1
|
Ohno M, Sekiya T, Obeng-Kyeremeh R, Handabile C, Haruta M, Nomura N, Kawakita T, Shingai M, Kida H. Optimization of the preparation method of inactivated intact virus particle vaccine for COVID-19. Vaccine 2025; 56:127173. [PMID: 40279928 DOI: 10.1016/j.vaccine.2025.127173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/15/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
It has been recognized that it is difficult to maintain the virus particle structure of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which may be associated with lower immunogenicity of inactivated vaccines against coronavirus disease 2019 (COVID-19). We have previously demonstrated that an intact structure of the virus particles is critical for influenza inactivated whole virus particle vaccine (WPV) to be immunogenically potent. Here, we tested 37, 35, 33, and 31 °C for the virus propagation temperatures and the timing of formaldehyde treatment of the virus before and after centrifugation-based purification to obtain virus particles with an intact structure. Virus particles cultured at 33 °C retained spike proteins on the surface the most abundantly. The pretreatment of the virus with formaldehyde prevented the dissociation of the spike proteins from the viral surface during the centrifugation-based purification. The immunogenicity of the prepared vaccines, intact WPV and non-intact WPV that had lost the spike proteins, was evaluated in a mouse model. A single dose of intact WPV effectively induced humoral immunity compared to non-intact WPV, as indicated by higher titers of neutralizing antibodies. After a virus challenge, the mice vaccinated with a single dose of inactivated intact WPV showed less severe weight loss and lower virus titers in the lungs compared to those vaccinated with non-intact WPV. These results demonstrate the importance of the structural integrity of WPV in inducing effective and protective immunity, and provide significant insight into the development of COVID-19 WPV for practical use.
Collapse
Affiliation(s)
- Marumi Ohno
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan; One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Toshiki Sekiya
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan; International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Richard Obeng-Kyeremeh
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Chimuka Handabile
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
| | - Minori Haruta
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
| | - Naoki Nomura
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Tomomi Kawakita
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan; Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo, Japan
| | - Masashi Shingai
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan; International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo, Japan
| | - Hiroshi Kida
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan; International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan; Division of Vaccine Immunology, Hokkaido University International Institute for Zoonosis Control, Sapporo, Japan.
| |
Collapse
|
2
|
Kawakita T, Sekiya T, Kameda Y, Nomura N, Ohno M, Handabile C, Yamaya A, Fukuhara H, Anraku Y, Kita S, Toba S, Tsukamoto H, Sawa T, Oshiumi H, Itoh Y, Maenaka K, Sato A, Sawa H, Suzuki Y, Brown LE, Jackson DC, Kida H, Matsumoto M, Seya T, Shingai M. ARNAX is an ideal adjuvant for COVID-19 vaccines to enhance antigen-specific CD4 + and CD8 + T-cell responses and neutralizing antibody induction. J Virol 2025; 99:e0229024. [PMID: 40231823 PMCID: PMC12090777 DOI: 10.1128/jvi.02290-24] [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: 01/13/2025] [Accepted: 03/02/2025] [Indexed: 04/16/2025] Open
Abstract
ARNAX is a synthetic nucleotide-based Toll-like receptor 3 (TLR3) ligand that specifically stimulates the TLR3/TIR domain-containing adaptor molecule 1 (TICAM-1) pathway without activating inflammatory responses. ARNAX activates cellular immunity via cross-presentation; hence, its practical application has been demonstrated in cancer immunotherapy. Given the importance of cellular immunity in virus infections, ARNAX is expected to be a more effective vaccine adjuvant for virus infections than alum, an adjuvant approved for human use that mainly enhances humoral immunity. In the present study, the trimeric recombinant spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was prepared as a vaccine antigen and formulated with ARNAX. When T-cell and neutralizing antibody responses were evaluated in immunized mice, antigen formulated with ARNAX generated significantly larger numbers of antigenspecific CD4+ and CD8+ T cells, as well as higher titers of neutralizing antibodies, compared to antigen alone or antigen formulated with alum. In experiments where immunized mice were challenged with a SARS-CoV-2 mouse-adapted virus derived from the ancestral strain, immunization with antigen formulated with ARNAX reduced virus titers in the lungs at 3 days post-infection to a much greater extent than did immunization with either antigen alone or that formulated with alum. These results show that ARNAX potently enhances the levels of both cellular and humoral immunity above those seen with alum, providing significantly greater viral clearing responses. Thus, ARNAX may act as a useful adjuvant for prophylactic vaccines, particularly for viral infectious diseases. IMPORTANCE Cellular immunity is a critical immunological defense system against virus infections. However, aluminum salts, the most widely used adjuvant for vaccines for human use, do not promote strong cellular immunity. To prepare for the next pandemic of viral origin, the development of Th1-type adjuvants with low adverse reactions that induce cellular immunity is necessary. ARNAX is a TLR3 agonist consisting of DNA-RNA hybrid nucleic acid, which is expected to be an adjuvant that induces cellular immunity. The present study using a coronavirus disease 2019 mouse model demonstrated that ARNAX potently induces cellular immunity in addition to humoral immunity with minimal induction of inflammatory cytokines. Therefore, ARNAX has the potential to be used as a potent and welltolerated adjuvant for vaccines against pandemic viruses emerging in the future.
Collapse
Affiliation(s)
- Tomomi Kawakita
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Toshiki Sekiya
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- The Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Yayoi Kameda
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Naoki Nomura
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Marumi Ohno
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Chimuka Handabile
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Akari Yamaya
- Nebuta Research Institute for Life Sciences, Aomori University, Aomori, Japan
| | - Hideo Fukuhara
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Pathogen Structure, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yuki Anraku
- Laboratory of Biomolecular Science, and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Shunsuke Kita
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Laboratory of Biomolecular Science, and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Shinsuke Toba
- Shionogi Pharmaceutical Research Center, Shionogi & Company, Limited, Toyonaka, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Hirotake Tsukamoto
- Division of Clinical Immunology and Cancer Immunotherapy, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasushi Itoh
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Katsumi Maenaka
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Pathogen Structure, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Laboratory of Biomolecular Science, and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Japan
| | - Akihiko Sato
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Shionogi Pharmaceutical Research Center, Shionogi & Company, Limited, Toyonaka, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Hirofumi Sawa
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yasuhiko Suzuki
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Lorena E. Brown
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- The Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - David C. Jackson
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- The Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Hiroshi Kida
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Misako Matsumoto
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Nebuta Research Institute for Life Sciences, Aomori University, Aomori, Japan
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tsukasa Seya
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Nebuta Research Institute for Life Sciences, Aomori University, Aomori, Japan
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masashi Shingai
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| |
Collapse
|
3
|
Posa A. Spike protein-related proteinopathies: A focus on the neurological side of spikeopathies. Ann Anat 2025; 260:152662. [PMID: 40254264 DOI: 10.1016/j.aanat.2025.152662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND The spike protein (SP) is an outward-projecting transmembrane glycoprotein on viral surfaces. SARS-CoV-2 (Severe Acute Respiratory Syndrome Coronavirus 2), responsible for COVID-19 (Coronavirus Disease 2019), uses SP to infect cells that express angiotensin converting enzyme 2 (ACE2) on their membrane. Remarkably, SP has the ability to cross the blood-brain barrier (BBB) into the brain and cause cerebral damage through various pathomechanisms. To combat the COVID-19 pandemic, novel gene-based products have been used worldwide to induce human body cells to produce SP to stimulate the immune system. This artificial SP also has a harmful effect on the human nervous system. STUDY DESIGN Narrative review. OBJECTIVE This narrative review presents the crucial role of SP in neurological complaints after SARS-CoV-2 infection, but also of SP derived from novel gene-based anti-SARS-CoV-2 products (ASP). METHODS Literature searches using broad terms such as "SARS-CoV-2", "spike protein", "COVID-19", "COVID-19 pandemic", "vaccines", "COVID-19 vaccines", "post-vaccination syndrome", "post-COVID-19 vaccination syndrome" and "proteinopathy" were performed using PubMed. Google Scholar was used to search for topic-specific full-text keywords. CONCLUSIONS The toxic properties of SP presented in this review provide a good explanation for many of the neurological symptoms following SARS-CoV-2 infection and after injection of SP-producing ASP. Both SP entities (from infection and injection) interfere, among others, with ACE2 and act on different cells, tissues and organs. Both SPs are able to cross the BBB and can trigger acute and chronic neurological complaints. Such SP-associated pathologies (spikeopathies) are further neurological proteinopathies with thrombogenic, neurotoxic, neuroinflammatory and neurodegenerative potential for the human nervous system, particularly the central nervous system. The potential neurotoxicity of SP from ASP needs to be critically examined, as ASPs have been administered to millions of people worldwide.
Collapse
Affiliation(s)
- Andreas Posa
- University Clinics and Outpatient Clinics for Radiology, Neuroradiology and Neurology, Martin Luther University Halle-Wittenberg, Ernst-Grube-Straße 40, Halle 06120, Germany.
| |
Collapse
|
4
|
Cilloniz C, Videla AJ, Pulido L, Uy-King MJ. Viral community-acquired pneumonia: what's new since COVID-19 emerged? Expert Rev Respir Med 2025; 19:347-362. [PMID: 40077864 DOI: 10.1080/17476348.2025.2479611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/28/2025] [Accepted: 03/11/2025] [Indexed: 03/14/2025]
Abstract
INTRODUCTION All over the world, viral pneumonia has a significant impact on morbidity and mortality, especially among vulnerable populations. The most common respiratory viruses causing pneumonia include influenza virus, respiratory syncytial virus, adenoviruses and rhinovirus. The COVID-19 pandemic has changed the landscape of viral pneumonia and has reshaped our understanding of the role of viruses in this disease. We are now more aware of the importance of early diagnosis, the impact of co-infections, the effects of viral variants, and the long-term consequences of post-viral pneumonia. AREAS COVERED We discuss the latest scientific evidence regarding epidemiology, diagnosis, treatment, and prevention of viral pneumonia. This review summarizes findings from a PubMed search on respiratory viruses in community-acquired pneumonia. EXPERT OPINION Our experience during the COVID-19 pandemic has changed our perspective on respiratory viruses and their role in viral pneumonia. Diagnostic advances have been made, co-infections have received greater recognition, immune responses to viral infections are better understood, and approaches to treating viral pneumonia have expanded. Despite this progress, however, research on the impact of respiratory viruses on pneumonia must continue to pursue the development of new antivirals and vaccines, and investigate the long-term sequelae, especially in cases of severe viral pneumonia.
