal Medicine, University of Health Sciences Ankara City Hospital, Ankara, Türkiye
| |
Collapse
|
111
|
Jamshidi E, Asgary A, Shafiekhani P, Khajeamiri Y, Mohamed K, Esmaily H, Jamal Rahi S, Mansouri N. Longevity of immunity following COVID-19 vaccination: a comprehensive review of the currently approved vaccines. Hum Vaccin Immunother 2022; 18:2037384. [PMID: 35417285 PMCID: PMC9115789 DOI: 10.1080/21645515.2022.2037384] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 01/31/2022] [Indexed: 11/04/2022] Open
Abstract
It is unknown how long the immunity following COVID-19 vaccination lasts. The current systematic review provides a perspective on the persistence of various antibodies for available vaccines.Both the BNT162b2 and the mRNA-1273 induce the production of IgA antibodies, reflecting the possible prevention of the asymptomatic spread. The mRNA-1273 vaccine's antibodies were detectable until 6 months, followed by the AZD1222, 3 months, the Ad26.COV2.S and the BNT162b2 vaccines within 2 months.The BNT162b2 produced anti-spike IgGs 11 days after the first dose and peaked at day 21, whereas the AZD1222 induced a neutralizing effect 22 days after the first dose.These vaccines induce T-cell mediated immune responses too. Each one of the AZD1222, Ad26.COV2.S, mRNA-1273 mediates T-cell response immunity at days 14-22, 15, and 43 after the first dose, respectively. Whereas for the BNT162b1 and BNT162b2 vaccines, T-cell immunity is induced 7 days and 12 weeks after the booster dose, respectively.
Collapse
Affiliation(s)
- Elham Jamshidi
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhossein Asgary
- Department of Biotechnology, College of Sciences, University of Tehran, Tehran, Iran
| | - Paria Shafiekhani
- USERN office, Functional Neurosurgery Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Yasaman Khajeamiri
- School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kawthar Mohamed
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Manama, Bahrain
| | - Hadi Esmaily
- School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahand Jamal Rahi
- Laboratory of the Physics of Biological Systems, Institute of Physics, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Nahal Mansouri
- Division of Pulmonary Medicine, Department of Medicine, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
- Research Group on Artificial Intelligence in Pulmonary Medicine, Division of Pulmonary Medicine, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, École polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland
| |
Collapse
|
112
|
Abullais SS, Arora S, Al Shahrani M, Khan AA, Al Shahrani W, Mahmood SE, Al Qahtani S, Maqbool M, Saib Jameel A, Saluja P. Knowledge, perception, and acceptance toward the booster dose of COVID-19 vaccine among patients visiting dental clinics in Aseer region of KSA. Hum Vaccin Immunother 2022; 18:2095162. [PMID: 35856819 DOI: 10.1080/21645515.2022.2095162] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Scientists have emerged with innovative research on non-human primates showing that the booster dose of the COVID-19 vaccine increases neutralizing antibody levels against all variants. The current cross-sectional survey was designed to evaluate the knowledge, perception, and acceptance of the booster dose of the COVID-19 vaccine among the patients visiting the various dental clinics in Aseer region, Kingdom of Saudi Arabia. A total of 609 dental patients were selected from various dental clinics by a simple random probability sampling method. The questionnaire was designed in multiple languages and categorized as demographic information, knowledge, perception, and acceptance of participants to a booster dose. An anonymous, self-administered, closed-ended online, and paper-based questionnaire was used to assess the above parameters. In the current survey, the majority of the participants were Saudis (80.8%) with an age mean of 37.7 ± 8.7 years. About 68.6% (418 out of 609) of participants had poor knowledge about the booster dose. Significant differences in the levels of knowledge were found in relation to gender, area of residency, education, nationality, and occupation. The distribution of level of perception of booster dose differs significantly among participants of different marital statuses and nationalities. Hesitation to booster was reported more in the rural than in the urban population. Despite a low level of knowledge, a higher level of good perception and acceptance of booster doses were reported among the studied population.
Collapse
Affiliation(s)
- Shahabe Saquib Abullais
- Department of Periodontics and Community Dental Sciences, College of Dentistry, King Khalid University, Abha, KSA
| | - Suraj Arora
- Department of Restorative Dentistry, College of Dentistry, King Khalid University, Abha, KSA
| | | | - Abdul Ahad Khan
- Department of Oral and Maxillofacial Surgery, College of Dentistry, King Khalid University, Abha, KSA
| | | | - Syed Esam Mahmood
- Department of Family and Community Medicine, College of Medicine, King Khalid University, Abha, KSA
| | - Saad Al Qahtani
- Department of Periodontics and Community Dental Sciences, College of Dentistry, King Khalid University, Abha, KSA
| | - Mohammed Maqbool
- Department of Periodontics and Community Dental Sciences, College of Dentistry, King Khalid University, Abha, KSA
| | - Ahmad Saib Jameel
- Department of Prosthetic Dental Sciences, College of Dentistry, Jazan University, Jazan, KSA
| | - Priyanka Saluja
- Department of Conservative Dentistry & Endodontics, JCD Dental College, Sirsa, India
| |
Collapse
|
113
|
Neutralizing antibodies against SARS-CoV-2 variants following mRNA booster vaccination in adults older than 65 years. Sci Rep 2022; 12:20373. [PMID: 36437298 PMCID: PMC9701678 DOI: 10.1038/s41598-022-24409-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
Immune response induced by COVID-19 vaccine booster against delta and omicron variants was assessed in 65 adults (65-84 years old) early aftesr a first booster dose. An increase in SARS-CoV-2 neutralizing antibodies was shown in individuals not previously infected without evidence of an age-related effect, with lower increase in those infected before a single dose of primary vaccination. Of note, humoral response was observed only starting from the 5th day after the boost.
Collapse
|
114
|
Valenzuela-Fernández A, Cabrera-Rodriguez R, Ciuffreda L, Perez-Yanes S, Estevez-Herrera J, González-Montelongo R, Alcoba-Florez J, Trujillo-González R, García-Martínez de Artola D, Gil-Campesino H, Díez-Gil O, Lorenzo-Salazar JM, Flores C, Garcia-Luis J. Nanomaterials to combat SARS-CoV-2: Strategies to prevent, diagnose and treat COVID-19. Front Bioeng Biotechnol 2022; 10:1052436. [PMID: 36507266 PMCID: PMC9732709 DOI: 10.3389/fbioe.2022.1052436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the associated coronavirus disease 2019 (COVID-19), which severely affect the respiratory system and several organs and tissues, and may lead to death, have shown how science can respond when challenged by a global emergency, offering as a response a myriad of rapid technological developments. Development of vaccines at lightning speed is one of them. SARS-CoV-2 outbreaks have stressed healthcare systems, questioning patients care by using standard non-adapted therapies and diagnostic tools. In this scenario, nanotechnology has offered new tools, techniques and opportunities for prevention, for rapid, accurate and sensitive diagnosis and treatment of COVID-19. In this review, we focus on the nanotechnological applications and nano-based materials (i.e., personal protective equipment) to combat SARS-CoV-2 transmission, infection, organ damage and for the development of new tools for virosurveillance, diagnose and immune protection by mRNA and other nano-based vaccines. All the nano-based developed tools have allowed a historical, unprecedented, real time epidemiological surveillance and diagnosis of SARS-CoV-2 infection, at community and international levels. The nano-based technology has help to predict and detect how this Sarbecovirus is mutating and the severity of the associated COVID-19 disease, thereby assisting the administration and public health services to make decisions and measures for preparedness against the emerging variants of SARS-CoV-2 and severe or lethal COVID-19.
Collapse
Affiliation(s)
- Agustín Valenzuela-Fernández
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Romina Cabrera-Rodriguez
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Laura Ciuffreda
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Silvia Perez-Yanes
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | - Judith Estevez-Herrera
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| | | | - Julia Alcoba-Florez
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Rodrigo Trujillo-González
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
- Departamento de Análisis Matemático, Facultad de Ciencias, Universidad de La Laguna, Santa Cruz de Tenerife, Spain
| | | | - Helena Gil-Campesino
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - Oscar Díez-Gil
- Servicio de Microbiología, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
| | - José M. Lorenzo-Salazar
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
| | - Carlos Flores
- Research Unit, Hospital Universitario N. S. de Candelaria, Santa Cruz de Tenerife, Spain
- Genomics Division, Instituto Tecnológico y de Energías Renovables, Santa Cruz de Tenerife, Spain
- CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Faculty of Health Sciences, University of Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain
| | - Jonay Garcia-Luis
- Laboratorio de Inmunología Celular y Viral, Unidad de Farmacología, Sección de Medicina, Facultad de Ciencias de la Salud, Universidad de La Laguna, San Cristóbal de La Laguna, Spain
| |
Collapse
|
115
|
Shi Y, Huang J, Liu Y, Liu J, Guo X, Li J, Gong L, Zhou X, Cheng G, Qiu Y, You J, Lou Y. Structural and biochemical characteristics of mRNA nanoparticles determine anti-SARS-CoV-2 humoral and cellular immune responses. SCIENCE ADVANCES 2022; 8:eabo1827. [PMID: 36417530 PMCID: PMC9683711 DOI: 10.1126/sciadv.abo1827] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic underlines the urgent need for effective mRNA vaccines. However, current understanding of the immunological outcomes of mRNA vaccines formulated under different nanoplatforms is insufficient. Here, severe acute respiratory syndrome coronavirus 2 receptor binding domain mRNA delivered via lipid nanoparticle (LNP), cationic nanoemulsion (CNE), and cationic liposome (Lipo) was constructed. Results demonstrated that the structural and biochemical characteristics of nanoparticles shaped their tissue dissemination, cellular uptake, and intracellular trafficking, which eventually determined the activation of antiviral humoral and cellular immunity. Specifically, LNP was mainly internalized by myocyte and subsequently circumvented lysosome degradation, giving rise to humoral-biased immune responses. Meanwhile, CNE and Lipo induced cellular-preferred immunity, which was respectively attributed to the better lysosomal escape in dendritic cells and the superior biodistribution in secondary lymphoid organs. Overall, this study may guide the design and clinical use of mRNA vaccines against COVID-19.
Collapse
Affiliation(s)
- Yingying Shi
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, People’s Republic of China
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People’s Republic of China
| | - Jiaxin Huang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People’s Republic of China
| | - Yu Liu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People’s Republic of China
| | - Jing Liu
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, People’s Republic of China
| | - Xuemeng Guo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People’s Republic of China
| | - Jianhua Li
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Binsheng Road, Hangzhou, Zhejiang 310051, People’s Republic of China
| | - Liming Gong
- Zhejiang Provincial Center for Disease Control and Prevention, 3399 Binsheng Road, Hangzhou, Zhejiang 310051, People’s Republic of China
| | - Xin Zhou
- Ausper Biopharma Inc., 688 Bin’an Road, Hangzhou, Zhejiang 310051, People’s Republic of China
| | - Guofeng Cheng
- Ausper Biopharma Inc., 688 Bin’an Road, Hangzhou, Zhejiang 310051, People’s Republic of China
| | - Yunqing Qiu
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, People’s Republic of China
| | - Jian You
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, People’s Republic of China
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, People’s Republic of China
| | - Yan Lou
- Zhejiang Provincial Key Laboratory for Drug Clinical Research and Evaluation, Department of Clinical Pharmacy, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang 310000, People’s Republic of China
| |
Collapse
|
116
|
Chao CH, Cheng D, Huang SW, Chuang YC, Yeh TM, Wang JR. Serological responses triggered by different SARS-CoV-2 vaccines against SARS-CoV-2 variants in Taiwan. Front Immunol 2022; 13:1023943. [PMID: 36458016 PMCID: PMC9705976 DOI: 10.3389/fimmu.2022.1023943] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/31/2022] [Indexed: 09/05/2023] Open
Abstract
Broadly neutralizing ability is critical for developing the next-generation SARS-CoV-2 vaccine. We collected sera samples between December 2021-January 2022 from 113 Taiwan naïve participants after their second dose of homologous vaccine (AZD1222, mRNA-1273, BNT162-b2, and MVC-COV1901) and compared the differences in serological responses of various SARS-CoV-2 vaccines. Compared to AZD1222, the two mRNA vaccines could elicit a higher level of anti-S1-RBD binding antibodies with higher broadly neutralizing ability evaluated using pseudoviruses of various SARS-CoV-2 lineages. The antigenic maps produced from the neutralization data implied that Omicron represents very different antigenic characteristics from the ancestral lineage. These results suggested that constantly administering the vaccine with ancestral Wuhan spike is insufficient for the Omicron outbreak. In addition, we found that anti-ACE2 autoantibodies were significantly increased in all four vaccinated groups compared to the unvaccinated pre-pandemic group, which needed to be investigated in the future.
Collapse
Affiliation(s)
- Chiao-Hsuan Chao
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Dayna Cheng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Wen Huang
- National Mosquito-Borne Diseases Control Research Center, National Health Research Institutes, Tainan, Taiwan
| | | | - Trai-Ming Yeh
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
| | - Jen-Ren Wang
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Tainan, Taiwan
| |
Collapse
|
117
|
Torres M, Corona M, Rodríguez-Mora S, Casado-Fernández G, Zurdo-Castronuño A, Mateos E, Ramos-Martín F, Sánchez-Menéndez C, Murciano-Antón MA, García-Pérez J, Alcamí J, Pérez-Olmeda M, Coiras M, López-Jiménez J, García-Gutiérrez V. Strong Humoral but Not Cellular Immune Responses against SARS-CoV-2 in Individuals with Oncohematological Disease Who Were Treated with Rituximab before Receiving a Vaccine Booster. Cancers (Basel) 2022; 14:5537. [PMID: 36428631 PMCID: PMC9688562 DOI: 10.3390/cancers14225537] [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: 09/24/2022] [Revised: 11/03/2022] [Accepted: 11/06/2022] [Indexed: 11/12/2022] Open
Abstract
The humoral immune response developed after receiving the full vaccination schedule against COVID-19 is impaired in individuals who received anti-CD20 therapy 6-9 months before vaccination. However, there is little information about the cellular immune responses elicited in these individuals. In this study, we analyzed the humoral and cellular immune responses in 18 individuals with hematological disease who received the last dose of rituximab 13.8 months (IQR 9.4-19) before the booster dose. One month after receiving the booster dose, the seroconversion rate in the rituximab-treated cohort increased from 83.3% to 88.9% and titers of specific IgGs against SARS-CoV-2 increased 1.53-fold (p = 0.0098), while the levels of neutralizing antibodies increased 3.03-fold (p = 0.0381). However, the cytotoxic activity of peripheral blood mononuclear cells (PBMCs) from rituximab-treated individuals remained unchanged, and both antibody-dependent cellular cytotoxicity (ADCC) and direct cellular cytotoxicity (CDD) were reduced 1.7-fold (p = 0.0047) and 2.0-fold (p = 0.0086), respectively, in comparison with healthy donors. Breakthrough infections rate was higher in our cohort of rituximab-treated individuals (33.33%), although most of the infected patients (83.4%) developed a mild form of COVID-19. In conclusion, our findings confirm a benefit in the humoral, but not in the cellular, immune response in rituximab-treated individuals after receiving a booster dose of an mRNA-based vaccine against COVID-19.
Collapse
Affiliation(s)
- Montserrat Torres
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Magdalena Corona
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
- Faculty of Sciences, Universidad de Alcalá, 28871 Alcalá de Henares, Spain
| | - Sara Rodríguez-Mora
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | - Guiomar Casado-Fernández
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Faculty of Sciences, Universidad de Alcalá, 28871 Alcalá de Henares, Spain
| | - Alejandro Zurdo-Castronuño
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Elena Mateos
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | - Fernando Ramos-Martín
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | - Clara Sánchez-Menéndez
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | | | - Javier García-Pérez
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28222 Majadahonda, Spain
| | - José Alcamí
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- AIDS Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28222 Majadahonda, Spain
| | - Mayte Pérez-Olmeda
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Serology Service, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mayte Coiras
- Immunopathology Unit, National Center of Microbiology, Instituto de Salud Carlos III, 28220 Majadahonda, Spain
- Biomedical Research Center Network in Infectious Diseases (CIBERINFEC), Instituto de Salud Carlos III, 28220 Majadahonda, Spain
| | - Javier López-Jiménez
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Valentín García-Gutiérrez
- Hematology and Hemotherapy Service, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | | |
Collapse
|
118
|
Corrao G, Franchi M, Cereda D, Bortolan F, Leoni O, Jara J, Valenti G, Pavesi G. Factors associated with severe or fatal clinical manifestations of SARS-CoV-2 infection after receiving the third dose of vaccine. J Intern Med 2022; 292:829-836. [PMID: 35943414 PMCID: PMC9539163 DOI: 10.1111/joim.13551] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Little is known about vulnerability to severe COVID-19 illness after vaccination completion with three doses of vaccine against COVID-19. OBJECTIVES To identify individual features associated with increased risk of severe clinical manifestation of SARS-CoV-2 infections after receiving the third dose of vaccine against COVID-19. METHODS We performed a nested case-control study based on 3,360,116 citizens from Lombardy, Italy, aged 12 years or older who received the third dose of vaccine against COVID-19 from 20 September through 31 December 2021. Individuals were followed from 14 days after vaccination completion until the occurrence of severe COVID-19 illness, death unrelated to COVID-19, emigration or 15 March 2022. For each case, controls were randomly selected to be 1:10 matched for the date of vaccination completion and municipality of residence. The association between candidate predictors and outcome was assessed through multivariable conditional logistic regression models. RESULTS During 12,538,330 person-months of follow-up, 5171 cases of severe illness occurred. As age increased, a trend towards increasing odds of severe illness was observed. Male gender was a significant risk factor. As the number of contacts with the Regional Health Service increased, a trend towards increasing odds of severe illness was observed. Having had a previous SARS-CoV-2 infection was a significant protective factor. Having received the Moderna vaccine significantly decreased the odds of severe illness. Significant higher odds were associated with 42 diseases/conditions. Odds ratios ranged from 1.23 (diseases of the musculoskeletal system) to 5.00 (autoimmune disease). CONCLUSIONS This study provides useful insights for establishing priority in fourth-dose vaccination programs.
Collapse
Affiliation(s)
- Giovanni Corrao
- National Centre for Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, Milan, Italy.,Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy.,Directorate General for Health, Lombardy Region, Milan, Italy
| | - Matteo Franchi
- National Centre for Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, Milan, Italy.,Unit of Biostatistics, Epidemiology and Public Health, Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy
| | - Danilo Cereda
- Directorate General for Health, Lombardy Region, Milan, Italy
| | | | - Olivia Leoni
- Directorate General for Health, Lombardy Region, Milan, Italy
| | | | | | - Giovanni Pavesi
- Directorate General for Health, Lombardy Region, Milan, Italy
| |
Collapse
|
119
|
Syversen SW, Jyssum I, Tveter AT, Sexton J, Christensen IE, Tran TT, Bjørlykke KH, Mjaaland S, Warren DJ, Kvien TK, Chopra A, Kro GB, Jahnsen J, Munthe LA, Haavardsholm EA, Grødeland G, Vaage JT, Provan SA, Jørgensen KK, Goll GL. Immunogenicity and safety of a three-dose SARS-CoV-2 vaccination strategy in patients with immune-mediated inflammatory diseases on immunosuppressive therapy. RMD Open 2022; 8:rmdopen-2022-002417. [PMID: 36328399 PMCID: PMC9638754 DOI: 10.1136/rmdopen-2022-002417] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 09/28/2022] [Indexed: 11/05/2022] Open
Abstract
Objectives Humoral vaccine responses to SARS-CoV-2 vaccines are impaired and short lasting in patients with immune-mediated inflammatory diseases (IMID) following two vaccine doses. To protect these vulnerable patients against severe COVID-19 disease, a three-dose primary vaccination strategy has been implemented in many countries. The aim of this study was to evaluate humoral response and safety of primary vaccination with three doses in patients with IMID. Methods Patients with IMID on immunosuppressive therapy and healthy controls receiving three-dose and two-dose primary SARS-CoV-2 vaccination, respectively, were included in this prospective observational cohort study. Anti-Spike antibodies were assessed 2–4 weeks, and 12 weeks following each dose. The main outcome was anti-Spike antibody levels 2–4 weeks following three doses in patients with IMID and two doses in controls. Additional outcomes were the antibody decline rate and adverse events. Results 1100 patients and 303 controls were included. Following three-dose vaccination, patients achieved median (IQR) antibody levels of 5720 BAU/mL (2138–8732) compared with 4495 (1591–6639) in controls receiving two doses, p=0.27. Anti-Spike antibody levels increased with median 1932 BAU/mL (IQR 150–4978) after the third dose. The interval between the vaccine doses and vaccination with mRNA-1273 or a combination of vaccines were associated with antibody levels following the third dose. Antibody levels had a slower decline-rate following the third than the second vaccine dose, p<0.001. Adverse events were reported by 464 (47%) patients and by 196 (78%) controls. Disease flares were reported by 70 (7%) patients. Conclusions This study shows that additional vaccine doses to patients with IMID contribute to strong and sustained immune-responses comparable to healthy persons vaccinated twice, and supports repeated vaccination of patients with IMID. Trial registration number NCT04798625.
Collapse
Affiliation(s)
- Silje Watterdal Syversen
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Ingrid Jyssum
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Anne Therese Tveter
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Joe Sexton
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | - Ingrid Egeland Christensen
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Trung T Tran
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Kristin Hammersbøen Bjørlykke
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | | | - David J Warren
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Tore K Kvien
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Adity Chopra
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | | | - Jorgen Jahnsen
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Ludvig A Munthe
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B cell Malignancies, University of Oslo, Oslo, Norway
| | - Espen A Haavardsholm
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gunnveig Grødeland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - John Torgils Vaage
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Sella Aarrestad Provan
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| | | | - Guro Løvik Goll
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, Oslo, Norway
| |
Collapse
|
120
|
Ponnampalli S, Venkata Suryanarayana Birudukota N, Kamal A. COVID-19: Vaccines and therapeutics. Bioorg Med Chem Lett 2022; 75:128987. [PMID: 36113669 PMCID: PMC9472710 DOI: 10.1016/j.bmcl.2022.128987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 12/15/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a communicable disease triggered by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), emerged as leading cause of death from a single infectious agent globally. Despite of rigorous protective measures, availability of multiple vaccines and with few approved therapeutics, the virus effect on the humankind throughout the world is perennial. COVID-19 has become the most urgent health concern with emergence of new challenging variants which outnumbered all other health issues and ensued in overwhelming number of reported deaths. In this unprecedented period of COVID-19 pandemic, scientists work round the clock for rapid development of efficient vaccines for prevention of infection and effective therapeutics for treatment. Here, we report the status of COVID-19 and highlight the ongoing research and development of vaccines and therapeutic strategies. It is necessary to know the present situation and available options to fight against the COVID-19 pandemic.
Collapse
Affiliation(s)
- Swapna Ponnampalli
- Department of Chemistry, CVR College of Engineering, Hyderabad 501510, India
| | | | - Ahmed Kamal
- Department of Pharmacy, Birla Institute of Technology & Science, Pilani, Hyderabad Campus, Hyderabad 500078, India; School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
121
|
Tobolowsky FA, Waltenburg MA, Moritz ED, Haile M, DaSilva JC, Schuh AJ, Thornburg NJ, Westbrook A, McKay SL, LaVoie SP, Folster JM, Harcourt JL, Tamin A, Stumpf MM, Mills L, Freeman B, Lester S, Beshearse E, Lecy KD, Brown LG, Fajardo G, Negley J, McDonald LC, Kutty PK, Brown AC. Longitudinal serologic and viral testing post-SARS-CoV-2 infection and post-receipt of mRNA COVID-19 vaccine in a nursing home cohort-Georgia, October 2020‒April 2021. PLoS One 2022; 17:e0275718. [PMID: 36301805 PMCID: PMC9612440 DOI: 10.1371/journal.pone.0275718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 09/21/2022] [Indexed: 11/18/2022] Open
Abstract
There are limited data describing SARS-CoV-2-specific immune responses and their durability following infection and vaccination in nursing home residents. We conducted a prospective longitudinal evaluation of 11 consenting SARS-CoV-2-positive nursing home residents to evaluate the quantitative titers and durability of binding antibodies detected after SARS-CoV-2 infection and subsequent COVID-19 vaccination. The evaluation included nine visits over 150 days from October 25, 2020, through April 1, 2021. Visits included questionnaire administration, blood collection for serology, and paired anterior nasal specimen collection for testing by BinaxNOW™ COVID-19 Ag Card (BinaxNOW), reverse transcription polymerase chain reaction (RT-PCR), and viral culture. We evaluated quantitative titers of binding SARS-CoV-2 antibodies post-infection and post-vaccination (beginning after the first dose of the primary series). The median age among participants was 74 years; one participant was immunocompromised. Of 10 participants with post-infection serology results, 9 (90%) had detectable Pan-Ig, IgG, and IgA antibodies, and 8 (80%) had detectable IgM antibodies. At first antibody detection post-infection, two-thirds (6/9, 67%) of participants were RT-PCR-positive, but none were culture- positive. Ten participants received vaccination; all had detectable Pan-Ig, IgG, and IgA antibodies through their final observation ≤90 days post-first dose. Post-vaccination geometric means of IgG titers were 10-200-fold higher than post-infection. Nursing home residents in this cohort mounted robust immune responses to SARS-CoV-2 post-infection and post-vaccination. The augmented antibody responses post-vaccination are potential indicators of enhanced protection that vaccination may confer on previously infected nursing home residents.