Collapse
Affiliation(s)
- Catia Cilloniz
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Faculty of Health Sciences, Continental University, Huancayo, Peru
| | - Alejandro J Videla
- Pulmonology Department, University Austral Hospital, Austral University, Buenos Aires, Argentina
| | - Laura Pulido
- Pulmonology Department, Italian Hospital of Rosario, Rosario, Argentina
| | - Mary Joy Uy-King
- Chairman Medical Research and Training Committee, Healthway QualiMed Hospital, San Jose Del Monte, Philippines
| |
Collapse
|
5
|
Rajan AK, Bashar MDA. Serious adverse events following immunization and predictors of mortality associated with COVID-19 vaccination in India: a secondary data analysis of nationwide causality assessments. Ther Adv Vaccines Immunother 2025; 13:25151355251321697. [PMID: 40034661 PMCID: PMC11873861 DOI: 10.1177/25151355251321697] [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: 08/21/2024] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Background Vaccines against COVID-19 were viewed as a way out to the ongoing pandemic and were given the emergency use authorization in India to initiate mass vaccination in January 2021. This study aimed to investigate the serious adverse events following immunizations (AEFIs) reported for COVID-19 vaccines and to identify predictors of mortality among these cases from India. Methods A secondary data analysis was conducted on the causality assessment reports for the 2708 serious AEFIs published by the National AEFI Committee under the Immunization Division, Ministry of Health and Family Welfare, Government of India. The analysis included all 21 reports published up until May 2023. The primary outcome variable analyzed was the survival/death status of each AEFI case, with various covariates from the published documents considered in the analysis. Results The majority of the serious AEFIs assessed were either coincidental (1220, 45%) or undetermined/unclassifiable (781, 28.8%). The majority of the serious AEFIs were reported among recipients of Covishield (1891, 69.8%) followed by Covaxin (347, 12.8%). Among these, 1114 (42.1%) died while the remaining 1594 (58.9%) were hospitalized and recovered. Systematically, AEFIs involving the cardiovascular system (696, 31.3%) were the most common, followed by those affecting the respiratory system (288, 13%) and neuropsychiatric system (295, 13.3%) which had a significant association with age (p < 0.001) and gender (p < 0.001). On multivariable analysis, females (p = 0.001), younger age groups (p < 0.001), AEFIs whose causality was determined and classified (p < 0.001), AEFI involving gastrointestinal and neuropsychiatric system (p < 0.001), AEFIs reported from North and Western India (p = 0.001) and those occurring during the winter season (p < 0.05) had significantly lower odds of mortality. Conclusion Among the cohort of serious AEFIs reported, older age, male sex, undetermined or unclassifiable causality classification, and involvement of the cardiovascular system were associated with significantly higher odds of mortality and require close monitoring following vaccination.
Collapse
Affiliation(s)
- Abin Kulathunkal Rajan
- Department of Community and Family Medicine, All India Institute of Medical Sciences, Gorakhpur, Uttar Pradesh, India
| | - MD. Abu Bashar
- Department of Community and Family Medicine, All India Institute of Medical Sciences, Room No. 229, Gorakhpur, Uttar Pradesh 273008, India
| |
Collapse
|
6
|
Li Y, Viswaroopan D, He W, Li J, Zuo X, Xu H, Tao C. Improving entity recognition using ensembles of deep learning and fine-tuned large language models: A case study on adverse event extraction from VAERS and social media. J Biomed Inform 2025; 163:104789. [PMID: 39923968 DOI: 10.1016/j.jbi.2025.104789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 01/07/2025] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
OBJECTIVE Adverse event (AE) extraction following COVID-19 vaccines from text data is crucial for monitoring and analyzing the safety profiles of immunizations, identifying potential risks and ensuring the safe use of these products. Traditional deep learning models are adept at learning intricate feature representations and dependencies in sequential data, but often require extensive labeled data. In contrast, large language models (LLMs) excel in understanding contextual information, but exhibit unstable performance on named entity recognition (NER) tasks, possibly due to their broad but unspecific training. This study aims to evaluate the effectiveness of LLMs and traditional deep learning models in AE extraction, and to assess the impact of ensembling these models on performance. METHODS In this study, we utilized reports and posts from the Vaccine Adverse Event Reporting System (VAERS) (n = 230), Twitter (n = 3,383), and Reddit (n = 49) as our corpora. Our goal was to extract three types of entities: vaccine, shot, and adverse event (ae). We explored and fine-tuned (except GPT-4) multiple LLMs, including GPT-2, GPT-3.5, GPT-4, Llama-2 7b, and Llama-2 13b, as well as traditional deep learning models like Recurrent neural network (RNN) and Bidirectional Encoder Representations from Transformers for Biomedical Text Mining (BioBERT). To enhance performance, we created ensembles of the three models with the best performance. For evaluation, we used strict and relaxed F1 scores to evaluate the performance for each entity type, and micro-average F1 was used to assess the overall performance. RESULTS The ensemble demonstrated the best performance in identifying the entities "vaccine," "shot," and "ae," achieving strict F1-scores of 0.878, 0.930, and 0.925, respectively, and a micro-average score of 0.903. These results underscore the significance of fine-tuning models for specific tasks and demonstrate the effectiveness of ensemble methods in enhancing performance. CONCLUSION In conclusion, this study demonstrates the effectiveness and robustness of ensembling fine-tuned traditional deep learning models and LLMs, for extracting AE-related information following COVID-19 vaccination. This study contributes to the advancement of natural language processing in the biomedical domain, providing valuable insights into improving AE extraction from text data for pharmacovigilance and public health surveillance.
Collapse
Affiliation(s)
- Yiming Li
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Deepthi Viswaroopan
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - William He
- Department of Electrical & Computer Engineering, Pratt School of Engineering, Duke University, 305 Tower Engineering Building, Durham, NC 27708, USA
| | - Jianfu Li
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Xu Zuo
- McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Hua Xu
- Department of Biomedical Informatics and Data Science, School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Cui Tao
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
7
|
Padhi BK, Gupta PC, Al Kaabi NA, Al-Subaie MF, Alrasheed HA, Sulaiman T, Rabaan AA, Khatib MN, Gaidhane S, Zahiruddin QS, Dziedzic A, Satapathy P, Fraj NJ, Aldrazi FA, Kukreti N, Rustagi S, Abu Serhan H. Incidence and Association of Uveitis with COVID-19 Vaccination: A Systematic Review and Meta-Analysis. Ophthalmic Epidemiol 2025; 32:112-120. [PMID: 38709174 DOI: 10.1080/09286586.2024.2343714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 04/02/2024] [Accepted: 04/10/2024] [Indexed: 05/07/2024]
Abstract
PURPOSE In the wake of the COVID-19 pandemic, vaccines have been pivotal in curbing disease spread and severity. However, concerns over post-vaccination adverse events, including uveitis, an inflammatory ocular condition, have been noted. This systematic review and meta-analysis aimed to evaluate the incidence and association of uveitis following COVID-19 vaccination. METHODS A literature search was performed across several databases on October 21, 2023. Human studies examining the incidence of uveitis post-COVID-19 vaccination were included. The Newcastle-Ottawa Scale was used for quality appraisal of the included studies. Meta-analysis was performed to assess the overall incidence of uveitis and the relative risk of developing the condition post-vaccination. All statistical analyses were performed using R software version 4.3. RESULTS Six studies involving over 2 billion vaccine doses were included. The overall incidence of uveitis was 0.016% (95% CI: 0.010 to 0.026). No significant association was found between vaccination and the onset of uveitis (Relative Risk: 1.45 (95% CI: 0.82 to 2.57, p = 0.12) from four studies. The evidence quality was rated very low due to the limited number of studies and imprecision. CONCLUSION This analysis indicates a low incidence of uveitis following COVID-19 vaccination and no significant association with the vaccine. The findings are constrained by the small number of studies and low certainty of evidence, underscoring the need for further research. Comprehensive and longitudinal studies are necessary to confirm these findings and reinforce public confidence in COVID-19 vaccination programs.
Collapse
Affiliation(s)
- Bijaya K Padhi
- Department of Community Medicine and School of Public Health, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Parul Chawla Gupta
- Department of Ophthalmology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Nawal A Al Kaabi
- College of Medicine and Health Science, Khalifa University, Abu Dhabi, United Arab Emirates
- Sheikh Khalifa Medical City, Abu Dhabi Health Services Company (SEHA), Abu Dhabi, United Arab Emirates
| | - Maha F Al-Subaie
- Research Center, Dr. Sulaiman Alhabib Medical Group, Riyadh, Saudi Arabia
- Department of Infectious Diseases, Dr. Sulaiman Alhabib Medical Group, Riyadh, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - Hayam A Alrasheed
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Tarek Sulaiman
- Infectious Diseases Section, Medical Specialties Department, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Ali A Rabaan
- College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia
- Department of Public Health and Nutrition, The University of Haripur, Haripur, Pakistan
| | - Mahalaqua Nazli Khatib
- Division of Evidence Synthesis, Global Consortium of Public Health and Research, Datta Meghe Institute of Higher Education, Wardha, India
| | - Shilpa Gaidhane
- One Health Centre (COHERD), Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education, Wardha, India
| | - Quazi Syed Zahiruddin
- South Asia Infant Feeding Research Network (SAIFRN), Division of Evidence Synthesis, Global Consortium of Public Health and Research, Datta Meghe Institute of Higher Education, Wardha, India
| | - Arkadiusz Dziedzic
- Department of Conservative Dentistry with Endodontics, Medical University of Silesia, Katowice, Poland
| | - Prakasini Satapathy
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Najat J Fraj
- Infection Prevention and Control Department, Dammam Medical Complex, Dammam, Saudi Arabia
| | | | - Neelima Kukreti
- School of Pharmacy, Graphic Era Hill University, Dehradun, India
| | - Sarvesh Rustagi
- School of Applied and Life Sciences, Uttaranchal University, Dehradun, India
| | | |
Collapse
|
8
|
Debbag R, Rudin D, Ceddia F, Watkins J. The Impact of Vaccination on COVID-19, Influenza, and Respiratory Syncytial Virus-Related Outcomes: A Narrative Review. Infect Dis Ther 2025; 14:63-97. [PMID: 39739199 PMCID: PMC11724835 DOI: 10.1007/s40121-024-01079-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/06/2024] [Indexed: 01/02/2025] Open
Abstract
Vaccination represents a core preventive strategy for public health, with interrelated and multifaceted effects across health and socioeconomic domains. Beyond immediate disease prevention, immunization positively influences downstream health outcomes by mitigating complications of preexisting comorbidities and promoting healthy aging. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza virus, and respiratory syncytial virus (RSV) are common respiratory viruses responsible for broad societal cost and substantial morbidity and mortality, particularly among at-risk individuals, including older adults and people with frailty or certain comorbid conditions. In this narrative review, we summarize the overall impact of vaccination for these 3 viruses, focusing on mRNA vaccines, each of which exhibits unique patterns of infection, risk, and transmission dynamics, but collectively represent a target for preventive strategies. Vaccines for COVID-19 (caused by SARS-CoV-2) and influenza are effective against the most severe outcomes, such as hospitalization and death; these vaccines represent the most potent and cost-effective interventions for the protection of population and individual health against COVID-19 and influenza, particularly for older adults and those with comorbid conditions. Based on promising results of efficacy for the prevention of RSV-associated lower respiratory tract disease, the first RSV vaccines were approved in 2023. Immunization strategies should account for various factors leading to poor uptake, including vaccine hesitancy, socioeconomic barriers to access, cultural beliefs, and lack of knowledge of vaccines and disease states. Coadministration of vaccines and combination vaccines, such as multicomponent mRNA vaccines, offer potential advantages in logistics and delivery, thus improving uptake and reducing barriers to adoption of new vaccines. The success of the mRNA vaccine platform was powerfully demonstrated during the COVID-19 pandemic; these and other new approaches show promise as a means to overcome existing challenges in vaccine development and to sustain protection against viral changes over time.A graphical abstract and video abstract is available with this article.
Collapse
Affiliation(s)
- Roberto Debbag
- Latin American Vaccinology Society, Buenos Aires, Argentina
| | | | | | - John Watkins
- Department of Population Medicine, Cardiff University, Cardiff, UK.
| |
Collapse
|
9
|
Balestracci B, Candido G, Federici L, Parretti C, Tartaglia R, Lachman P, Bianco A, La Regina M. Anti-COVID-19 Vaccination in the Italian General Population: Proactive Clinical Risk Analysis Using Failure Mode, Effects, and Criticality Analysis Technique. Healthcare (Basel) 2024; 12:2541. [PMID: 39765968 PMCID: PMC11675166 DOI: 10.3390/healthcare12242541] [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: 10/24/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Large-scale vaccination was crucial to address the global COVID-19 pandemic and its associated health risks, including fatal and disabling diseases. However, there were significant challenges to be overcome to ensure the safe and effective implementation of the vaccination program. The aim of the present study was to assess patient safety threats related to the anti-COVID-19 large-scale vaccination process. Methods: Between February and May 2021, we conducted a proper analysis to proactively identify risks and potential Failure Modes (FMs) in the COVID-19 vaccination process using the Failure Mode, Effects, and Criticality Analysis (FMECA) technique at an Italian Public Health Authority. A standardized risk scoring system was used to assess the severity, frequency, and detectability of events associated with potential failures. Criticalities were identified in both the preparatory and operational areas of the vaccination process, and several potential FMs were listed in descending order of risk score (Risk Priority Number, RPN) to ensure prioritization of interventions. Results: The most critical steps were found to be in the operational area rather than in the preparatory one. The highest RPNs were associated with failure or inadequate management of severe allergic reactions that can lead to serious harm and even death of the vaccinated person (RPN 60) and failure to keep updated vaccination teams' knowledge (RPN 36). Conclusions: Ensuring patient safety and effective clinical risk management are crucial in mass vaccination campaigns. By prioritizing these aspects through collaboration with various stakeholders and implementing preventive measures, patient trust-on which vaccination campaign success relies-can be built and maintained.
Collapse
Affiliation(s)
| | - Giuseppe Candido
- Department of Engineering Sciences, Guglielmo Marconi University, 00193 Rome, Italy; (G.C.); (C.P.)
| | - Lorenzo Federici
- Internal Medicine, Ligurian Health Authority n. 5, 19121 La Spezia, Italy;
| | - Chiara Parretti
- Department of Engineering Sciences, Guglielmo Marconi University, 00193 Rome, Italy; (G.C.); (C.P.)
| | - Riccardo Tartaglia
- Department of Engineering Sciences, Guglielmo Marconi University, 00193 Rome, Italy; (G.C.); (C.P.)
| | - Peter Lachman
- Department of Quality Improvement, Royal College of Physicians of Ireland, D02 X266 Dublin, Ireland;
| | - Alessandra Bianco
- Communications Department, Ligurian Health Authority n. 5, 19121 La Spezia, Italy;
| | - Micaela La Regina
- Clinical Governance and Risk Management, Ligurian Health Authority n. 5, 19121 La Spezia, Italy;
| |
Collapse
|
10
|
Mok CC, Cheung CCL, Chan KL, Tse SM, To CH. Effect of SARS-CoV2 infection on disease flares in patients with systemic lupus erythematosus: a case-control study. Rheumatology (Oxford) 2024; 63:3390-3396. [PMID: 37947324 DOI: 10.1093/rheumatology/kead601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 10/08/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023] Open
Abstract
OBJECTIVES To study the effect of SARS-CoV2 infection on flares of systemic lupus erythematosus (SLE). METHODS Patients who fulfilled the ACR/SLICC criteria for SLE and had documented COVID-19 between February and November 2022 were identified retrospectively from our hospital COVID-19 registry. SLE controls who did not have SARS-CoV2 infection were randomly matched for age, sex and the time of infection in a 2:1 ratio with those infected. The primary outcome of interest was clinical flare of SLE within 90 days of COVID-19. The rate of SLE flares (mild/moderate or severe) was compared between SARS-CoV2-infected SLE patients and controls. RESULTS Ninety-one SLE patients with COVID-19 (age 48.6 (14.0) years; 95.6% women) and 182 SLE controls (age 48.7 (13.8) years; 95.6% women) were studied. Eleven of 91 (12.1%) SARS-CoV2-infected patients had serious manifestations. One (1.1%) patient died and 7 (7.7%) developed severe complications. Within 90 days of SARS-CoV2 infection, 14 (15.4%) patients developed mild/moderate clinical SLE flares and two (2.2%) patients had severe SLE flares. The incidence of SLE flares in SARS-CoV2-infected patients was significantly higher than in those without the infection (17.6% vs 5.5%; odds ratio 3.67 [95% CI: 1.59, 8.46]; P = 0.001). The changes in anti-dsDNA and complement levels, however, were not significantly different between the two groups. Among SARS-CoV2-infected SLE patients, those with clinical SLE flares had significantly lower C3 values (P = 0.004) before the infection than those without. CONCLUSION Clinical flares within 90 days were significantly more common in SLE patients infected with SARS-CoV2 than matched non-infected SLE controls.
Collapse
Affiliation(s)
- Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, Hong Kong SAR, China
| | - Chris Ching Lam Cheung
- Department of Medicine, Tuen Mun Hospital, Hong Kong SAR, China
- Rutonjee Hospital, Hong Kong SAR, China
| | - Kar Li Chan
- Department of Medicine, Tuen Mun Hospital, Hong Kong SAR, China
| | - Sau Mei Tse
- Department of Medicine, Tuen Mun Hospital, Hong Kong SAR, China
| | - Chi Hung To
- Department of Medicine, Tuen Mun Hospital, Hong Kong SAR, China
- Pok Oi Hospital, Hong Kong SAR, China
| |
Collapse
|
11
|
Mohammadi D, Ghasemi M, Manouchehrian N, Zafarmand M, Akbari M, Boroumand AB. COVID-19 vaccines: current and future challenges. Front Pharmacol 2024; 15:1434181. [PMID: 39568586 PMCID: PMC11576167 DOI: 10.3389/fphar.2024.1434181] [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: 05/17/2024] [Accepted: 09/18/2024] [Indexed: 11/22/2024] Open
Abstract
As of December 2020, around 200 vaccine candidates for Coronavirus Disease 2019 (COVID-19) are being developed. COVID-19 vaccines have been created on a number of platforms and are still being developed. Nucleic acid (DNA, RNA) vaccines, viral vector vaccines, inactivated vaccines, protein subunit vaccines, and live attenuated vaccines are among the COVID-19 vaccine modalities. At this time, at least 52 candidate vaccines are being studied. Spike protein is the primary protein that COVID-19 vaccines are targeting. Therefore, it is critical to determine whether immunizations provide complete or fractional protection, whether this varies with age, whether vaccinated people are protected from reoccurring diseases, and whether they need booster shots if they've already been inoculated. Despite the enormous achievement of bringing several vaccine candidates to market in less than a year, acquiring herd immunity at the national level and much more so at the global level remains a major challenge. Therefore, we gathered information on the mechanism of action of presently available COVID-19 vaccines in this review and essential data on the vaccines' advantages and downsides and their future possibilities.
Collapse
Affiliation(s)
| | - Matin Ghasemi
- Islamic Azad University Tonekabon, Tonekabon, Mazandaran, Iran
| | - Nahid Manouchehrian
- Department of Anesthesilogy, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | | | - Mitra Akbari
- Eye Research Center, Department of Eye, Amiralmomenin Hospital, School of Medicine, Guilan University of Medical Science, Rasht, Iran
| | | |
Collapse
|
12
|
Samiei-Abianeh H, Nazarian S, Kordbacheh E, Felegary A. Recombinant receptor-binding motif of spike COVID-19 vaccine candidate induces SARS-CoV-2 neutralizing antibody response. BIOIMPACTS : BI 2024; 15:30520. [PMID: 40256231 PMCID: PMC12008496 DOI: 10.34172/bi.30520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/21/2024] [Accepted: 09/23/2024] [Indexed: 04/22/2025]
Abstract
Introduction The SARS-CoV-2 pandemic necessitates effective therapeutic solutions. The receptor-binding motif (RBM) is a subdomain of the spike protein's receptor-binding domain (RBD) and is critical for facilitating the binding of SARS-CoV-2 to the human ACE2 receptor. This study investigates the use of the receptor-binding motif (RBM) domain as an immunogen to produce potent neutralizing antibodies against SARS-CoV-2. Methods The RBM gene was codon-optimized and cloned into the pET17b vector for expression in E. coli BL21 (DE3) cells, induced with 1 mM IPTG. The recombinant RBM protein was purified using Ni-NTA affinity chromatography. After validating the recombinant RBM by Western blotting with anti-His tag antibodies, BALB/c mice were immunized with 20 µg of the purified RBM protein. Anti-RBM IgG was subsequently purified using protein G resin, and its neutralizing capacity was assessed using the Pishtaz Teb Zaman Neutralization Assay Kit. Results The recombinant RBM protein, with a molecular weight of 10 kDa, was expressed as inclusion bodies. the typical yield of purification was 27 mg/L of bacterial culture. The neutralization test demonstrated a concentration of 36 µg/mL of neutralizing antibodies in the immunized serum, preventing the spike protein from binding to ACE2. Conclusion Our study demonstrated that anti-RBM antibodies exhibited neutralization effects on SARS-CoV-2. These findings provide evidence for the development of a vaccine candidate through the induction of antibodies against the RBM, necessitating further studies with adjuvants suitable for human use to evaluate its potential for human vaccination.