Collapse
Affiliation(s)
- Farrell A. Tobolowsky
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | | | - Erin D. Moritz
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Melia Haile
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Juliana C. DaSilva
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Amy J. Schuh
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Natalie J. Thornburg
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Adrianna Westbrook
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Susannah L. McKay
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Stephen P. LaVoie
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jennifer M. Folster
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jennifer L. Harcourt
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Azaibi Tamin
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Megan M. Stumpf
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Lisa Mills
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Brandi Freeman
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Sandra Lester
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Elizabeth Beshearse
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Kristin D. Lecy
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Laura G. Brown
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Geroncio Fajardo
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jeanne Negley
- Georgia Department of Public Health, Atlanta, Georgia, United States of America
| | - L. Clifford McDonald
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Preeta K. Kutty
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Allison C. Brown
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | | |
Collapse
|
122
|
Schmiedeberg K, Abela IA, Pikor NB, Vuilleumier N, Schwarzmueller M, Epp S, Pagano S, Grabherr S, Patterson AB, Nussberger M, Trkola A, Ludewig B, von Kempis J, Rubbert-Roth A. Postvaccination anti-S IgG levels predict anti-SARS-CoV-2 neutralising activity over 24 weeks in patients with RA. RMD Open 2022; 8:rmdopen-2022-002575. [PMID: 36288822 PMCID: PMC9615173 DOI: 10.1136/rmdopen-2022-002575] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 09/30/2022] [Indexed: 11/03/2022] Open
Abstract
OBJECTIVES To correlate immune responses following a two-dose regimen of mRNA anti-SARS-CoV-2 vaccines in patients with rheumatoid arthritis (RA) to the development of a potent neutralising antiviral activity. METHODS The RECOVER study was a prospective, monocentric study including patients with RA and healthy controls (HCs). Assessments were performed before, and 3, 6, 12 and 24 weeks, after the first vaccine dose, respectively, and included IgG, IgA and IgM responses (against receptor binding domain, S1, S2, N), IFN-γ ELISpots as well as neutralisation assays. RESULTS In patients with RA, IgG responses developed slower with lower peak titres compared with HC. Potent neutralising activity assessed by a SARS-CoV-2 pseudovirus neutralisation assay after 12 weeks was observed in all 21 HCs, and in 60.3% of 73 patients with RA. A significant correlation between peak anti-S IgG levels 2 weeks after the second vaccine dose and potent neutralising activity against SARS-CoV-2 was observed at weeks 12 and 24. The analysis of IgG, IgA and IgM isotype responses to different viral proteins demonstrated a delay in IgG but not in IgA and IgM responses. T cell responses were comparable in HC and patients with RA but declined earlier in patients with RA. CONCLUSION In patients with RA, vaccine-induced IgG antibody levels were diminished, while IgA and IgM responses persisted, indicating a delayed isotype switch. Anti-S IgG levels 2 weeks after the second vaccine dose correlate with the development of a potent neutralising activity after 12 and 24 weeks and may allow to identify patients who might benefit from additional vaccine doses or prophylactic regimen.
Collapse
Affiliation(s)
- Kristin Schmiedeberg
- Division of Rheumatology and Immunology, Kantonsspital St Gallen, Sankt Gallen, Switzerland
| | - Irene A Abela
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland,Division of Infectious Diseases and Hospital Epidemiology, University of Zurich, Zurich, Switzerland
| | | | | | - Magdalena Schwarzmueller
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland,Division of Infectious Diseases and Hospital Epidemiology, University of Zurich, Zurich, Switzerland
| | - Selina Epp
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland,Division of Infectious Diseases and Hospital Epidemiology, University of Zurich, Zurich, Switzerland
| | - Sabrina Pagano
- Laboratory Medicine Division, University of Geneva, Geneve, Switzerland
| | - Sarah Grabherr
- Institute of Immunobiology, Kantonsspital St Gallen, Sankt Gallen, Switzerland
| | | | - Madalina Nussberger
- Division of Rheumatology and Immunology, Kantonsspital St Gallen, Sankt Gallen, Switzerland
| | - Alexandra Trkola
- Institute of Medical Virology, University of Zurich, Zurich, Switzerland,Division of Infectious Diseases and Hospital Epidemiology, University of Zurich, Zurich, Switzerland
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St Gallen, Sankt Gallen, Switzerland,Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Johannes von Kempis
- Division of Rheumatology and Immunology, Kantonsspital St Gallen, Sankt Gallen, Switzerland
| | - Andrea Rubbert-Roth
- Division of Rheumatology and Immunology, Kantonsspital St Gallen, Sankt Gallen, Switzerland
| |
Collapse
|
123
|
Anderson EM, Li SH, Awofolaju M, Eilola T, Goodwin E, Bolton MJ, Gouma S, Manzoni TB, Hicks P, Goel RR, Painter MM, Apostolidis SA, Mathew D, Dunbar D, Fiore D, Brock A, Weaver J, Millar JS, DerOhannessian S, Greenplate AR, Frank I, Rader DJ, Wherry EJ, Bates P, Hensley SE. SARS-CoV-2 infections elicit higher levels of original antigenic sin antibodies compared with SARS-CoV-2 mRNA vaccinations. Cell Rep 2022; 41:111496. [PMID: 36261003 PMCID: PMC9578169 DOI: 10.1016/j.celrep.2022.111496] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 07/19/2022] [Accepted: 09/21/2022] [Indexed: 11/30/2022] Open
Abstract
It is important to determine if severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections and SARS-CoV-2 mRNA vaccinations elicit different types of antibodies. Here, we characterize the magnitude and specificity of SARS-CoV-2 spike-reactive antibodies from 10 acutely infected health care workers with no prior SARS-CoV-2 exposure history and 23 participants who received SARS-CoV-2 mRNA vaccines. We found that infection and primary mRNA vaccination elicit S1- and S2-reactive antibodies, while secondary vaccination boosts mostly S1 antibodies. Using absorption assays, we found that SARS-CoV-2 infections elicit a large proportion of original antigenic sin-like antibodies that bind efficiently to the spike of common seasonal human coronaviruses but poorly to the spike of SARS-CoV-2. In converse, vaccination modestly boosts antibodies reactive to the spike of common seasonal human coronaviruses, and these antibodies cross-react more efficiently to the spike of SARS-CoV-2. Our data indicate that SARS-CoV-2 infections and mRNA vaccinations elicit fundamentally different antibody responses.
Collapse
Affiliation(s)
- Elizabeth M Anderson
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shuk Hang Li
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Moses Awofolaju
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Theresa Eilola
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eileen Goodwin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marcus J Bolton
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sigrid Gouma
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tomaz B Manzoni
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Philip Hicks
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rishi R Goel
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark M Painter
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sokratis A Apostolidis
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Rheumatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Divij Mathew
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Debora Dunbar
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Danielle Fiore
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amanda Brock
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - JoEllen Weaver
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John S Millar
- Department of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephanie DerOhannessian
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Allison R Greenplate
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ian Frank
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel J Rader
- Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics and Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - E John Wherry
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA 19104, USA; Parker Institute for Cancer Immunotherapy, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - Paul Bates
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
124
|
Yu H, Liu H, Yang Y, Guan X. Development and Evaluation of a Rapid Neutralizing Antibody Assay for COVID-19 Vaccination. ACS OMEGA 2022; 7:36254-36262. [PMID: 36278077 PMCID: PMC9583339 DOI: 10.1021/acsomega.2c03677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 09/28/2022] [Indexed: 06/16/2023]
Abstract
SARS-CoV-2 neutralizing antibodies have excellent application prospects in the prevention and treatment of COVID-19. This study established a competitive colloidal gold immunochromatography assay (GICA) to detect neutralizing antibodies against the receptor-binding domain (RBD) of SARS-CoV-2 in postvaccination serum. The sensitivity, stability, and specificity of GICA were evaluated using neutralizing antibody solution reference material and positive serum. The consistency and correlation between GICA, pseudovirus neutralization (PN) assay, and ELISA were compared. Consistency analysis of serum neutralizing antibody and specific IgG antibody titers was conducted, and changes in neutralizing antibodies and specific IgG antibodies in serum after inoculation with the homologous booster inactivated vaccine and recombinant vaccine were noted. The sensitivity of the reagent was 20.66 IU/L, and the specificity was 100%. There was a strong consistency and correlation between GICA and PN (κ = 0.886, n = 165; r = 0.918, P < 0.001). The correlation coefficient of serum anti-RBD antibody and specific IgG antibody titers was 0.5253 (P < 0.001). The specific IgG antibody titers in serum after (W4) inoculation with homologous booster inactivated vaccine were 10.80 (S/CO).The anti-RBD antibody titers were 28.33. The anti-RBD omicron variant (B.1.1.529) antibody titers were 11.67. After inoculation with the recombinant vaccine, the specific IgG antibody titers in the serum of W4 were 10.68. The serum anti-RBD antibody titers of W4 were 103.30. The serum anti-RBD omicron variant (B.1.1.529) antibody titers of W4 were 56.67. Therefore, vaccination of the third dose of the homologous booster inactivated vaccine and recombinant vaccine can enhance the level of neutralizing antibodies against the omicron variant (B.1.1.529). This study demonstrates that a GICA kit for neutralizing antibodies against the RBD of SARS-CoV-2 can be used for COVID-19 vaccine evaluation. Changes in titers enable long-term monitoring of a population's immunity and guide interventions when their immunity declines.
Collapse
Affiliation(s)
- Heshan Yu
- Affiliated
Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110032, Liaoning, China
| | - Huan Liu
- Liaoning
University of Traditional Chinese Medicine, Shenyang 110847, Liaoning, China
| | - Yongju Yang
- Affiliated
Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110032, Liaoning, China
| | - Xuefeng Guan
- Liaoning
University of Traditional Chinese Medicine, Shenyang 110847, Liaoning, China
| |
Collapse
|
125
|
Kang B, Chu H, Youn BY, Leem J. Prevention and management of adverse events following COVID-19 vaccination using traditional Korean medicine: An online survey of public health doctors. World J Clin Cases 2022; 10:10053-10065. [PMID: 36246807 PMCID: PMC9561582 DOI: 10.12998/wjcc.v10.i28.10053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/14/2022] [Accepted: 08/22/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Since February 2021, vaccination against coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 has started in Korea.
AIM To perform a questionnaire survey about the prevention and management of adverse events of COVID-19 vaccination among public health doctors of Korean Medicine (PHKMDs).
METHODS An online questionnaire was developed, comprising categories about adverse events of vaccination (AEVs) and perception of using Korean medicine (KM) in managing vaccine adverse events (AEs) among PHKMDs. PHKMDs’ experience of AEVs, usage of Korean medicine for AEVs, and perception and attitude in using KM for AEVs were surveyed. The survey web-link was emailed to the association of PHKMDs. Online links were sent through Survey Monkey to all PHKMDs in Korea.
RESULTS A total of 143 participants were recruited for this study; 65% of participants were vaccinated at the same of the survey (n = 93). Although most participants did not take any medication for prevention of the adverse events, 62% (n = 58) of participants had taken herbal medicine to treat the adverse events (n = 52). Approximately 35% of participants (n = 33) said that they would recommend taking herbal medicine to prevent any AEVs. From various KM interventions, the participants strongly recommended taking an herbal medicine (n = 69, 74.19%) to treat AEs, and the second-highest was acupuncture treatment (n = 19, 20.43%).
CONCLUSION Overall, this research demonstrated a high prevalence of KM usage by the PHKMDs. Hence, this study's results may serve as fundamental evidence for health professionals to consider using KM treatments when treating or preventing AEVs in the near future.