Collapse
Affiliation(s)
- Hossein Samiei-Abianeh
- Department of Medical Biotechnology and Nanotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Biology, Faculty of Basic Sciences, Imam Hossein University, Tehran, Iran
| | - Shahram Nazarian
- Department of Biology, Faculty of Basic Sciences, Imam Hossein University, Tehran, Iran
| | - Emad Kordbacheh
- Department of Biology, Faculty of Basic Sciences, Imam Hossein University, Tehran, Iran
| | - Alireza Felegary
- Department of Biology, Faculty of Basic Sciences, Imam Hossein University, Tehran, Iran
| |
Collapse
|
13
|
Mirzakhani M, Bayat M, Dashti M, Tahmasebi S, Rostamtabar M, Esmaeili Gouvarchin Ghaleh H, Amani J. The Assessment of Anti-SARS-CoV-2 Antibodies in Different Vaccine Platforms: A Systematic Review and Meta-Analysis of COVID-19 Vaccine Clinical Trial Studies. Rev Med Virol 2024; 34:e2579. [PMID: 39327654 DOI: 10.1002/rmv.2579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/13/2024] [Accepted: 08/20/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND AND OBJECTIVE The COVID-19 pandemic spread rapidly throughout the world and caused millions of deaths globally. Several vaccines have been developed to control the COVID-19 pandemic and reduce the burden it placed on public health. This study aimed to assess the efficacy of different vaccine platforms in inducing potent antibody responses. Moreover, the seroconversion rate and common side effects of vaccine platforms were evaluated. METHODS This meta-analysis included clinical trials of COVID-19 vaccines that met the eligibility criteria. Electronic databases (including PubMed, Scopus, and Web of Science) and Google Scholar search engine were searched for eligible studies. Regarding the methodological heterogeneity between the included studies, we selected a random-effects model. The geometric mean ratio (GMR) was chosen as the effect size for this meta-analysis. RESULTS Of the 1838 records identified through screening and after removing duplicate records, the full texts of 1076 records were assessed for eligibility. After the full-text assessment, 56 records were eligible and included in the study. Overall, vaccinated participants had a 150.8-fold increased rate of anti-spike IgG titres compared with the placebo group (GMR = 150.8; 95% CI, 95.9-237.1; I2 = 100%). Moreover, vaccinated participants had a 37.3-fold increased rate of neutralising antibody titres compared with the placebo group (GMR = 37.3; 95% CI, 28.5-48.7; I2 = 99%). The mRNA platform showed a higher rate of anti-spike IgG (GMR = 1263.5; 95% CI, 431.1-3702.8; I2 = 99%), while neutralising antibody titres were higher in the subunit platform (GMR = 53.4; 95% CI, 32.8-87.1; I2 = 99%) than in other platforms. Different vaccine platforms showed different rates of both anti-spike IgG and neutralising antibody titres with interesting results. The seroconversion rate of anti-spike IgG and neutralising antibody titres was more than 98% in the vaccinated participants. CONCLUSION Inactivated and subunit vaccines produced a high percentage of neutralising antibodies and had a low common adverse reaction rate compared to other platforms. In this regard, subunit and inactivated vaccines can still be used as the main vaccine platforms for effectively controlling infections with high transmission rates.
Collapse
Affiliation(s)
- Mohammad Mirzakhani
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Maryam Bayat
- Department of Immunology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammadreza Dashti
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Kashmar School of Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Safa Tahmasebi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Rostamtabar
- Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Hadi Esmaeili Gouvarchin Ghaleh
- Applied Virology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Jafar Amani
- Applied Microbiology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
SeyedAlinaghi S, Pashapouryeganeh A, Dehghani S, Mirzapour P, Abbaspour F, Afroughi F, Rahimzadeh P, Najafi M, Ghasemi H, Mozafari N, Soltanali Z, Mehraeen E. Feasibility and Effectiveness of Vaccines for COVID-19: An Umbrella Review. ARCHIVES OF ACADEMIC EMERGENCY MEDICINE 2024; 13:e6. [PMID: 39318867 PMCID: PMC11417638 DOI: 10.22037/aaem.v12i1.2357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Introduction In January 2020, WHO declared the 2019 Coronavirus Disease (COVID-19) a pandemic. Though COVID-19 vaccines are recommended, ongoing surveillance is crucial due to potential unforeseen events. Evaluation of long-term effectiveness and safety and addressing emerging variants are vital. This study integrates systematic reviews to assess COVID-19 vaccine efficacy, immunogenicity, and safety comprehensively. Methods This study was an umbrella review study on the feasibility and effectiveness of vaccines for COVID-19. We conducted a comprehensive search in PubMed, Web of Sciences, and Scopus, using MeSH terms and keywords related to COVID-19 vaccines. Inclusion criteria comprised peer-reviewed systematic reviews and meta-analyses in English, focusing on feasibility and effectiveness. Exclusion criteria targeted non-systematic reviews exclusively on vaccine safety and duplicates. Two independent reviewers screened and resolved discrepancies. Data extraction included key details. Methodological quality was assessed using the ROBIS tool. Data synthesis involves narrative and, if applicable, quantitative synthesis (meta-analysis). Reporting followed PRISMA guidelines. Results A total of 32 systematic reviews were included in the study, of which 20 also conducted a meta-analysis. The studies investigated in the included reviews ranged from 7 to 74. The included articles were conducted in various countries around the globe. The findings indicated that COVID-19 vaccines are generally safe and effective for individuals with various medical conditions. The overall risk of bias for the included studies was assessed as low risk. Conclusion The study outcomes indicated that mRNA vaccines exhibit a higher incidence of adverse events but demonstrate greater efficacy. Conversely, inactivated and protein subunit vaccines are safer but exhibit lower efficiency. Moreover, the vaccine is considered safe for individuals with specific conditions such as inflammatory bowel disease, solid organ transplant recipients, children, pregnant individuals, and those with hematologic problems. Ultimately, the acceptance of the COVID-19 vaccine among individuals is influenced by various factors, including geographic, socioeconomic, and pandemic-related considerations.
Collapse
Affiliation(s)
- SeyedAhmad SeyedAlinaghi
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirreza Pashapouryeganeh
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
- Cofirst author
| | - Soheil Dehghani
- Prevention ofMetabolic Disorders Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pegah Mirzapour
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Faeze Abbaspour
- School ofMedicine, Tehran University ofMedical Sciences, Tehran, Iran
| | - Fatemeh Afroughi
- Iranian Research Center for HIV/AIDS, Iranian Institute for Reduction of High-Risk Behaviors, Tehran University of Medical Sciences, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Morvarid Najafi
- School ofMedicine, Tehran University ofMedical Sciences, Tehran, Iran
| | - Hoomaan Ghasemi
- School ofMedicine, Tehran University ofMedical Sciences, Tehran, Iran
| | - Navid Mozafari
- School ofMedicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Zahra Soltanali
- Medical student in Ilam University of Medical Sciences, Ilam, Iran
| | - Esmaeil Mehraeen
- Department of Health Information Technology, Khalkhal University of Medical Sciences, Khalkhal, Iran
| |
Collapse
|
15
|
Portilho AI, Silva VO, Da Costa HHM, Yamashiro R, de Oliveira IP, de Campos IB, Prudencio CR, Matsuda EM, de Macedo Brígido LF, De Gaspari E. An unexpected IgE anti-receptor binding domain response following natural infection and different types of SARS-CoV-2 vaccines. Sci Rep 2024; 14:20003. [PMID: 39198569 PMCID: PMC11358332 DOI: 10.1038/s41598-024-71047-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024] Open
Abstract
Humoral response to SARS-CoV-2 has been studied, predominantly the classical IgG and its subclasses. Although IgE antibodies are typically specific to allergens or parasites, a few reports describe their production in response to SARS-CoV-2 and other viruses. Here, we investigated IgE specific to receptor binding domain (RBD) of SARS-CoV-2 in a Brazilian cohort following natural infection and vaccination. Samples from 59 volunteers were assessed after infection (COVID-19), primary immunization with vectored (ChAdOx1) or inactivated (CoronaVac) vaccines, and booster immunization with mRNA (BNT162b2) vaccine. Natural COVID-19 induced IgE, but vaccination increased its levels. Subjects vaccinated with two doses of ChAdOx1 exhibited a more robust response than those immunized with two doses of CoronaVac; however, after boosting with BNT162b2, all groups presented similar IgE levels. IgE showed intermediate-to-high avidity, especially after the booster vaccine. We also found IgG4 antibodies, mainly after the booster, and they moderately correlated with IgE. ELISA results were confirmed by control assays, using IgG depletion by protein G and lack of reactivity with heterologous antigen. In our cohort, no clinical data could be associated with the IgE response. We advocate for further research on IgE and its role in viral immunity, extending beyond allergies and parasitic infections.