Collapse
Affiliation(s)
- Byungsoo Kang
- College of Korean Medicine, Dongshin University, Naju-si 58245, South Korea
| | - Hongmin Chu
- Daecheong Public Health Subcenter, Ongjin Public Healthcenter, Incheon 23105, Incheon, South Korea
| | - Bo-Young Youn
- Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul 02447, South Korea
| | - Jungtae Leem
- College of Korean Medicine, Wonkwang University, Iksan 54538, Jeollabuk-do, South Korea
| |
Collapse
|
126
|
Salgado Del Riego E, Saiz ML, Corte-Iglesias V, Leoz Gordillo B, Martin-Martin C, Rodríguez-Pérez M, Escudero D, Lopez-Larrea C, Suarez-Alvarez B. Divergent SARS-CoV-2-specific T cell responses in intensive care unit workers following mRNA COVID-19 vaccination. Front Immunol 2022; 13:942192. [PMID: 36275696 PMCID: PMC9582956 DOI: 10.3389/fimmu.2022.942192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 09/16/2022] [Indexed: 11/13/2022] Open
Abstract
The cellular immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in response to full mRNA COVID-19 vaccination could be variable among healthy individuals. Studies based only in specific antibody levels could show an erroneous immune protection at long times. For that, we analyze the antibody levels specific to the S protein and the presence of SARS-CoV-2-specific T cells by ELISpot and AIM assays in intensive care unit (ICU) workers with no antecedents of COVID-19 and vaccinated with two doses of mRNA COVID-19 vaccines. All individuals were seronegative for the SARS-CoV-2 protein S before vaccination (Pre-v), but 34.1% (14/41) of them showed pre-existing T lymphocytes specific for some viral proteins (S, M and N). One month after receiving two doses of COVID-19 mRNA vaccine (Post-v1), all cases showed seroconversion with high levels of total and neutralizing antibodies to the spike protein, but six of them (14.6%) had no T cells reactive to the S protein. Specifically, they lack of specific CD8+ T cells, but maintain the contribution of CD4+ T cells. Analysis of the immune response against SARS-CoV-2 at 10 months after full vaccination (Post-v10), exhibited a significant reduction in the antibody levels (p<0.0001) and protein S-reactive T cells (p=0.0073) in all analyzed individuals, although none of the individuals become seronegative and 77% of them maintained a competent immune response. Thus, we can suggest that the immune response to SARS-CoV-2 elicited by the mRNA vaccines was highly variable among ICU workers. A non-negligible proportion of individuals did not develop a specific T cell response mediated by CD8+ T cells after vaccination, that may condition the susceptibility to further viral infections with SARS-CoV-2. By contrast, around 77% of individuals developed strong humoral and cellular immune responses to SARS-CoV-2 that persisted even after 10 months. Analysis of the cellular immune response is highly recommended for providing exact information about immune protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Estefanía Salgado Del Riego
- Servicio de Medicina Intensiva, Hospital Universitario Central de Asturias, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - María Laura Saiz
- Translational Immunology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Viviana Corte-Iglesias
- Translational Immunology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Blanca Leoz Gordillo
- Servicio de Medicina Intensiva, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Cristina Martin-Martin
- Translational Immunology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Mercedes Rodríguez-Pérez
- Servicio de Microbiología, Hospital Universitario Central de Asturias, Oviedo, Spain
- Translational Microbiology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Dolores Escudero
- Servicio de Medicina Intensiva, Hospital Universitario Central de Asturias, Oviedo, Spain
- Translational Microbiology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Carlos Lopez-Larrea
- Translational Immunology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
- Servicio de Inmunología, Hospital Universitario Central De Asturias, Oviedo, Spain
| | - Beatriz Suarez-Alvarez
- Translational Immunology, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| |
Collapse
|
127
|
Lenart K, Hellgren F, Ols S, Yan X, Cagigi A, Cerveira RA, Winge I, Hanczak J, Mueller SO, Jasny E, Schwendt K, Rauch S, Petsch B, Loré K. A third dose of the unmodified COVID-19 mRNA vaccine CVnCoV enhances quality and quantity of immune responses. Mol Ther Methods Clin Dev 2022; 27:309-323. [PMID: 36217434 PMCID: PMC9535876 DOI: 10.1016/j.omtm.2022.10.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 10/04/2022] [Indexed: 10/24/2022]
Abstract
A third vaccine dose is often required to achieve potent, long-lasting immune responses. We investigated the impact of three 8 μg doses of CVnCoV, CureVac's SARS-CoV-2 vaccine candidate containing sequence-optimized unmodified mRNA encoding spike (S) glycoprotein, administered at 0, 4 and 28 weeks on immune responses in rhesus macaques. Following the third dose S-specific binding and neutralizing antibodies increased 50-fold compared with post-dose 2 levels, with increased responses also evident in the lower airways and against the SARS-CoV-2 B.1.1.7 (Alpha), B.1.351 (Beta), P.1 (Gamma) and B.1.617.2 (Delta) variants. Enhanced binding affinity of serum antibodies after the third dose correlated with higher somatic hypermutation in S-specific B cells, corresponding with improved binding properties of monoclonal antibodies expressed from isolated B cells. Administration of low dose mRNA led to fewer cells expressing antigen in vivo at the injection site and in the draining lymph nodes compared with a tenfold higher dose, possibly reducing the engagement of precursor cells with the antigen and resulting in the suboptimal response observed following two-dose vaccination schedules in phase IIb/III clinical trials of CVnCoV. However, when immune memory is established, a third dose efficiently boosts the immunological responses as well as improves antibody affinity and breadth.
Collapse
Affiliation(s)
- Klara Lenart
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fredrika Hellgren
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sebastian Ols
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Xianglei Yan
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Alberto Cagigi
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rodrigo Arcoverde Cerveira
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Inga Winge
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jakub Hanczak
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | - Karin Loré
- Department of Medicine Solna, Division of Immunology and Allergy, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden,Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden,Correspondence should be addressed to: Karin Loré, Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Visionsgatan 4, BioClinicum J7:30, Karolinska University Hospital, 171 64 Stockholm, Sweden. E-mail address:
| |
Collapse
|
128
|
Christensen IE, Jyssum I, Tveter AT, Sexton J, Tran TT, Mjaaland S, Kro GB, Kvien TK, Warren DJ, Jahnsen J, Munthe LA, Haavardsholm EA, Vaage JT, Grødeland G, Lund-Johansen F, Jørgensen KK, Syversen SW, Goll GL, Provan SA. The persistence of anti-Spike antibodies following two SARS-CoV-2 vaccine doses in patients on immunosuppressive therapy compared to healthy controls-a prospective cohort study. BMC Med 2022; 20:378. [PMID: 36199139 PMCID: PMC9534475 DOI: 10.1186/s12916-022-02587-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 09/28/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The durability of vaccine-induced humoral immunity against SARS-CoV-2 in patients with immune-mediated inflammatory diseases (IMIDs) on immunosuppressive therapy is not known. The aim of this study was to compare the persistence of anti-Spike antibodies following two-dose SARS-CoV-2 vaccination between IMID patients and healthy controls and to identify factors associated with antibody decline. METHODS IMID patients on immunosuppressive medication enrolled in the prospective observational Nor-vaC study were included. Participants received two-dose SARS-CoV-2 vaccination. Serum collected at two time points following vaccination (first assessment within 6-48 days, second within 49-123 days) were analyzed for antibodies binding the receptor-binding domain (RBD) of the SARS-CoV-2 Spike protein. Multivariable regression models estimated percent reduction in anti-RBD over 30 days and factors associated with reduction. RESULTS A total of 1108 patients (403 rheumatoid arthritis, 195 psoriatic arthritis, 195 spondyloarthritis, 124 ulcerative colitis, 191 Crohn's disease) and 134 controls provided blood samples within the defined intervals (median 19 days [IQR 15-24] and 97 days [87-105] after second vaccine dose). Antibody levels were lower in patients compared to controls at both time points, with median anti-RBD 2806 BAU/ml [IQR 1018-6068] in patients and 6187 BAU/ml [4105-7496] in controls (p<0.001) at first assessment, and 608 BAU/ml [IQR 58-1053] in patients and 1520 BAU/ml [979-3766] in controls (p<0.001) at second assessment. At second assessment, low anti-RBD antibody levels (defined as <200 BAU/ml) were found in 449 (41%) patients, and 6 (5%) controls (p<0.001). The change was - 83% in patients and - 66% in controls (p<0.001). Patients had a greater estimated 30 days percent reduction in anti-RBD levels compared to controls - 4.9 (95% CI - 7.4 to - 2.4), (p<0.05). Among therapies, mono- or combination treatment with tumor necrosis factor inhibitors was associated with the greatest decline. CONCLUSIONS Within 4 months after vaccination, antibody levels declined considerably in both IMID patients and controls. Patients had lower initial antibody levels and a more pronounced decline compared to healthy controls and were therefore more likely to decline to low antibody levels. These results support that IMID patients need additional vaccine doses at an earlier stage than healthy individuals.
Collapse
Affiliation(s)
- Ingrid Egeland Christensen
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, P.O Box 23, Vinderen, N-0319, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Ingrid Jyssum
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, P.O Box 23, Vinderen, N-0319, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anne Therese Tveter
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, P.O Box 23, Vinderen, N-0319, Oslo, Norway
| | - Joseph Sexton
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, P.O Box 23, Vinderen, N-0319, Oslo, Norway
| | - Trung T Tran
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | | | | | - Tore K Kvien
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, P.O Box 23, Vinderen, N-0319, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - David John Warren
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Jørgen Jahnsen
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway
| | - Ludvig A Munthe
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Espen A Haavardsholm
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, P.O Box 23, Vinderen, N-0319, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - John Torgils Vaage
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Gunnveig Grødeland
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | | | - Silje Watterdal Syversen
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, P.O Box 23, Vinderen, N-0319, Oslo, Norway
| | - Guro Løvik Goll
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, P.O Box 23, Vinderen, N-0319, Oslo, Norway
| | - Sella Aarrestad Provan
- Center for treatment of Rheumatic and Musculoskeletal Diseases (REMEDY), Diakonhjemmet Hospital, P.O Box 23, Vinderen, N-0319, Oslo, Norway
- Department of Public Health and Sport Sciences, Inland Norway University of Applied Sciences, Elverum, Norway
| |
Collapse
|
129
|
Li Z, Zhang X, Ho W, Bai X, Jaijyan DK, Li F, Kumar R, Kolloli A, Subbian S, Zhu H, Xu X. Lipid-Polymer Hybrid "Particle-in-Particle" Nanostructure Gene Delivery Platform Explored for Lyophilizable DNA and mRNA COVID-19 Vaccines. ADVANCED FUNCTIONAL MATERIALS 2022; 32:2204462. [PMID: 35942271 PMCID: PMC9349454 DOI: 10.1002/adfm.202204462] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/05/2022] [Indexed: 05/06/2023]
Abstract
SARS-CoV-2 has led to a worldwide pandemic, catastrophically impacting public health and the global economy. Herein, a new class of lipid-modified polymer poly (β-amino esters) (L-PBAEs) is developed via enzyme-catalyzed esterification and further formulation of the L-PBAEs with poly(d,l-lactide-coglycolide)-b-poly(ethylene glycol) (PLGA-PEG) leads to self-assembly into a "particle-in-particle" (PNP) nanostructure for gene delivery. Out of 24 PNP candidates, the top-performing PNP/C12-PBAE nanoparticles efficiently deliver both DNA and mRNA in vitro and in vivo, presenting enhanced transfection efficacy, sustained gene release behavior, and excellent stability for at least 12 months of storage at -20 °C after lyophilization without loss of transfection efficacy. Encapsulated with spike encoded plasmid DNA and mRNA, the lipid-modified polymeric PNP COVID-19 vaccines successfully elicit spike-specific antibodies and Th1-biased T cell immune responses in immunized mice even after 12 months of lyophilized storage at -20 °C. This newly developed lipid-polymer hybrid PNP nanoparticle system demonstrates a new strategy for both plasmid DNA and mRNA delivery with the capability of long-term lyophilized storage.
Collapse
Affiliation(s)
- Zhongyu Li
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Xue‐Qing Zhang
- Engineering Research Center of Cell & Therapeutic Antibody Ministry of EducationSchool of PharmacyShanghai Jiao Tong UniversityShanghai200240P. R. China
| | - William Ho
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Xin Bai
- Engineering Research Center of Cell & Therapeutic Antibody Ministry of EducationSchool of PharmacyShanghai Jiao Tong UniversityShanghai200240P. R. China
| | - Dabbu Kumar Jaijyan
- Department of MicrobiologyBiochemistry and Molecular GeneticsRutgers—New Jersey Medical SchoolNewarkNJ07103USA
| | - Fengqiao Li
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Ranjeet Kumar
- Public Health Research Institute (PHRI)Rutgers—New Jersey Medical SchoolNewarkNJ07103USA
| | - Afsal Kolloli
- Public Health Research Institute (PHRI)Rutgers—New Jersey Medical SchoolNewarkNJ07103USA
| | - Selvakumar Subbian
- Public Health Research Institute (PHRI)Rutgers—New Jersey Medical SchoolNewarkNJ07103USA
| | - Hua Zhu
- Department of MicrobiologyBiochemistry and Molecular GeneticsRutgers—New Jersey Medical SchoolNewarkNJ07103USA
| | - Xiaoyang Xu
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
- Department of Biomedical EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| |
Collapse
|
130
|
Immunogenicity decay and case incidence six months post Sinovac-CoronaVac vaccine in autoimmune rheumatic diseases patients. Nat Commun 2022; 13:5801. [PMID: 36192386 PMCID: PMC9527375 DOI: 10.1038/s41467-022-33042-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 07/25/2022] [Indexed: 11/17/2022] Open
Abstract
The determination of durability and vaccine-associated protection is essential for booster doses strategies, however data on the stability of SARS-CoV-2 immunity are scarce. Here we assess anti-SARS-CoV-2 immunogenicity decay and incident cases six months after the 2nd dose of Sinovac-CoronaVac inactivated vaccine (D210) in 828 autoimmune rheumatic diseases patients compared with 207 age/sex-balanced control individuals. The primary outcome is the presence of anti-S1/S2 SARS-CoV-2 IgG at 6 months compared to 6 weeks after 2nd vaccine dose for decay evaluation. Secondary outcomes are presence of neutralizing antibodies, percent inhibition by neutralizing, geometric mean titers and cumulative incident cases at 6 months after 2nd dose. Anti-S1/S2 IgG positivity and titers reduce to 23.8% and 38% in patients (p < 0.001) during the six-month follow up and 20% and 51% in controls (p < 0.001), respectively. Neutralizing antibodies positivity and percent inhibition declines 41% and 54% in patients (p < 0.001) and 39.7% and 47% in controls (p < 0.001). Multivariate logistic regression analysis show males (OR = 0.56;95% CI0.40-0.79), prednisone (OR = 0.56; 95% CI0.41-0.76), anti-TNF (OR = 0.66;95% CI0.45-0.96), abatacept (OR = 0.29; 95% CI0.15-0.56) and rituximab (OR = 0.32;95% CI0.11-0.90) associate with a substantial reduction in IgG response at day 210 in patients. Although cellular immunity was not assessed, a decrease of COVID-19 cases (from 27.5 to 8.1/100 person-years; p < 0.001) is observed despite the concomitant emergence and spread of the Delta variant. Altogether we show a reduction in immunity 6-months of Sinovac-CoronaVac 2nd dose, particularly in males and those under immunosuppressives therapies, without a concomitant rise in COVID-19 cases. (CoronavRheum clinicaltrials.gov:NCT04754698). Characterising the response to SARS-CoV-2 post vaccination is critical in the appraisement of the induced immune response, performance and protective potential. Here the authors present data from a phase 4 clinical trial in autoimmune rheumatic disease patients 6 months post second dose of Sinovac-CoronaVac inactivated vaccine that show a marked reduction in antibody particularly in males or those under treatment with immune targeting therapies but saw no rise in COVID-19 disease.
Collapse
|
131
|
Jin J, Wang X, Carapito R, Moog C, Su B. Advances in Research on COVID-19 Vaccination for People Living with HIV. INFECTIOUS DISEASES & IMMUNITY 2022; 2:213-218. [PMID: 37520898 PMCID: PMC9612416 DOI: 10.1097/id9.0000000000000065] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Indexed: 01/24/2023]
Affiliation(s)
- Junyan Jin
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Xiuwen Wang
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Raphael Carapito
- Laboratoire d’ImmunoRhumatologie Moléculaire, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg 67000, France
| | - Christiane Moog
- Laboratoire d’ImmunoRhumatologie Moléculaire, Institut National de la Santé et de la Recherche Médicale (INSERM) UMR_S 1109, Institut Thématique Interdisciplinaire (ITI) de Médecine de Précision de Strasbourg, Transplantex NG, Faculté de Médecine, Fédération Hospitalo-Universitaire OMICARE, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg 67000, France
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| |
Collapse
|
132
|
Park T, Hwang H, Moon S, Kang SG, Song S, Kim YH, Kim H, Ko EJ, Yoon SD, Kang SM, Hwang HS. Vaccines against SARS-CoV-2 variants and future pandemics. Expert Rev Vaccines 2022; 21:1363-1376. [PMID: 35924678 PMCID: PMC9979704 DOI: 10.1080/14760584.2022.2110075] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 08/02/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Vaccination continues to be the most effective method for controlling COVID-19 infectious diseases. Nonetheless, SARS-CoV-2 variants continue to evolve and emerge, resulting in significant public concerns worldwide, even after more than 2 years since the COVID-19 pandemic. It is important to better understand how different COVID-19 vaccine platforms work, why SARS-CoV-2 variants continue to emerge, and what options for improving COVID-19 vaccines can be considered to fight against SARS-CoV-2 variants and future pandemics. AREA COVERED Here, we reviewed the innate immune sensors in the recognition of SARS-CoV-2 virus, innate and adaptive immunity including neutralizing antibodies by different COVID-19 vaccines. Efficacy comparison of the several COVID-19 vaccine platforms approved for use in humans, concerns about SARS-CoV-2 variants and breakthrough infections, and the options for developing future COIVD-19 vaccines were also covered. EXPERT OPINION Owing to the continuous emergence of novel pathogens and the reemergence of variants, safer and more effective new vaccines are needed. This review also aims to provide the knowledge basis for the development of next-generation COVID-19 and pan-coronavirus vaccines to provide cross-protection against new SARS-CoV-2 variants and future coronavirus pandemics.
Collapse
Affiliation(s)
- Taeyoung Park
- Department of Biology, College of Life Science and Industry, Sunchon National University (SCNU), Suncheon, South Korea
| | - Hyogyeong Hwang
- Department of Biology, College of Life Science and Industry, Sunchon National University (SCNU), Suncheon, South Korea
| | - Suhyeong Moon
- Department of Biology, College of Life Science and Industry, Sunchon National University (SCNU), Suncheon, South Korea
| | - Sang Gu Kang
- Department of Biology, College of Life Science and Industry, Sunchon National University (SCNU), Suncheon, South Korea
| | - Seunghyup Song
- Department of Biology, College of Life Science and Industry, Sunchon National University (SCNU), Suncheon, South Korea
| | - Young Hun Kim
- Department of Biology, College of Life Science and Industry, Sunchon National University (SCNU), Suncheon, South Korea
| | - Hanbi Kim
- Department of Biology, College of Life Science and Industry, Sunchon National University (SCNU), Suncheon, South Korea
| | - Eun-Ju Ko
- College of Veterinary Medicine and Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, South Korea
| | - Soon-Do Yoon
- Department of Chemical and Biomolecular Engineering, Chonnam National University, Yeosu, South Korea
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Hye Suk Hwang
- Department of Biology, College of Life Science and Industry, Sunchon National University (SCNU), Suncheon, South Korea
| |
Collapse
|
133
|
Brunner WM, Freilich D, Victory J, Krupa N, Scribani MB, Jenkins P, Lasher EG, Fink A, Shah A, Cross P, Bush V, Peek LJ, Pestano GA, Gadomski AM. Comparison of antibody response durability of mRNA-1273, BNT162b2, and Ad26.COV2.S SARS-CoV-2 vaccines in healthcare workers. Int J Infect Dis 2022; 123:183-191. [PMID: 36044963 PMCID: PMC9420538 DOI: 10.1016/j.ijid.2022.08.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES There are limited comparative immunologic durability data post COVID-19 vaccinations. METHODS Approximately 8.4 months after primary COVID-19 vaccination, 647 healthcare workers completed surveys about COVID-19 vaccinations/infections and blood draws. The groups included participants vaccinated with mRNA-1273 (n = 387), BNT162b2 (n = 212), or Ad26.COV2.S (n = 10) vaccines; unvaccinated participants (n = 10); and participants who received a booster dose (n = 28). The primary outcome was immunoglobin anti-spike titer. Secondary/tertiary outcomes included neutralizing antibodies (enzyme-linked immunosorbent assay-based pseudoneutralization) and vaccine effectiveness (VE). Antibody levels were compared using analysis of variance and linear regression. RESULTS Mean age was 49.7 and 75.3% of the participants were female. Baseline variables were balanced except for immunosuppression, previous COVID-19 infection, and post-primary vaccination time. Unadjusted median (interquartile range [IQR]) anti-spike titers (AU/ml) were 1539.5 (876.7-2626.7) for mRNA-1273, 751.2 (422.0-1381.5) for BNT162b2, 451.6 (103.0-2396.7) for Ad26.COV2.S, 113.4 (3.7-194.0) for unvaccinated participants, and 31898.8 (21347.1-45820.1) for participants administered with booster dose (mRNA-1273 vs BNT162b2, P <.001; mRNA-1273, BNT162b2, or boosted vs unvaccinated, P <.006; mRNA-1273, BNT162b2, Ad26.COV2.S, or unvaccinated vs boosted, P <.001). Unadjusted median (IQR) pseudoneutralization was as follows: 90.9% (80.1-95.0) for mRNA-1273, 77.2% (59.1-89.9) for BNT162b2, 57.9% (36.6-95.8) for Ad26.COV2.S, 40.1% (21.7-60.6) for unvaccinated, and 96.4% (96.1-96.6) for participants administered with booster dose (mRNA-1273 vs BNT162b2, P <.001; mRNA-1273, BNT162b2, or boosted vs unvaccinated, P <.028; mRNA-1273, BNT162b2, Ad26.COV2.S, or unvaccinated vs boosted, P <.001). VE was 87-89% for participants administered mRNA-1273 vaccine, BNT162b2 vaccine, and booster dose, and 33% for Ad26.COV2.S (none significantly different). CONCLUSION Antibody responses 8.4 months after primary vaccination were significantly higher with mRNA-1273 than those observed with BNT162b2.
Collapse
Affiliation(s)
- Wendy M. Brunner
- Bassett Research Institute, Bassett Medical Center, Cooperstown, 13326, USA,Corresponding author: Wendy M. Brunner, 1 Atwell Road, Cooperstown, New York, 13326, New York, USA. Tel: 607-547-6559
| | - Daniel Freilich
- Department of Internal Medicine, Bassett Medical Center, Cooperstown, 13326, USA
| | - Jennifer Victory
- Bassett Research Institute, Bassett Medical Center, Cooperstown, 13326, USA
| | - Nicole Krupa
- Bassett Research Institute, Bassett Medical Center, Cooperstown, 13326, USA
| | | | - Paul Jenkins
- Bassett Research Institute, Bassett Medical Center, Cooperstown, 13326, USA
| | - Emily G. Lasher
- Bassett Research Institute, Bassett Medical Center, Cooperstown, 13326, USA
| | - Amanda Fink
- Bassett Research Institute, Bassett Medical Center, Cooperstown, 13326, USA
| | - Anshini Shah
- Medical Education, Bassett Medical Center, Cooperstown, 13326, USA
| | - Peggy Cross
- Bassett Research Institute, Bassett Medical Center, Cooperstown, 13326, USA
| | - Valerie Bush
- Bassett Healthcare Network Clinical Laboratory, Cooperstown, 13326, USA
| | | | | | - Anne M. Gadomski
- Bassett Research Institute, Bassett Medical Center, Cooperstown, 13326, USA
| |
Collapse
|
134
|
Affiliation(s)
- Dan H Barouch
- From Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, and the Ragon Institute of MGH, MIT, and Harvard, Cambridge - both in Massachusetts
| |
Collapse
|
135
|
Kushima H, Ishii H, Kinoshita Y, Koide Y, Komiya Y, Kato J, Umehara M, Sakata M, Miyazaki M, Ikuta M. The Limited Effect of a History of COVID-19 on Antibody Titers and Adverse Reactions Following BNT162b2 Vaccination: A Single-Center Prospective Study. J Clin Med 2022; 11:jcm11185388. [PMID: 36143040 PMCID: PMC9505143 DOI: 10.3390/jcm11185388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/11/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Objectives: The need for, and ideal frequency of, the vaccination against coronavirus disease 2019 (COVID-19) of previously infected individuals have not yet been sufficiently evaluated. The aim of this study was to examine the anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibody status and adverse reactions after vaccination among medical staff with or without a history of COVID-19. Materials and Methods: A single-center prospective study was performed at Fukuoka University Chikushi Hospital. We investigated the presence of the anti-SARS-CoV-2 antibody titer among medical staff before and after mRNA vaccination with the BNT162b2. The levels of immunoglobulin G antibody were quantitatively measured at six points—before vaccination, after the first vaccination, at three points after the second vaccination, and finally, after the third vaccination—and the levels were then compared based on the COVID-19 infection history. Results: The previously infected (before the first vaccination) subjects (n = 17) showed a marked increase in antibody titers two weeks after the first vaccination and four weeks after the second vaccination. Although they were able to maintain a certain level of antibody titers until 30 weeks after the second vaccination, the titers fell in the same way as observed in the non-infected subjects. The subjects who did not receive the third vaccination due to adverse reactions to previous vaccines (n = 1) or who were positive for COVID-19 prior to the third vaccination (n = 2) were excluded from the subsequent analyses. Among non-infected subjects (n = 36), smokers had lower peak antibody titers than the others. The previously infected subjects had a significantly higher incidence of adverse reactions after the first vaccination but had a similar incidence of adverse reactions after the second and third vaccinations compared to the non-infected subjects. Conclusions: A history of COVID-19 may influence only the initial increase in anti-SARS-CoV-2 antibody titers and the occurrence of adverse reactions after the first vaccination.