Collapse
Affiliation(s)
- Amanda Izeli Portilho
- Immunology Center, Adolfo Lutz Institute, Av Dr Arnaldo, 355, 11th floor, room 1116, Pacaembu, São Paulo, SP, 01246-902, Brazil
- Post Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, SP, Brazil
| | - Valéria Oliveira Silva
- Virology Center, Adolfo Lutz Institute, São Paulo, SP, Brazil
- Post Graduate Program in Public Health Surveillance, Disease Control Coordination, São Paulo, SP, Brazil
| | - Hernan Hermes Monteiro Da Costa
- Immunology Center, Adolfo Lutz Institute, Av Dr Arnaldo, 355, 11th floor, room 1116, Pacaembu, São Paulo, SP, 01246-902, Brazil
- Post Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, SP, Brazil
| | - Rosemeire Yamashiro
- Immunology Center, Adolfo Lutz Institute, Av Dr Arnaldo, 355, 11th floor, room 1116, Pacaembu, São Paulo, SP, 01246-902, Brazil
| | | | | | - Carlos Roberto Prudencio
- Immunology Center, Adolfo Lutz Institute, Av Dr Arnaldo, 355, 11th floor, room 1116, Pacaembu, São Paulo, SP, 01246-902, Brazil
- Post Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, SP, Brazil
| | | | - Luís Fernando de Macedo Brígido
- Virology Center, Adolfo Lutz Institute, São Paulo, SP, Brazil
- Post Graduate Program in Public Health Surveillance, Disease Control Coordination, São Paulo, SP, Brazil
| | - Elizabeth De Gaspari
- Immunology Center, Adolfo Lutz Institute, Av Dr Arnaldo, 355, 11th floor, room 1116, Pacaembu, São Paulo, SP, 01246-902, Brazil.
- Post Graduate Program Interunits in Biotechnology, University of São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
16
|
Lakin KS, Wu Y, Gordon JK, Kwakkenbos L, Carrier ME, Henry RS, Denton CP, Mouthon L, Spiera RF, Thombs BD. COVID-19 vaccinations and infections among individuals with systemic sclerosis: A Scleroderma Patient-centered Intervention Network (SPIN) Cohort study. Semin Arthritis Rheum 2024; 67:152453. [PMID: 38851172 DOI: 10.1016/j.semarthrit.2024.152453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 06/10/2024]
Abstract
BACKGROUND/PURPOSE We previously surveyed adults with systemic sclerosis (SSc) regarding COVID-19 vaccination in April-May 2021. The objective of the present study was to update through June-July 2022 and assess self-reported (1) COVID-19 vaccination rates, including boosters; (2) vaccine-related adverse events; (3) peri‑vaccination immunosuppressive medication management; (4) vaccine hesitancy; and (5) prevalence and severity of COVID-19 infections. METHODS In April-May 2021 and June-July 2022, SPIN Cohort participants completed surveys on COVID-19 vaccination and infection. Primary vaccine series was defined according to the standard for each COVID-19 vaccine; additional vaccine administrations were considered booster doses. Fully vaccinated was defined as having completed a primary vaccine series and at least one booster dose. RESULTS 544 participants completed the 2021 survey only, 101 the 2022 survey only, and 388 both surveys. Among 489 participants with 2022 data, 437 (89 %) had received both primary and booster vaccines. Among all 1,033 participants, 960 (93 %) received at least one dose. At least one adverse reaction was reported by 34 % (330 of 960 participants) following first, 48 % (314 of 657 participants) following second, and 34 % (147 of 437 participants) following booster vaccine doses (primarily sore arm and fatigue); no severe adverse reactions were reported. SSc symptom worsening was reported in 6 % (53 of 960) after the first, 6 % after the second (39 of 657), and 4 % (17 of 437) after the booster dose. Of participants taking methotrexate or mycophenolate (including Cellcept or Myfortic), 34 of 266 (13 %) reported that they temporarily stopped or decreased their medication at the first dose, 32 of 215 (15 %) at the second dose, and 28 of 148 (19 %) for booster vaccination. Of 52 individuals not fully vaccinated with primary and booster doses in 2022, 29 (56 %) reported worry about vaccine related SSc flares. 172 of 489 (35 %) 2022 participants reported a history of at least one COVID-19 infection; 114 (66 %) occurred after receiving at least a primary vaccine series. Among initial COVID-19 infections, 9 (5 %) were asymptomatic, 66 (38 %) involved mild symptoms, 82 (48 %) moderate symptoms, and 15 (9 %) required hospitalization. CONCLUSION Most people with SSc in the study were fully vaccinated, and most continued their methotrexate or mycophenolate post-primary and booster vaccinations. Over half of vaccine-hesitant participants were concerned regarding risk of SSc flare; however, few vaccinated participants reported this. These data may be useful for counselling people with SSc regarding COVID-19 vaccine safety and outcomes.
Collapse
Affiliation(s)
| | - Yin Wu
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | | | - Linda Kwakkenbos
- Department of Clinical Psychology, Behavioural Science Institute, Radboud University, Nijmegen, The Netherlands; Department of IQ Healthcare, Radboud University Medical Center, Nijmegen, the Netherlands; Department of Psychiatry, Centre for Mindfulness, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marie-Eve Carrier
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Richard S Henry
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Christopher P Denton
- University College London, Division of Medicine, Centre for Rheumatology and Connective Tissue Diseases, London, UK
| | - Luc Mouthon
- Service de Médecine Interne, Centre de Référence Maladies Autoimmunes et Autoinflammatoires Systémiques Rares d'Ile de France, de l'Est et de l'Ouest, Hôpital Cochin, Paris, France; Assistance Publique Hôpitaux de Paris-Centre, Hôpital Cochin, Université Paris Cité, Paris, France
| | | | - Brett D Thombs
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Department of Psychiatry, McGill University, Montreal, Quebec, Canada; Department of Medicine, McGill University, Montreal, Quebec, Canada; Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Quebec, Canada; Biomedical Ethics Unit, McGill University, Montreal, Quebec, Canada; Department of Psychology, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
17
|
Cahuapaza-Gutierrez NL, Campos-Escalante TS. Aplastic Anemia Following COVID-19 Vaccination: A Systematic Review of Case Reports and Case Series. Int J Hematol Oncol Stem Cell Res 2024; 18:297-305. [PMID: 39257704 PMCID: PMC11381666 DOI: 10.18502/ijhoscr.v18i3.16111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 12/23/2023] [Indexed: 09/12/2024] Open
Abstract
Background: Aplastic anemia (AA) is the prototypical bone marrow failure syndrome due to the destruction of hematopoietic stem cells by cytotoxic T cells. According to case reports, vaccines could lead to the development of AA. We conducted the present systematic review to evaluate cases of AA following vaccination against coronavirus disease (COVID-19). Materials and Methods: We searched the following databases: PubMed, Scopus, and EMBASE in English, Portuguese, and Spanish languages until April 24, 2023. Published reports and case series on AA following vaccination against COVID-19 were included. The Joanna Brigs Institute (JBI) was used to assess study quality and risk of bias. Results: Six studies were selected from 102 research studies, and data were extracted according to the inclusion criteria. All case reports and case series reported the occurrence of AA following COVID-19 vaccination. AA events were mainly observed in vaccines with messenger ribonucleic acid technology (Moderna; Pfizer-BioNTech). AA was diagnosed by bone marrow biopsy, and severity was determined by Camitta criteria. Conclusion: All cases of AA were properly diagnosed. The sample size was small; therefore, further investigations are required to demonstrate and elucidate the complete pathophysiological mechanisms of AA development after receiving COVID-19 vaccination.
Collapse
Affiliation(s)
- Nelson Luis Cahuapaza-Gutierrez
- Facultad de Ciencias de la Salud, Carrera de Medicina Humana, Universidad Científica Del Sur, Lima, Perú
- Change Research Working Group, Universidad Científica del Sur, Lima, Perú
| | | |
Collapse
|
18
|
Yang D, Tian J, Shen C, Li Q. An overview and single-arm meta-analysis of immune-mediated adverse events following COVID-19 vaccination. Front Pharmacol 2024; 15:1308768. [PMID: 38933672 PMCID: PMC11200080 DOI: 10.3389/fphar.2024.1308768] [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: 10/16/2023] [Accepted: 05/13/2024] [Indexed: 06/28/2024] Open
Abstract
Background We conducted an overview to assess immune adverse effects associated with the COVID-19 vaccine, guiding safer choices and providing evidence-based information to clinicians. Methods Forty-three studies on adverse effects of vaccines were reviewed from PubMed, Embase, and Web of Science. Single-arm meta-analyses estimated summary effects, incidence, presentation, etc. An overview using single-arm meta-analysis and reported the findings following the guidelines outlined in the 'Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) specifically focusing on myocarditis and thrombosis. After screening 2,591 articles, 42 studies met the inclusion criteria. Methodological quality was evaluated using AMSTAR 2. Disagreements were resolved via consensus. Data analysis utilized a random-effects model in R software to estimate incidence rates of selected adverse events. Results After removing 1,198 duplicates and screening out irrelevant articles from a total of 2,591, we included 42 studies. Adverse reactions to vaccinations include myocarditis, thrombosis, skin reactions, GBS, etc. thrombosis and myocarditis are the most dangerous diseases associated with vaccination. Myocarditis occurred in 6% of Vector vaccine recipients, compared to 61% of mRNA vaccine recipients. Thrombosis was more common after Vector vaccination (91%) than after mRNA vaccination (9%). Furthermore, eight studies conducted anti-PF4 antibody tests and yielded a positivity rate of 67%. Meta-analysis showed that among all patients with Vaccine-induced Thrombotic Thrombocytopenia, cerebral venous sinus thrombosis occurred in 66%, and intracranial hemorrhage occurred in 43%. The rates of deep vein thrombosis and pulmonary thromboembolism in vaccinated patients were 13% and 23%, respectively, with a pooled case fatality rate of 30%. Conclusion The results of this overview indicate the majority of adverse reactions are self-limiting and require minimal intervention, while rare occurrences such as myocarditis and thrombosis pose a potentially fatal threat.
Collapse
Affiliation(s)
- Donghua Yang
- Department of Public Health and Hospital Infection Management, Qinghai University Affiliated Hospital, Xining, China
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Caiyi Shen
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Qin Li
- Hunan University of Medicine, Huaihua, China
| |
Collapse
|
19
|
Panos LD, Bargiotas P, Hadjigeorgiou G, Panos GD. Neurovascular Adverse Effects of Sars-Cov-2 Vaccination. Drug Des Devel Ther 2024; 18:1891-1905. [PMID: 38836116 PMCID: PMC11147783 DOI: 10.2147/dddt.s464394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 05/22/2024] [Indexed: 06/06/2024] Open
Abstract
The global deployment of SARS-CoV-2 vaccines has been pivotal in curbing the COVID-19 pandemic, reducing morbidity and mortality associated with the virus. While most of these vaccines have demonstrated high efficacy and overall safety, emerging reports have highlighted potential neurovascular adverse effects, albeit uncommon, associated with these vaccinations. This review aims to assess and summarize the current knowledge on the neurovascular complications arising post-SARS-CoV-2 vaccination. We conducted an extensive literature review, focusing on clinical studies and case reports to identify reported neurovascular events, such as ischemic stroke, cerebral sinus venous thrombosis, intracerebral hemorrhage, pituitary apoplexy and primary CNS angiitis Despite the relative rarity of these events, their impact on affected individuals underscores the importance of ongoing surveillance, early detection, and management strategies. We aim to provide healthcare professionals with the latest evidence on neurovascular adverse effects, facilitating informed decision-making in the context of SARS-CoV-2 vaccination programs. Furthermore, we highlight areas requiring further research to understand the pathophysiology of these adverse events better and to develop targeted prevention and treatment strategies.
Collapse
Affiliation(s)
- Leonidas D Panos
- Department of Neurology, Bern University Hospital « Inselspital », Bern, Switzerland
- Department of Neurology, School of Medicine, University of Cyprus, Nicosia, Cyprus
| | - Panagiotis Bargiotas
- Department of Neurology, School of Medicine, University of Cyprus, Nicosia, Cyprus
| | | | - Georgios D Panos
- Department of Ophthalmology, Queen’s Medical Centre, Nottingham University Hospitals (NUH), Nottingham, U.K
- Division of Ophthalmology and Visual Sciences, School of Medicine, University of Nottingham, Nottingham, U.K
| |
Collapse
|
20
|
Liu Y, Lam DMK, Luan M, Zheng W, Ai H. Recent development of oral vaccines (Review). Exp Ther Med 2024; 27:223. [PMID: 38590568 PMCID: PMC11000446 DOI: 10.3892/etm.2024.12511] [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: 08/24/2023] [Accepted: 02/08/2024] [Indexed: 04/10/2024] Open
Abstract
Oral immunization can elicit an effective immune response and immune tolerance to specific antigens. When compared with the traditional injection route, delivering antigens via the gastrointestinal mucosa offers superior immune effects and compliance, as well as simplicity and convenience, making it a more optimal route for immunization. At present, various oral vaccine delivery systems exist. Certain modified bacteria, such as Salmonella, Escherichia coli and particularly Lactobacillus, are considered promising carriers for oral vaccines. These carriers can significantly enhance immunization efficiency by actively replicating in the intestinal tract following oral administration. The present review provided a discussion of the main mechanisms of oral immunity and the research progress made in the field of oral vaccines. Additionally, it introduced the advantages and disadvantages of the currently more commonly administered injectable COVID-19 vaccines, alongside the latest advancements in this area. Furthermore, recent developments in oral vaccines are summarized, and their potential benefits and side effects are discussed.
Collapse
Affiliation(s)
- Ying Liu
- Key Laboratory of Follicular Development and Reproductive Health in Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | | | - Mei Luan
- Department of Geriatric Medicine, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| | - Wenfu Zheng
- Chinese Academy of Sciences Key Lab for Biological Effects of Nanomaterials and Nanosafety, National Center for NanoScience and Technology, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Hao Ai
- Key Laboratory of Follicular Development and Reproductive Health in Liaoning Province, Jinzhou Medical University, Jinzhou, Liaoning 121000, P.R. China
| |
Collapse
|
21
|
Mendes D, Machira Krishnan S, O'Brien E, Padgett T, Harrison C, Strain WD, Manca A, Ustianowski A, Butfield R, Hamson E, Reynard C, Yang J. Modelling COVID-19 Vaccination in the UK: Impact of the Autumn 2022 and Spring 2023 Booster Campaigns. Infect Dis Ther 2024; 13:1127-1146. [PMID: 38662331 PMCID: PMC11098993 DOI: 10.1007/s40121-024-00965-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/21/2024] [Indexed: 04/26/2024] Open
Abstract
INTRODUCTION The delivery of COVID-19 vaccines was successful in reducing hospitalizations and mortality. However, emergence of the Omicron variant resulted in increased virus transmissibility. Consequently, booster vaccination programs were initiated to decrease the risk of severe disease and death among vulnerable members of the population. This study aimed to estimate the effects of the booster program and alternative vaccination strategies on morbidity and mortality due to COVID-19 in the UK. METHOD A Susceptible-Exposed-Infectious-Recovered (SEIR) model was used to assess the impact of several vaccination strategies on severe outcomes associated with COVID-19, including hospitalizations, mortality, National Health Service (NHS) capacity quantified by hospital general ward and intensive care unit (ICU) bed days, and patient productivity. The model accounted for age-, risk- and immunity-based stratification of the UK population. Outcomes were evaluated over a 48-week time horizon from September 2022 to August 2023 considering the actual UK autumn 2022/spring 2023 booster campaigns and six counterfactual strategies. RESULTS The model estimated that the autumn 2022/spring 2023 booster campaign resulted in a reduction of 18,921 hospitalizations and 1463 deaths, compared with a no booster scenario. Utilization of hospital bed days due to COVID-19 decreased after the autumn 2022/spring 2023 booster campaign. Expanding the booster eligibility criteria and improving uptake improved all outcomes, including averting twice as many ICU admissions, preventing more than 20% additional deaths, and a sevenfold reduction in long COVID, compared with the autumn 2022/spring 2023 booster campaign. The number of productive days lost was reduced by fivefold indicating that vaccinating a wider population has a beneficial impact on the morbidities associated with COVID-19. CONCLUSION Our modelling demonstrates that the autumn 2022/spring 2023 booster campaign reduced COVID-19-associated morbidity and mortality. Booster campaigns with alternative eligibility criteria warrant consideration in the UK, given their potential to further reduce morbidity and mortality as future variants emerge.
Collapse
Affiliation(s)
| | | | - Esmé O'Brien
- Health Economics and Outcomes Research Ltd, Cardiff, UK
| | | | - Cale Harrison
- Health Economics and Outcomes Research Ltd, Cardiff, UK
| | | | | | - Andrew Ustianowski
- Manchester University Foundation Trust, University of Manchester, Manchester, UK
| | | | | | | | - Jingyan Yang
- Pfizer Inc, New York, USA
- Institute for Social and Economic Research and Policy, Columbia University, New York, USA
| |
Collapse
|
22
|
Sanchez-Martinez ZV, Alpuche-Lazcano SP, Stuible M, Durocher Y. CHO cells for virus-like particle and subunit vaccine manufacturing. Vaccine 2024; 42:2530-2542. [PMID: 38503664 DOI: 10.1016/j.vaccine.2024.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/27/2024] [Accepted: 03/13/2024] [Indexed: 03/21/2024]
Abstract
Chinese Hamster Ovary (CHO) cells, employed primarily for manufacturing monoclonal antibodies and other recombinant protein (r-protein) therapeutics, are emerging as a promising host for vaccine antigen production. This is exemplified by the recently approved CHO cell-derived subunit vaccines (SUV) against respiratory syncytial virus (RSV) and varicella-zoster virus (VZV), as well as the enveloped virus-like particle (eVLP) vaccine against hepatitis B virus (HBV). Here, we summarize the design, production, and immunogenicity features of these vaccine and review the most recent progress of other CHO-derived vaccines in pre-clinical and clinical development. We also discuss the challenges associated with vaccine production in CHO cells, with a focus on ensuring viral clearance for eVLP products.
Collapse
Affiliation(s)
- Zalma V Sanchez-Martinez
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Sergio P Alpuche-Lazcano
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Matthew Stuible
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université du Québec à Montréal, 201 Avenue du Président Kennedy, Montréal, QC H2X 3Y7, Canada.
| |
Collapse
|
23
|
Ryoo S, Choi M, Choi NK, Shin HS, Woo JH, Park BJ, Oh S. Psychiatric adverse events associated with the COVID-19 vaccines approved in the Republic of Korea: a systematic review. Osong Public Health Res Perspect 2024; 15:107-114. [PMID: 38621764 PMCID: PMC11082440 DOI: 10.24171/j.phrp.2023.0325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 04/17/2024] Open
Abstract
This systematic review evaluated psychiatric adverse events (AEs) following vaccination against coronavirus disease 2019 (COVID-19). We included studies that reported or investigated psychiatric AEs in individuals who had received an approved COVID-19 vaccine in the Republic of Korea. Systematic electronic searches of Ovid-Medline, Embase, CENTRAL, and KoreaMed databases were conducted on March 22, 2023. Risk of bias was assessed using the Risk of Bias Assessment Tool for Non-randomized Studies 2.0. The study protocol was registered in the International Prospective Register of Systematic Reviews (CRD42023449422). Of the 301 articles initially selected, 7 were included in the final analysis. All studies reported on sleep disturbances, and 2 highlighted anxiety-related AEs. Sleep disorders like insomnia and narcolepsy were the most prevalent AEs, while depression was not reported. Our review suggests that these AEs may have been influenced by biological mechanisms as well as the broader psychosocial context of the COVID-19 pandemic. Although this study had limitations, such as a primary focus on the BNT162b2 vaccine and an observational study design, it offered a systematic, multi-vaccine analysis that fills a critical gap in the existing literature. This review underscores the need for continued surveillance of psychiatric AEs and guides future research to investigate underlying mechanisms, identify risk factors, and inform clinical management.
Collapse
Affiliation(s)
- Seungeun Ryoo
- Division of Healthcare Technology Assessment Research, National Evidence-based Healthcare Collaborating Agency, Seoul, Republic of Korea
| | - Miyoung Choi
- Division of Healthcare Technology Assessment Research, National Evidence-based Healthcare Collaborating Agency, Seoul, Republic of Korea
| | - Nam-Kyong Choi
- COVID-19 Vaccine Safety Research Center, Seoul, Republic of Korea
- Department of Health Convergence, College of Science & Industry Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Hyoung-Shik Shin
- COVID-19 Vaccine Safety Research Center, Seoul, Republic of Korea
- Department of Infectious Diseases, Daejeon Eulji Medical Center, Eulji University School of Medicine, Daejeon, Republic of Korea
| | - Jun Hee Woo
- COVID-19 Vaccine Safety Research Center, Seoul, Republic of Korea
- National Academy of Medicine of Korea, Seoul, Republic of Korea
| | - Byung-Joo Park
- COVID-19 Vaccine Safety Research Center, Seoul, Republic of Korea
- National Academy of Medicine of Korea, Seoul, Republic of Korea
| | - Sanghoon Oh
- COVID-19 Vaccine Safety Research Center, Seoul, Republic of Korea
- Department of Psychiatry, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu, Republic of Korea
| |
Collapse
|
24
|
Yıldız A, Răileanu C, Beissert T. Trans-Amplifying RNA: A Journey from Alphavirus Research to Future Vaccines. Viruses 2024; 16:503. [PMID: 38675846 PMCID: PMC11055088 DOI: 10.3390/v16040503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Replicating RNA, including self-amplifying RNA (saRNA) and trans-amplifying RNA (taRNA), holds great potential for advancing the next generation of RNA-based vaccines. Unlike in vitro transcribed mRNA found in most current RNA vaccines, saRNA or taRNA can be massively replicated within cells in the presence of RNA-amplifying enzymes known as replicases. We recently demonstrated that this property could enhance immune responses with minimal injected RNA amounts. In saRNA-based vaccines, replicase and antigens are encoded on the same mRNA molecule, resulting in very long RNA sequences, which poses significant challenges in production, delivery, and stability. In taRNA-based vaccines, these challenges can be overcome by splitting the replication system into two parts: one that encodes replicase and the other that encodes a short antigen-encoding RNA called transreplicon. Here, we review the identification and use of transreplicon RNA in alphavirus research, with a focus on the development of novel taRNA technology as a state-of-the art vaccine platform. Additionally, we discuss remaining challenges essential to the clinical application and highlight the potential benefits related to the unique properties of this future vaccine platform.