Collapse
Affiliation(s)
- Hisako Kushima
- Department of Infection Control and Prevention, Fukuoka University Chikushi Hospital, Chikushino 818-8502, Japan
- Department of Respiratory Medicine, Fukuoka University Chikushi Hospital, Chikushino 818-8502, Japan
| | - Hiroshi Ishii
- Department of Infection Control and Prevention, Fukuoka University Chikushi Hospital, Chikushino 818-8502, Japan
- Department of Respiratory Medicine, Fukuoka University Chikushi Hospital, Chikushino 818-8502, Japan
- Correspondence: ; Tel.: +81-92-921-1011
| | - Yoshiaki Kinoshita
- Department of Respiratory Medicine, Fukuoka University Chikushi Hospital, Chikushino 818-8502, Japan
| | - Yohei Koide
- Department of Infection Control and Prevention, Fukuoka University Chikushi Hospital, Chikushino 818-8502, Japan
- Department of Respiratory Medicine, Fukuoka University Chikushi Hospital, Chikushino 818-8502, Japan
| | - Yukie Komiya
- Department of Clinical Laboratory, Fukuoka University Chikushi Hospital, Chikushino 818-8502, Japan
| | - Junko Kato
- Department of Clinical Laboratory, Fukuoka University Chikushi Hospital, Chikushino 818-8502, Japan
| | - Mayumi Umehara
- Department of Infection Control and Prevention, Fukuoka University Chikushi Hospital, Chikushino 818-8502, Japan
| | - Miho Sakata
- Department of Infection Control and Prevention, Fukuoka University Chikushi Hospital, Chikushino 818-8502, Japan
| | - Motoyasu Miyazaki
- Department of Pharmacy, Fukuoka University Chikushi Hospital, Chikushino 818-8502, Japan
| | - Mikihiro Ikuta
- Department of Clinical Laboratory, Fukuoka University Chikushi Hospital, Chikushino 818-8502, Japan
| |
Collapse
|
136
|
Methodology to estimate natural- and vaccine-induced antibodies to SARS-CoV-2 in a large geographic region. PLoS One 2022; 17:e0273694. [PMID: 36084125 PMCID: PMC9462720 DOI: 10.1371/journal.pone.0273694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 08/14/2022] [Indexed: 11/19/2022] Open
Abstract
Accurate estimates of natural and/or vaccine-induced antibodies to SARS-CoV-2 are difficult to obtain. Although model-based estimates of seroprevalence have been proposed, they require inputting unknown parameters including viral reproduction number, longevity of immune response, and other dynamic factors. In contrast to a model-based approach, the current study presents a data-driven detailed statistical procedure for estimating total seroprevalence (defined as antibodies from natural infection or from full vaccination) in a region using prospectively collected serological data and state-level vaccination data. Specifically, we conducted a longitudinal statewide serological survey with 88,605 participants 5 years or older with 3 prospective blood draws beginning September 30, 2020. Along with state vaccination data, as of October 31, 2021, the estimated percentage of those 5 years or older with naturally occurring antibodies to SARS-CoV-2 in Texas is 35.0% (95% CI = (33.1%, 36.9%)). This is 3× higher than, state-confirmed COVID-19 cases (11.83%) for all ages. The percentage with naturally occurring or vaccine-induced antibodies (total seroprevalence) is 77.42%. This methodology is integral to pandemic preparedness as accurate estimates of seroprevalence can inform policy-making decisions relevant to SARS-CoV-2.
Collapse
|
137
|
Wang F, Huang B, Lv H, Feng L, Ren W, Wang X, Tang L, Liu Q, Wu D, Zheng H, An Z, Deng Y, Zhao L, Ye F, Wang W, Zhang H, Chang S, Liao Y, Chen F, Rodewald LE, Gao GF, Yin Z, Tan W. Factors associated with neutralizing antibody levels induced by two inactivated COVID-19 vaccines for 12 months after primary series vaccination. Front Immunol 2022; 13:967051. [PMID: 36159863 PMCID: PMC9501884 DOI: 10.3389/fimmu.2022.967051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 08/22/2022] [Indexed: 11/21/2022] Open
Abstract
Background BBIBP-CorV and CoronaVac inactivated COVID-19 vaccines are widely-used, World Health Organization-emergency-listed vaccines. Understanding antibody level changes over time after vaccination is important for booster dose policies. We evaluated neutralizing antibody (nAb) titers and associated factors for the first 12 months after primary-series vaccination with BBIBP-CorV and CoronaVac. Methods Our study consisted of a set of cross-sectional sero-surveys in Zhejiang and Shanxi provinces, China. In 2021, we enrolled 1,527 consenting 18-59-year-olds who received two doses of BBIBP-CorV or CoronaVac 1, 3, 6, 9, or 12 months earlier and obtained blood samples and demographic and medical data. We obtained 6-month convalescent sera from 62 individuals in Hebei province. Serum nAb titers were measured by standard micro-neutralization cytopathic effect assay in Vero cells with ancestral SARS-CoV-2 strain HB01. We used the first WHO International Standard (IS) for anti-SARS-CoV-2 immunoglobulin (NIBSC code 20/136) to standardized geometric mean concentrations (IU/mL) derived from the nAb geometric mean titers (GMT over 1:4 was considered seropositive). We analyzed nAb titer trends using Chi-square and factors related to nAb titers with logistic regression and linear models. Results Numbers of subjects in each of the five month-groupings ranged from 100 to 200 for each vaccine and met group-specific target sample sizes. Seropositivity rates from BBIBP-CorV were 98.0% at 1 month and 53.5% at 12 months, and GMTs were 25.0 and 4.0. Respective seropositivity rates from CoronaVac were 90.0% and 62.5%, and GMTs were 20.2 and 4.1. One-, three-, six-, nine-, and twelve-month GMCs were 217.2, 84.1, 85.7, 44.6, and 10.9 IU/mL in BBIBP-CorV recipients and 195.7, 94.6, 51.7, 27.6, and 13.4 IU/mL in CoronaVac recipients. Six-month convalescent seropositivity was 95.2%; GMC was 108.9 IU/mL. Seropositivity and GMCs were associated with age, sex, and time since vaccination. Conclusions Neutralizing Ab levels against ancestral SARS-CoV-2 from BBIBP-CorV or CoronaVac vaccination were similar and decreased with increasing time since vaccination; over half of 12-month post-vaccination subjects were seropositive. Seropositivity and GMCs from BBIBP-CorV and CoronaVac six and nine months after vaccination were similar to or slightly lower than in six-month convalescent sera. These real-world data suggest necessity of six-month booster doses.
Collapse
Affiliation(s)
- Fuzhen Wang
- National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Baoying Huang
- National Health Commission (NHC) Key Laboratory of Biosafety, Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Huakun Lv
- Immunization Program Institute, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Lizhong Feng
- Immunization Program Institute, Shanxi Provincial Center for Disease Control and Prevention, Taiyuan, China
| | - Weihong Ren
- Xingtai Center for Disease Control and Prevention, Xingtai, China
| | - Xiaoqi Wang
- National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Lin Tang
- National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qianqian Liu
- National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Dan Wu
- National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hui Zheng
- National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhijie An
- National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yao Deng
- National Health Commission (NHC) Key Laboratory of Biosafety, Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Li Zhao
- National Health Commission (NHC) Key Laboratory of Biosafety, Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Fei Ye
- National Health Commission (NHC) Key Laboratory of Biosafety, Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wenling Wang
- National Health Commission (NHC) Key Laboratory of Biosafety, Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hangjie Zhang
- Immunization Program Institute, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Shaoying Chang
- Immunization Program Institute, Shanxi Provincial Center for Disease Control and Prevention, Taiyuan, China
| | - Yuting Liao
- School of Public Health, Xiamen University, Xiamen, China
| | - Fengyang Chen
- Xingtai Center for Disease Control and Prevention, Xingtai, China
| | - Lance E. Rodewald
- National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
| | - George F. Gao
- Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zundong Yin
- National Immunization Program, Chinese Center for Disease Control and Prevention, Beijing, China
- *Correspondence: Zundong Yin, ; Wenjie Tan,
| | - Wenjie Tan
- National Health Commission (NHC) Key Laboratory of Biosafety, Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- *Correspondence: Zundong Yin, ; Wenjie Tan,
| |
Collapse
|
138
|
Marta RA, Nakamura GEK, de Matos Aquino B, Bignardi PR. COVID-19 vaccines: Update of the vaccines in use and under development. VACUNAS 2022; 23:S88-S102. [PMID: 35761987 PMCID: PMC9217136 DOI: 10.1016/j.vacun.2022.06.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/02/2022] [Indexed: 12/30/2022]
Abstract
The ongoing COVID-19 pandemic has imposed a series of challenges on the scientific community. One of the biggest was the development of safe and effective vaccines in record time, which could be achieved through a global effort. A topic of great discussion has been the technology surrounding these vaccines: ranging from the well-known inactivated virus vaccines to the latest RNA vaccines. As vaccines became available, another point also came into question: their efficacy and effectiveness against the original Wuhan strain and its variants. Among the numerous variants, 5 of them (Alpha, Beta, Gamma, Delta and, more recently, Omicron) gained greater prominence due to their epidemiological relevance. In this scenario, with numerous variants and several vaccine options, scientific information can often be mismatched. This review aims to provide an overview of the efficacy, effectiveness, and safety of 11 vaccines in use or under development against the original Wuhan strain and the variants of concern identified by the World Health Organisation (WHO). Simultaneously, we aim to explore possible scenarios that can be expected shortly regarding new variants and vaccines. Overall, COVID-19 vaccines have satisfactory efficacy and loss of effectiveness against SARS-CoV-2 variants, especially the Omicron strain.
Collapse
Affiliation(s)
| | | | | | - Paulo R Bignardi
- School of Medicine, Pontifical Catholic University of Paraná, Londrina, Brazil
| |
Collapse
|
139
|
Differential persistence of neutralizing antibody against SARS-CoV-2 in post immunized Bangladeshi population. Sci Rep 2022; 12:14681. [PMID: 36038600 PMCID: PMC9421641 DOI: 10.1038/s41598-022-18302-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Abstract
Development of effective vaccines have been immensely welcomed by the world to prevent the transmission of SARS-CoV-2. However, the duration and clinical implications of antibody-mediated natural immunity in SARS-CoV-2 have not been adequately elucidated alongside some other immune system transforming factors. In a cohort study, we measured NAb titer following the 2nd immunization dosage of the CoviShield (AZD1222) vaccine. The enzyme-linked immunoassay was used to look for SARS-CoV-2—specific NAb. We measured NAb at 30 days after the 2nd dosage of immunization and > 96% titer was detected in 42.9% of subjects, but only 5.1% of subjects retained the same level after 180 days. The median NAb titer dropped significantly, from 92% at 30 days to 58% at 180 days (p < 0.001). Besides, there were significant differences observed in NAb titer after 180 days by age, sex, COVID-19 infection, tobacco use, and asthma patients. However, SARS-CoV-2 infection along with two dosages of immunization upheld NAb titer (p < 0.001) even at the end of the study period.
Collapse
|
140
|
Čiučiulkaitė I, Möhlendick B, Thümmler L, Fisenkci N, Elsner C, Dittmer U, Siffert W, Lindemann M. GNB3 c.825c>T polymorphism influences T-cell but not antibody response following vaccination with the mRNA-1273 vaccine. Front Genet 2022; 13:932043. [PMID: 36105097 PMCID: PMC9465595 DOI: 10.3389/fgene.2022.932043] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/01/2022] [Indexed: 11/29/2022] Open
Abstract
Background: Immune responses following vaccination against COVID-19 with different vaccines and the waning of immunity vary within the population. Genetic host factors are likely to contribute to this variability. However, to the best of our knowledge, no study on G protein polymorphisms and vaccination responses against COVID-19 has been published so far. Methods: Antibodies against the SARS-CoV-2 spike protein and T-cell responses against a peptide pool of SARS-CoV-2 S1 proteins were measured 1 and 6 months after the second vaccination with mRNA-1273 in the main study group of 204 participants. Additionally, antibodies against the SARS-CoV-2 spike protein were measured in a group of 597 participants 1 month after the second vaccination with mRNA-1273. Genotypes of GNB3 c.825C>T were determined in all participants. Results: The median antibody titer against the SARS-CoV-2 spike protein and median values of spots increment in the SARS-CoV-2 IFN-γ ELISpot assay against the S1-peptide pool were significantly decreased from months 1 to 6 (p < 0.0001). Genotypes of GNB3 c.825C>T had no influence on the humoral immune response. At month 1, CC genotype carriers had significantly increased T-cell responses compared to CT (p = 0.005) or TT (p = 0.02) genotypes. CC genotype carriers had an almost 6-fold increased probability compared to TT genotype carriers and an almost 3-fold increased probability compared to T-allele carriers to mount a SARS-CoV-2-specific T-cell response above the median value. Conclusion: CC genotype carriers of the GNB3 c.825C>T polymorphism have an increased T-cell immune response to SARS-CoV-2, which may indicate better T-cell-mediated protection against COVID-19 after vaccination with mRNA-1273.