Collapse
Affiliation(s)
| | | | - Tim Beissert
- TRON—Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (A.Y.); (C.R.)
| |
Collapse
|
25
|
Ouaddouh C, Duijster JW, Lieber T, van Hunsel FPAM. The role of co-morbidities in the development of an AEFI after COVID-19 vaccination in a large prospective cohort with patient-reported outcomes in the Netherlands. Expert Opin Drug Saf 2024; 23:323-331. [PMID: 37796980 DOI: 10.1080/14740338.2023.2267971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/25/2023] [Indexed: 10/07/2023]
Abstract
BACKGROUND The effect of a preexisting comorbidity on the occurrence of adverse events after immunization (AEFIs) has been studied poorly. In this longitudinal cohort study, we assess the association between co-morbidities and the occurrence of AEFIs after COVID-19 vaccination. Also, we described the occurrence of flare-ups and their manifestation after COVID-19 vaccination in people with rheumatic diseases. RESEARCH DESIGN AND METHODS We performed multivariable logistic regression to investigate the association between the occurrence of AEFIs and 10 common comorbidities using patient-reported data from people vaccinated with the AstraZeneca, Johnson&Johnson, Moderna, or Pfizer vaccine. RESULTS Occurrence of any AEFI, injection site reactions, headache, fatigue, and/or malaise was significantly associated with presence of comorbidities, including psychological disorders, musculoskeletal disorders, and endocrine disorders after the first and second doses (OR ranges 1.23-1.77). One participant with rheumatoid arthritis experienced a flare-up after receiving the first dose of the AstraZeneca vaccine. DISCUSSION/CONCLUSION The results showed that the odds of reporting an AEFI after COVID-19 vaccination is significantly higher in the presence of some comorbidities whilst flare-ups are uncommon after receiving COVID-19 vaccination in people with rheumatic disease. In-depth research is needed to validate our results and unravel the observed associations from a mechanistic perspective.
Collapse
Affiliation(s)
- C Ouaddouh
- Vaccine Team, Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, Netherlands
| | - J W Duijster
- Vaccine Team, Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, Netherlands
| | - T Lieber
- Vaccine Team, Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, Netherlands
| | - F P A M van Hunsel
- Vaccine Team, Netherlands Pharmacovigilance Centre Lareb, 's-Hertogenbosch, Netherlands
- Faculty of Science and Engineering, FarmacoTherapie, Epidemiologie en Economie Groningen Research Institute of Pharmacy, Groningen, the Netherlands
| |
Collapse
|
26
|
Collins CP, Longo DL, Murphy WJ. The immunobiology of SARS-CoV-2 infection and vaccine responses: potential influences of cross-reactive memory responses and aging on efficacy and off-target effects. Front Immunol 2024; 15:1345499. [PMID: 38469293 PMCID: PMC10925677 DOI: 10.3389/fimmu.2024.1345499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 02/12/2024] [Indexed: 03/13/2024] Open
Abstract
Immune responses to both SARS-CoV-2 infection and its associated vaccines have been highly variable within the general population. The increasing evidence of long-lasting symptoms after resolution of infection, called post-acute sequelae of COVID-19 (PASC) or "Long COVID," suggests that immune-mediated mechanisms are at play. Closely related endemic common human coronaviruses (hCoV) can induce pre-existing and potentially cross-reactive immunity, which can then affect primary SARS-CoV-2 infection, as well as vaccination responses. The influence of pre-existing immunity from these hCoVs, as well as responses generated from original CoV2 strains or vaccines on the development of new high-affinity responses to CoV2 antigenic viral variants, needs to be better understood given the need for continuous vaccine adaptation and application in the population. Due in part to thymic involution, normal aging is associated with reduced naïve T cell compartments and impaired primary antigen responsiveness, resulting in a reliance on the pre-existing cross-reactive memory cell pool which may be of lower affinity, restricted in diversity, or of shorter duration. These effects can also be mediated by the presence of down-regulatory anti-idiotype responses which also increase in aging. Given the tremendous heterogeneity of clinical data, utilization of preclinical models offers the greatest ability to assess immune responses under a controlled setting. These models should now involve prior antigen/viral exposure combined with incorporation of modifying factors such as age on immune responses and effects. This will also allow for mechanistic dissection and understanding of the different immune pathways involved in both SARS-CoV-2 pathogen and potential vaccine responses over time and how pre-existing memory responses, including potential anti-idiotype responses, can affect efficacy as well as potential off-target effects in different tissues as well as modeling PASC.
Collapse
Affiliation(s)
- Craig P. Collins
- Graduate Program in Immunology, University of California (UC) Davis, Davis, CA, United States
| | - Dan L. Longo
- Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, United States
| | - William J. Murphy
- Departments of Dermatology and Internal Medicine (Hematology/Oncology), University of California (UC) Davis School of Medicine, Sacramento, CA, United States
| |
Collapse
|
27
|
Wu X, Xu K, Zhan P, Liu H, Zhang F, Song Y, Lv T. Comparative efficacy and safety of COVID-19 vaccines in phase III trials: a network meta-analysis. BMC Infect Dis 2024; 24:234. [PMID: 38383356 PMCID: PMC10880292 DOI: 10.1186/s12879-023-08754-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 10/25/2023] [Indexed: 02/23/2024] Open
Abstract
BACKGROUND Over a dozen vaccines are in or have completed phase III trials at an unprecedented speed since the World Health Organization (WHO) declared COVID-19 a pandemic. In this review, we aimed to compare and rank these vaccines indirectly in terms of efficacy and safety using a network meta-analysis. METHODS We searched Embase, MEDLINE, and the Cochrane Library for phase III randomized controlled trials (RCTs) from their inception to September 30, 2023. Two investigators independently selected articles, extracted data, and assessed the risk of bias. Outcomes included efficacy in preventing symptomatic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and the incidence of serious adverse events (SAEs) according to vaccine type and individual vaccines in adults and elderly individuals. The risk ratio and mean differences were calculated with 95% confidence intervals using a Bayesian network meta-analysis. RESULTS A total of 25 RCTs involving 22 vaccines were included in the study. None of vaccines had a higher incidence of SAEs than the placebo. Inactivated virus vaccines might be the safest, with a surface under the cumulative ranking curve (SUCRA) value of 0.16. BIV1-CovIran showed the highest safety index (SUCRA value: 0.13), followed by BBV152, Soberana, Gam-COVID-Vac, and ZF2001. There were no significant differences among the various types of vaccines regarding the efficacy in preventing symptomatic SARS-CoV-2 infection, although there was a trend toward higher efficacy of the mRNA vaccines (SUCRA value: 0.09). BNT162b2 showed the highest efficacy (SUCRA value: 0.02) among the individual vaccines, followed by mRNA-1273, Abdala, Gam-COVID-Vac, and NVX-CoV2373. BNT162b2 had the highest efficacy (SUCRA value: 0.08) in the elderly population, whereas CVnCoV, CoVLP + AS03, and CoronaVac were not significantly different from the placebo. CONCLUSIONS None of the different types of vaccines were significantly superior in terms of efficacy, while mRNA vaccines were significantly inferior in safety to other types. BNT162b2 had the highest efficacy in preventing symptomatic SARS-CoV-2 infection in adults and the elderly, whereas BIV1-CovIran had the lowest incidence of SAEs in adults.
Collapse
Affiliation(s)
- Xiaodi Wu
- Medical School of Nanjing University, Nanjing, 210000, China
| | - Ke Xu
- Medical School of Nanjing University, Nanjing, 210000, China
| | - Ping Zhan
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| | - Hongbing Liu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| | - Fang Zhang
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| | - Yong Song
- Medical School of Nanjing University, Nanjing, 210000, China.
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China.
| | - Tangfeng Lv
- Medical School of Nanjing University, Nanjing, 210000, China.
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China.
| |
Collapse
|
28
|
Handabile C, Ohno M, Sekiya T, Nomura N, Kawakita T, Kawahara M, Endo M, Nishimura T, Okumura M, Toba S, Sasaki M, Orba Y, Chua BY, Rowntree LC, Nguyen THO, Shingai M, Sato A, Sawa H, Ogasawara K, Kedzierska K, Kida H. Immunogenicity and protective efficacy of a co-formulated two-in-one inactivated whole virus particle COVID-19/influenza vaccine. Sci Rep 2024; 14:4204. [PMID: 38378856 PMCID: PMC10879490 DOI: 10.1038/s41598-024-54421-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 02/13/2024] [Indexed: 02/22/2024] Open
Abstract
Due to the synchronous circulation of seasonal influenza viruses and severe acute respiratory coronavirus 2 (SARS-CoV-2) which causes coronavirus disease 2019 (COVID-19), there is need for routine vaccination for both COVID-19 and influenza to reduce disease severity. Here, we prepared individual WPVs composed of formalin-inactivated SARS-CoV-2 WK 521 (Ancestral strain; Co WPV) or influenza virus [A/California/07/2009 (X-179A) (H1N1) pdm; Flu WPV] to produce a two-in-one Co/Flu WPV. Serum analysis from vaccinated mice revealed that a single dose of Co/Flu WPV induced antigen-specific neutralizing antibodies against both viruses, similar to those induced by either type of WPV alone. Following infection with either virus, mice vaccinated with Co/Flu WPV showed no weight loss, reduced pneumonia and viral titers in the lung, and lower gene expression of proinflammatory cytokines, as observed with individual WPV-vaccinated. Furthermore, a pentavalent vaccine (Co/qFlu WPV) comprising of Co WPV and quadrivalent influenza vaccine (qFlu WPV) was immunogenic and protected animals from severe COVID-19. These results suggest that a single dose of the two-in-one WPV provides efficient protection against SARS-CoV-2 and influenza virus infections with no evidence of vaccine interference in mice. We propose that concomitant vaccination with the two-in-one WPV can be useful for controlling both diseases.