Collapse
Affiliation(s)
- Ieva Čiučiulkaitė
- Institute of Pharmacogenetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- *Correspondence: Ieva Čiučiulkaitė,
| | - Birte Möhlendick
- Institute of Pharmacogenetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Laura Thümmler
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Neslinur Fisenkci
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Carina Elsner
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Winfried Siffert
- Institute of Pharmacogenetics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Monika Lindemann
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
141
|
Zhang J, Han ZB, Liang Y, Zhang XF, Jin YQ, Du LF, Shao S, Wang H, Hou JW, Xu K, Lei W, Lei ZH, Liu ZM, Zhang J, Hou YN, Liu N, Shen FJ, Wu JJ, Zheng X, Li XY, Li X, Huang WJ, Wu GZ, Su JG, Li QM. A mosaic-type trimeric RBD-based COVID-19 vaccine candidate induces potent neutralization against Omicron and other SARS-CoV-2 variants. eLife 2022; 11:e78633. [PMID: 36004719 PMCID: PMC9481243 DOI: 10.7554/elife.78633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 08/22/2022] [Indexed: 11/29/2022] Open
Abstract
Large-scale populations in the world have been vaccinated with COVID-19 vaccines, however, breakthrough infections of SARS-CoV-2 are still growing rapidly due to the emergence of immune-evasive variants, especially Omicron. It is urgent to develop effective broad-spectrum vaccines to better control the pandemic of these variants. Here, we present a mosaic-type trimeric form of spike receptor-binding domain (mos-tri-RBD) as a broad-spectrum vaccine candidate, which carries the key mutations from Omicron and other circulating variants. Tests in rats showed that the designed mos-tri-RBD, whether used alone or as a booster shot, elicited potent cross-neutralizing antibodies against not only Omicron but also other immune-evasive variants. Neutralizing antibody ID50 titers induced by mos-tri-RBD were substantially higher than those elicited by homo-tri-RBD (containing homologous RBDs from prototype strain) or the BIBP inactivated COVID-19 vaccine (BBIBP-CorV). Our study indicates that mos-tri-RBD is highly immunogenic, which may serve as a broad-spectrum vaccine candidate in combating SARS-CoV-2 variants including Omicron.
Collapse
Affiliation(s)
- Jing Zhang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Zi Bo Han
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Yu Liang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xue Feng Zhang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Yu Qin Jin
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
| | - Li Fang Du
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Shuai Shao
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Hui Wang
- Beijing Institute of Biological Products Company LimitedBeijingChina
| | - Jun Wei Hou
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Ke Xu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC)BeijingChina
| | - Wenwen Lei
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC)BeijingChina
| | - Ze Hua Lei
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Zhao Ming Liu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Jin Zhang
- Beijing Institute of Biological Products Company LimitedBeijingChina
| | - Ya Nan Hou
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Ning Liu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Fu Jie Shen
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Jin Juan Wu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xiang Zheng
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xin Yu Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xin Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Wei Jin Huang
- National Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Gui Zhen Wu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC)BeijingChina
| | - Ji Guo Su
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Qi Ming Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| |
Collapse
|
142
|
Speich B, Chammartin F, Abela IA, Amico P, Stoeckle MP, Eichenberger AL, Hasse B, Braun DL, Schuurmans MM, Müller TF, Tamm M, Audigé A, Mueller NJ, Rauch A, Günthard HF, Koller MT, Trkola A, Briel M, Kusejko K, Bucher HC. Antibody Response in Immunocompromised Patients After the Administration of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Vaccine BNT162b2 or mRNA-1273: A Randomized Controlled Trial. Clin Infect Dis 2022; 75:e585-e593. [PMID: 35234868 PMCID: PMC8903480 DOI: 10.1093/cid/ciac169] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND BNT162b2 by Pfizer-BioNTech and mRNA-1273 by Moderna are the most commonly used vaccines to prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. Head-to-head comparison of the efficacy of these vaccines in immunocompromised patients is lacking. METHODS Parallel, 2-arm (allocation 1:1), open-label, noninferiority randomized clinical trial nested into the Swiss HIV Cohort Study and the Swiss Transplant Cohort Study. People living with human immunodeficiency virus (PLWH) or solid organ transplant recipients (SOTR; ie, lung and kidney) from these cohorts were randomized to mRNA-1273 or BNT162b2. The primary endpoint was antibody response to SARS-CoV-2 spike (S1) protein receptor binding domain (Elecsys Anti-SARS-CoV-2 immunoassay, Roche; cutoff ≥0.8 units/mL) 12 weeks after first vaccination (ie, 8 weeks after second vaccination). In addition, antibody response was measured with the Antibody Coronavirus Assay 2 (ABCORA 2). RESULTS A total of 430 patients were randomized and 412 were included in the intention-to-treat analysis (341 PLWH and 71 SOTR). The percentage of patients showing an immune response was 92.1% (95% confidence interval [CI]: 88.4-95.8; 186/202) for mRNA-1273 and 94.3% (95% CI: 91.2-97.4; 198/210) for BNT162b2 (difference: -2.2%; 95% CI: -7.1 to 2.7), fulfilling noninferiority of mRNA-1273. With the ABCORA 2 test, 89.1% had an immune response to mRNA-1273 (95% CI: 84.8-93.4; 180/202) and 89.5% to BNT162b2 (95% CI: 85.4-93.7; 188/210). Based on the Elecsys test, all PLWH had an antibody response (100.0%; 341/341), whereas for SOTR, only 60.6% (95% CI: 49.2-71.9; 43/71) had titers above the cutoff level. CONCLUSIONS In immunocompromised patients, the antibody response of mRNA-1273 was noninferior to BNT162b2. PLWH had in general an antibody response, whereas a high proportion of SOTR had no antibody response.
Collapse
Affiliation(s)
- Benjamin Speich
- Basel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
- Centre for Statistics in Medicine, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Frédérique Chammartin
- Basel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Irene A Abela
- University of Zurich, Institute of Medical Virology, Zurich, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Patrizia Amico
- Clinic for Transplantation Immunology and Nephrology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Marcel P Stoeckle
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Basel, University of Basel, Switzerland
| | - Anna L Eichenberger
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Barbara Hasse
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Dominique L Braun
- University of Zurich, Institute of Medical Virology, Zurich, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Macé M Schuurmans
- Division of Pulmonology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas F Müller
- Nephrology Clinic, University Hospital Zurich, Zürich, Switzerland
| | - Michael Tamm
- Clinic of Respiratory Medicine and Pulmonary Cell Research, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Annette Audigé
- University of Zurich, Institute of Medical Virology, Zurich, Switzerland
| | - Nicolas J Mueller
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Andri Rauch
- Department of Infectious Diseases, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Huldrych F Günthard
- University of Zurich, Institute of Medical Virology, Zurich, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Michael T Koller
- Swiss Transplant Cohorts Study, University Hospital Basel, University of Basel, Basel, Switzerlandand
| | - Alexandra Trkola
- University of Zurich, Institute of Medical Virology, Zurich, Switzerland
| | - Matthias Briel
- Basel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Katharina Kusejko
- University of Zurich, Institute of Medical Virology, Zurich, Switzerland
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, Zurich, Switzerland
| | - Heiner C Bucher
- Basel Institute for Clinical Epidemiology and Biostatistics, Department of Clinical Research, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
143
|
Saresella M, Piancone F, Marventano I, Hernis A, Trabattoni D, Invernizzi M, La Rosa F, Clerici M. Innate immune responses to three doses of the BNT162b2 mRNA SARS-CoV-2 vaccine. Front Immunol 2022; 13:947320. [PMID: 36072604 PMCID: PMC9443429 DOI: 10.3389/fimmu.2022.947320] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022] Open
Abstract
To explore the effects of SARS-CoV-2-mRNA vaccines on innate immune responses we enrolled 58 individuals who received 3 doses of the BNT162b2 vaccine in a longitudinal study; 45 of these individuals had never been SARS-CoV-2 infected. Results showed that vaccination significantly increased: 1) classical and intermediate inflammatory monocytes, 2) CD56bright, CD56dim, and CD56dim/CD16dim NK cells, and 3) IFN-γ+ ;production as well as perforin and granzyme content by NK cells. Vaccination also reduced expression of the NK inhibitory receptor ILT-2, increasing that of the stimulatory molecule 2DS2. These effects were long-lasting and were boosted by every vaccine dose. Notably, ILT-2 expressing NK cells were reduced even more robustly in COVID-19-recovereed vaccines. BNT162b1 mRNA vaccine is known to induce potent adaptive immune responses; results herein show its ability to modulate innate immune responses as well, offering further support to the indication to proceed with worldwide vaccination efforts to end the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Marina Saresella
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- *Correspondence: Marina Saresella,
| | | | | | - Ambra Hernis
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Daria Trabattoni
- Departments of Biomedical and Clinical Sciences L. Sacco, University of Milan, Milan, Italy
| | | | | | - Mario Clerici
- IRCCS Fondazione Don Carlo Gnocchi, Milan, Italy
- Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
144
|
Psaridi L, Maltezou HC, Simonidou S, Lialliou I, Athanasopoulou D, Haila Z, Kyrimi A, Giannopoulou I, Giannousa S, Pseimada M, Christofilea O, Dounias G, Lanitis S, Kremasmenou E. Neutralizing antibody responses in healthcare personnel after three doses of mRNA BNT162b2 vaccine and association with baseline characteristics and past SARS-CoV-2 infection. Vaccine 2022; 40:5752-5756. [PMID: 36008235 PMCID: PMC9388445 DOI: 10.1016/j.vaccine.2022.08.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 07/12/2022] [Accepted: 08/15/2022] [Indexed: 11/24/2022]
Abstract
Aim To estimate neutralizing antibody (NAb) immunity against SARS-CoV-2 in 739 healthcare personnel (HCP) vaccinated with three doses of BNT162b2 mRNA vaccine. Methods Serum samples were collected at 3, 6, and 9 months after the second vaccine dose and at 7–55 days after the third dose. Samples were tested for NAbs against SARS-CoV-2 receptor binding domain. Results The mean inhibition rates at 3, 6, and 9 months after the second dose were 86.33%, 73.38%, and 61.18%, and increased to 95.57% after the booster dose. Younger HCP and HCP with past SARS-CoV-2 infection had higher inhibition rates while there was an inverse correlation between NAb levels and comorbidities or tobacco use (p-values < 0.001). Increased NAb titers were also noticed in women (p-value = 0.033), especially at the end of the 9-month study period. Conclusion NAb levels increased considerably after a booster mRNA vaccine dose. Host factors and past SARS-CoV-2 infection influence NAb titers.
Collapse
Affiliation(s)
- Loukia Psaridi
- Department of Immunology, Red Cross General Hospital of Athens, Greece
| | - Helena C Maltezou
- Directorate of Research, Studies, and Documentation, National Public Health Organization, Athens, Greece.
| | - Sofia Simonidou
- Department of Immunology, Red Cross General Hospital of Athens, Greece
| | - Ioanna Lialliou
- Department of Immunology, Red Cross General Hospital of Athens, Greece
| | | | - Zoi Haila
- Department of Immunology, Red Cross General Hospital of Athens, Greece
| | - Areti Kyrimi
- Department of Immunology, Red Cross General Hospital of Athens, Greece
| | | | | | - Maria Pseimada
- Department of Immunology, Red Cross General Hospital of Athens, Greece
| | | | - George Dounias
- Department of Public Health Policies, School of Public Health, University of West Attica, Athens, Greece
| | - Sophocles Lanitis
- 2(nd) Surgical Department and Unit of Surgical Oncology, Red Cross General Hospital of Athens, Athens, Greece
| | | |
Collapse
|
145
|
Campbell C, Roblin D, Padmanabhan N, Romero D, Joe J, Fathi L, Whiting T, Williamson J, Goodwin P, Mckie C, Deneal A, Greenberg L, Sigal G. Saliva-based SARS-CoV-2 serology using at-home collection kits returned via mail. Sci Rep 2022; 12:14061. [PMID: 35982133 PMCID: PMC9387411 DOI: 10.1038/s41598-022-17057-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 07/20/2022] [Indexed: 12/16/2022] Open
Abstract
Serology provides tools for epidemiologic studies, and may have a role in vaccine prioritization and selection. Automated serologic testing of saliva, especially specimens that are self-collected at home and sent to a laboratory via the mail without refrigeration, could be a highly-scalable strategy for population-wide testing. In this prospective study, non-vaccinated patients were recruited after PCR testing to self-collect saliva and return their specimens via mail. Longitudinal specimens were analyzed in order to monitor seroconversion in the weeks after a diagnostic PCR test for SARS-CoV-2. Diverse users self-collected saliva and returned specimens via mail in compliance with shipping regulations. At our pre-established threshold (0.963 AU/mL), salivary IgG reactivity to full-length spike protein achieved 95.8% sensitivity and 92.4% specificity at 2–4 weeks after diagnostic testing, which is comparable to the typical sensitivity and specificity achieved for serum testing. Reactivity to N antigen also was detected with 92.6% sensitivity and 90.7% specificity at 4–8 weeks after diagnostic testing. Moreover, serologic testing for endemic coronaviruses performed in multiplex with SARS-CoV-2 antigens has the potential to identify samples that may require retesting due to effects of pre-analytical factors. The easy-to-use saliva collection kit, coupled with thresholds for positivity and methods of flagging samples for retest, provides a framework for large-scale serosurveillance of SARS-CoV-2.
Collapse
Affiliation(s)
- Christopher Campbell
- Meso Scale Diagnostics, LLC., Rockville, MD, USA. .,Meso Scale Diagnostics, LLC, 16020 Industrial Drive, Gaithersburg, MD, 20877, USA.
| | - Douglas Roblin
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente, Rockville, MD, USA
| | | | | | - Jessica Joe
- Meso Scale Diagnostics, LLC., Rockville, MD, USA
| | - Lily Fathi
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente, Rockville, MD, USA
| | - Thomas Whiting
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente, Rockville, MD, USA
| | - Jared Williamson
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente, Rockville, MD, USA
| | - Paul Goodwin
- Meso Scale Diagnostics, LLC., Rockville, MD, USA
| | - Charmaine Mckie
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente, Rockville, MD, USA
| | - Adrienne Deneal
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente, Rockville, MD, USA
| | - Leslie Greenberg
- Mid-Atlantic Permanente Research Institute, Kaiser Permanente, Rockville, MD, USA
| | - George Sigal
- Meso Scale Diagnostics, LLC., Rockville, MD, USA
| |
Collapse
|
146
|
He Q, Mao Q, Zhang J, Gao F, Bai Y, Cui B, Liu J, An C, Wang Q, Yan X, Yang J, Song L, Song Z, Liu D, Yuan Y, Sun J, Zhao J, Bian L, Wu X, Huang W, Li C, Wang J, Liang Z, Xu M. Heterologous immunization with adenovirus vectored and inactivated vaccines effectively protects against SARS-CoV-2 variants in mice and macaques. Front Immunol 2022; 13:949248. [PMID: 36059554 PMCID: PMC9428284 DOI: 10.3389/fimmu.2022.949248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
To cope with the decline in COVID-19 vaccine-induced immunity caused by emerging SARS-CoV-2 variants, a heterologous immunization regimen using chimpanzee adenovirus vectored vaccine expressing SARS-CoV-2 spike (ChAd-S) and an inactivated vaccine (IV) was tested in mice and non-human primates (NHPs). Heterologous regimen successfully enhanced or at least maintained antibody and T cell responses and effectively protected against SARS-CoV-2 variants in mice and NHPs. An additional heterologous booster in mice further improved and prolonged the spike-specific antibody response and conferred effective neutralizing activity against the Omicron variant. Interestingly, priming with ChAd-S and boosting with IV reduced the lung injury risk caused by T cell over activation in NHPs compared to homologous ChAd-S regimen, meanwhile maintained the flexibility of antibody regulation system to react to virus invasion by upregulating or preserving antibody levels. This study demonstrated the satisfactory compatibility of ChAd-S and IV in prime-boost vaccination in animal models.
Collapse
Affiliation(s)
- Qian He
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Qunying Mao
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Jialu Zhang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Fan Gao
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Yu Bai
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Bopei Cui
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Jianyang Liu
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Chaoqiang An
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Qian Wang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Xujia Yan
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Jinghuan Yang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Lifang Song
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Ziyang Song
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Dong Liu
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Yadi Yuan
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Jing Sun
- Guangzhou Laboratory, Guangzhou, China
| | | | - Lianlian Bian
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Xing Wu
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Weijin Huang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Changgui Li
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
| | - Junzhi Wang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
- *Correspondence: Junzhi Wang, ; Zhenglun Liang, ; Miao Xu,
| | - Zhenglun Liang
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
- *Correspondence: Junzhi Wang, ; Zhenglun Liang, ; Miao Xu,
| | - Miao Xu
- Division of Hepatitis and Enterovirus Vaccines, Institute of Biological Products, National Institutes for Food and Drug Control, Beijing, China
- *Correspondence: Junzhi Wang, ; Zhenglun Liang, ; Miao Xu,
| |
Collapse
|
147
|
Association between Vitamin D Serum Levels and Immune Response to the BNT162b2 Vaccine for SARS-CoV-2. Biomedicines 2022; 10:biomedicines10081993. [PMID: 36009540 PMCID: PMC9405810 DOI: 10.3390/biomedicines10081993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022] Open
Abstract
The use of micronutrients such as vitamin D could improve the response to viral vaccines, particularly in immunosuppressed and immunosenescent subjects. Here, we analysed the association between serum 25-hydroxyvitamin D (25OHD) levels and the immune response elicited by the BNT162b2 vaccine in a cohort of 101 healthcare workers naïve for SARS-CoV-2 infection. We observed no significant differences in anti-spike (S) IgG and T-cell responses according to the 25OHD status at baseline. However, significant correlations between the 25OHD concentration at baseline and (i) the anti-S response (p < 0.020) and (ii) the neutralizing antibody (NT) titre (p = 0.040) at six months after the second dose were detected. We concluded that adequate levels of vitamin D may improve the immune response to mRNA vaccines such as BNT162b2, and that further larger studies are warranted in order to confirm these preliminary observations.
Collapse
|
148
|
Zhang Y, Lu M, Mahesh KC, Kim E, Shamseldin MM, Ye C, Dravid P, Chamblee M, Park JG, Hall JM, Trivedi S, Chaiwatpongsakorn S, Kenny AD, Murthy SS, Sharma H, Liang X, Yount JS, Kapoor A, Martinez-Sobrido L, Dubey P, Boyaka PN, Peeples ME, Li J. A highly efficacious live attenuated mumps virus-based SARS-CoV-2 vaccine candidate expressing a six-proline stabilized prefusion spike. Proc Natl Acad Sci U S A 2022; 119:e2201616119. [PMID: 35895717 PMCID: PMC9388148 DOI: 10.1073/pnas.2201616119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/24/2022] [Indexed: 11/18/2022] Open
Abstract
With the rapid increase in SARS-CoV-2 cases in children, a safe and effective vaccine for this population is urgently needed. The MMR (measles/mumps/rubella) vaccine has been one of the safest and most effective human vaccines used in infants and children since the 1960s. Here, we developed live attenuated recombinant mumps virus (rMuV)-based SARS-CoV-2 vaccine candidates using the MuV Jeryl Lynn (JL2) vaccine strain backbone. The soluble prefusion SARS-CoV-2 spike protein (preS) gene, stablized by two prolines (preS-2P) or six prolines (preS-6P), was inserted into the MuV genome at the P-M or F-SH gene junctions in the MuV genome. preS-6P was more efficiently expressed than preS-2P, and preS-6P expression from the P-M gene junction was more efficient than from the F-SH gene junction. In mice, the rMuV-preS-6P vaccine was more immunogenic than the rMuV-preS-2P vaccine, eliciting stronger neutralizing antibodies and mucosal immunity. Sera raised in response to the rMuV-preS-6P vaccine neutralized SARS-CoV-2 variants of concern, including the Delta variant equivalently. Intranasal and/or subcutaneous immunization of IFNAR1-/- mice and golden Syrian hamsters with the rMuV-preS-6P vaccine induced high levels of neutralizing antibodies, mucosal immunoglobulin A antibody, and T cell immune responses, and were completely protected from challenge by both SARS-CoV-2 USA-WA1/2020 and Delta variants. Therefore, rMuV-preS-6P is a highly promising COVID-19 vaccine candidate, warranting further development as a tetravalent MMR vaccine, which may include protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Yuexiu Zhang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Mijia Lu
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - K C Mahesh
- Center for Vaccines and Immunity, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205
| | - Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Mohamed M. Shamseldin
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Chengjin Ye
- Department of Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX 78227
| | - Piyush Dravid
- Center for Vaccines and Immunity, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205
| | - Michelle Chamblee
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Jun-Gyu Park
- Department of Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX 78227
| | - Jesse M. Hall
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Sheetal Trivedi
- Center for Vaccines and Immunity, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205
| | - Supranee Chaiwatpongsakorn
- Center for Vaccines and Immunity, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205
| | - Adam D. Kenny
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Satyapramod Srinivasa Murthy
- Center for Vaccines and Immunity, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205
| | - Himanshu Sharma
- Center for Vaccines and Immunity, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205
| | - Xueya Liang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
| | - Jacob S. Yount
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210
| | - Amit Kapoor
- Center for Vaccines and Immunity, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Luis Martinez-Sobrido
- Department of Disease Intervention and Prevention, Texas Biomedical Research Institute, San Antonio, TX 78227
| | - Purnima Dubey
- Department of Microbial Infection and Immunity, College of Medicine, The Ohio State University, Columbus, OH 43210
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210
| | - Prosper N. Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210
| | - Mark E. Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43205
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210
| | - Jianrong Li
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH 43210
- Infectious Disease Institute, The Ohio State University, Columbus, OH 43210
| |
Collapse
|
149
|
Vallivedu Chennakesavulu P, Cuddapah GV, Keesara MR, Inampudi JV, Arremsetty A, Moka S, Miryala S. COVID-19 Vaccine Clinical Trials: A Bird’s Eye Perspective. Cureus 2022; 14:e28066. [PMID: 36120231 PMCID: PMC9477158 DOI: 10.7759/cureus.28066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2022] [Indexed: 11/23/2022] Open
Abstract
Several Phase-III clinical studies investigating vaccine safety and effectiveness have been published a year following the first breakout of the COVID-19 pandemic. These vaccine candidates were produced using a variety of vaccination technologies, including mRNA, recombinant protein, adenoviral vector, and inactivated virus-based platforms, by various research organizations and pharmaceutical firms. Despite many successful clinical studies, participants are restricted by trial inclusion and exclusion criteria, geographic location, and the current state of the virus epidemic. Many concerns remain, particularly for specific populations such as the elderly, women who are pregnant or nursing, and teenagers. Vaccine effectiveness against asymptomatic infection and particular viral variations, on the other hand, is still largely unclear. This review will focus on vaccination candidates that have completed Phase-III clinical trials and will examine the scientific evidence that has been gathered so far for these vaccine candidates for various subgroups of individuals and virus variations.
Collapse
|
150
|
Oâ Connor MA, Hawman DW, Meade-White K, Leventhal S, Song W, Randall S, Archer J, Lewis TB, Brown B, Iwayama N, Ahrens C, Garrison W, Wangari S, Guerriero KA, Hanley P, Lovaglio J, Saturday G, Edlefsen PT, Khandhar A, Feldmann H, Fuller DH, Erasmus JH. A replicon RNA vaccine induces durable protective immunity from SARS-CoV-2 in nonhuman primates after neutralizing antibodies have waned. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.08.08.503239. [PMID: 35982677 PMCID: PMC9387133 DOI: 10.1101/2022.08.08.503239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The global SARS-CoV-2 pandemic prompted rapid development of COVID-19 vaccines. Although several vaccines have received emergency approval through various public health agencies, the SARS-CoV-2 pandemic continues. Emergent variants of concern, waning immunity in the vaccinated, evidence that vaccines may not prevent transmission and inequity in vaccine distribution have driven continued development of vaccines against SARS-CoV-2 to address these public health needs. In this report, we evaluated a novel self-amplifying replicon RNA vaccine against SARS-CoV-2 in a pigtail macaque model of COVID-19 disease. We found that this vaccine elicited strong binding and neutralizing antibody responses. While binding antibody responses were sustained, neutralizing antibody waned to undetectable levels after six months but were rapidly recalled and conferred protection from disease when the animals were challenged 7 months after vaccination as evident by reduced viral replication and pathology in the lower respiratory tract, reduced viral shedding in the nasal cavity and lower concentrations of pro-inflammatory cytokines in the lung. Cumulatively, our data demonstrate in pigtail macaques that a self-amplifying replicon RNA vaccine can elicit durable and protective immunity to SARS-CoV-2 infection. Furthermore, these data provide evidence that this vaccine can provide durable protective efficacy and reduce viral shedding even after neutralizing antibody responses have waned to undetectable levels.
Collapse
|