Collapse
Affiliation(s)
- Chimuka Handabile
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Marumi Ohno
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Toshiki Sekiya
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Naoki Nomura
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Tomomi Kawakita
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Mamiko Kawahara
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | | | | | | | - Shinsuke Toba
- Shionogi Pharmaceutical Research Center, Shionogi & Company, Limited, Toyonaka, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Michihito Sasaki
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yasuko Orba
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Brendon Y Chua
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Louise C Rowntree
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Masashi Shingai
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Akihiko Sato
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Shionogi Pharmaceutical Research Center, Shionogi & Company, Limited, Toyonaka, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Hirofumi Sawa
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Kazumasa Ogasawara
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Katherine Kedzierska
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Hiroshi Kida
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan.
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan.
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan.
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
29
|
Moradi Hasan-Abad A, Arbabi M, Gilasi H, Motedayyen H. Immunogenicity and adverse events of the COVID-19 vaccines in healthy and individuals with autoimmune diseases in an Iranian population. Int J Immunopathol Pharmacol 2024; 38:3946320241239202. [PMID: 38494849 PMCID: PMC10946076 DOI: 10.1177/03946320241239202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024] Open
Abstract
Introduction: Recent studies have proposed various COVID-19 vaccines to control the disease and protect susceptible individuals. However, immunogenicity and safety of COVID-19 vaccines in various populations are not well identified yet. Therefore, this study aimed to elucidate the efficacy and safety of the BBIBP-CorV (Sinopharm) and ChAdOx1 nCoV-19 (Oxford-AstraZeneca) vaccines in healthy subjects and patients with autoimmune diseases.Methods: Study population included 121 healthy subjects and 100 patients with autoimmune diseases. Immunization was performed based on the national vaccination protocols. Of the 221 volunteers, 201 subjects received Sinopharm and 20 cases were vaccinated with Oxford-AstraZeneca. During a 1-year follow-up, the immunogenicity was measured by ELISA before primary vaccination and 1 to 3 months after secondary immunization. Side effects were studied before entering the study and 1 week after the second dose.Results: Vaccination had a positive impact on the induction of immunogenic response (p < .0001). The rates of seropositive vaccine responses were 80% and 75% in subjects vaccinated with the Sinopharm and Oxford-AstraZeneca, respectively. The neutralizing antibody values were significantly higher in subjects with autoimmune diseases than those without autoimmunity (p < .05). The rate of adverse events were 38% and 42% in subjects vaccinated with the Sinopharm and Oxford-AstraZeneca, respectively. The rates of immunogenic responses induced with the Sinopharm and Oxford-AstraZeneca were, respectively, 76% and 81.5% in seropositive subjects, while they were 63.8% and 79.1% in seronegative subjects vaccinated with the Sinopharm and Oxford-AstraZeneca, respectively. Individuals previously infected with SARS-CoV-2 showed a significant reduction in the value of SARS-CoV-2 neutralizing antibodies compared with seronegative subjects (p < .01-.05). Seropositive individuals vaccinated with the Sinopharm had significantly higher the percentages of vaccine-related adverse events than seronegative persons (p < .05). There was no significant difference between seronegative and seropositive individuals vaccinated with the Oxford-AstraZeneca.Conclusion: Our findings revealed that the Sinopharm and Oxford-AstraZeneca vaccines are effective in the production of neutralizing antibodies in healthy subjects and patients with autoimmune disorders undergoing immunosuppressive therapies without considerable reactogenicity.
Collapse
Affiliation(s)
- Amin Moradi Hasan-Abad
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohsen Arbabi
- Department of Medical Parasitology And Mycology, Kashan University Of Medical Sciences, Kashan, Iran
| | - Hamidreza Gilasi
- Department of Epidemiology & Biostatistics, Kashan University of Medical Sciences, Kashan, Iran
| | - Hossein Motedayyen
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
30
|
Kim KH. The Role of COVID-19 Vaccination for Patients With Atherosclerotic Cardiovascular Disease in the Upcoming Endemic Era. J Lipid Atheroscler 2024; 13:21-28. [PMID: 38299160 PMCID: PMC10825569 DOI: 10.12997/jla.2024.13.1.21] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/02/2024] Open
Abstract
COVID-19 vaccination has played a pivotal role in coping with the COVID-19 pandemic by providing a powerful tool to curb the spread of the virus, reduce severe illness and hospitalizations, and ultimately save lives and facilitate a return to normal daily routines. As COVID-19 vaccination has become more widespread and more individuals have recovered from the infection, COVID-19 has entered an endemic disease phase. This phase is characterized by a less severe and more stable pattern of infection within certain regions, similar to the predictability of seasonal influenza. In this endemic era, COVID-19 vaccines may appear to be less important, and many people are reluctant to receive COVID-19 vaccination for various reasons, including the fear of adverse events. However, COVID-19 remains a major public health problem, in that the incidence rate of new COVID-19 infections is still high and the morbidity and mortality in high-risk populations are substantial. Therefore, the role of COVID-19 vaccines in protecting high-risk individuals is crucial, and ongoing research and surveillance are imperative to refine vaccination recommendations in the ever-changing landscape of the COVID-19 endemic era. This review explores the role of COVID-19 vaccination in the upcoming COVID-19 endemic era.
Collapse
Affiliation(s)
- Kye Hun Kim
- Department of Cardiology, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, Korea
| |
Collapse
|
31
|
Cosenza LC, Marzaro G, Zurlo M, Gasparello J, Zuccato C, Finotti A, Gambari R. Inhibitory effects of SARS-CoV-2 spike protein and BNT162b2 vaccine on erythropoietin-induced globin gene expression in erythroid precursor cells from patients with β-thalassemia. Exp Hematol 2024; 129:104128. [PMID: 37939833 DOI: 10.1016/j.exphem.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 09/18/2023] [Accepted: 11/01/2023] [Indexed: 11/10/2023]
Abstract
During the recent coronavirus disease 2019 (COVID-19) pandemic several patients with β-thalassemia have been infected by severe acute respiratory syndrome coronavirus (SARS-CoV-2), and most patients were vaccinated against SARS-CoV-2. Recent studies demonstrate an impact of SARS-CoV-2 infection on the hematopoietic system. The main objective of this study was to verify the effects of exposure of erythroid precursor cells (ErPCs) from patients with β-thalassemia to SARS-CoV-2 spike protein (S-protein) and the BNT162b2 vaccine. Erythropoietin (EPO)-cultured ErPCs have been either untreated or treated with S-protein or BNT162b2 vaccine. The employed ErPCs were from a β-thalassemia cellular Biobank developed before the COVID-19 pandemic. The genotypes were β+-IVSI-110/β+-IVSI-110 (one patient), β039/β+-IVSI-110 (3 patients), and β039/ β039 (2 patients). After treatment with S-protein or BNT162b2 for 5 days, lysates were analyzed by high performance liquid chromatography (HPLC), for hemoglobin production, and isolated RNA was assayed by RT-qPCR, for detection of globin gene expression. The main conclusions of the results obtained are that SARS-CoV-2 S-protein and BNT162b2 vaccine (a) inhibit fetal hemoglobin (HbF) production by β-thalassemic ErPCs and (b) inhibit γ-globin mRNA accumulation. In addition, we have performed in silico studies suggesting a high affinity of S-protein to HbF. Remarkably, the binding interaction energy of fetal hemoglobin to S-protein was comparable with that of angiotensin-converting enzyme 2 (ACE2). Our results are consistent with the hypothesis of a relevant impact of SARS-CoV-2 infection and COVID-19 vaccination on the hematopoietic system.
Collapse
Affiliation(s)
- Lucia Carmela Cosenza
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Giovanni Marzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Matteo Zurlo
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Jessica Gasparello
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy
| | - Cristina Zuccato
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy; Center "Chiara Gemmo and Elio Zago" for the Research on Thalassemia, Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy; Center "Chiara Gemmo and Elio Zago" for the Research on Thalassemia, Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, Ferrara, Italy; Center "Chiara Gemmo and Elio Zago" for the Research on Thalassemia, Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
32
|
Alshehri S, Sallam M. Vaccine conspiracy association with higher COVID-19 vaccination side effects and negative attitude towards booster COVID-19, influenza and monkeypox vaccines: A pilot study in Saudi Universities. Hum Vaccin Immunother 2023; 19:2275962. [PMID: 37941437 PMCID: PMC10653693 DOI: 10.1080/21645515.2023.2275962] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/06/2023] [Accepted: 10/24/2023] [Indexed: 11/10/2023] Open
Abstract
Conspiracies regarding vaccines are widely prevalent, with negative consequences on health-seeking behaviors. The current study aimed to investigate the possible association between the embrace of vaccine conspiracies and the attitude to booster COVID-19, seasonal influenza, and monkeypox (mpox) vaccinations as well as the perceived side effects following COVID-19 vaccination. The target population involved academic staff and university students in health colleges in the Kingdom of Saudi Arabia. A self-administered questionnaire was distributed in January 2023 to collect data on participants' demographics, self-reported side effects following each dose, willingness to get booster COVID-19, seasonal influenza, and mpox vaccinations, as well as an evaluation of vaccine conspiracies and attitude to mandatory vaccination. Among the 273 participants, the willingness to receive yearly booster COVID-19 vaccination was observed among 26.0% of the participants, while it was 46.9% and 34.1% for seasonal influenza and mpox vaccinations, respectively. Multinomial logistic regression analyses demonstrated a significant correlation between endorsing vaccine conspiracies and higher frequency of self-reported side effects following uptake of the second and third doses of COVID-19 vaccines. Vaccine conspiracies were also correlated with attitude toward booster COVID-19, influenza, mpox, and mandatory vaccination. The findings of this pilot study highlighted the potential adverse impact of the preexisting notions and negative attitudes toward vaccines, which could have contributed to heightened perceived side effects following COVID-19 vaccination. The study also highlighted the ongoing divisions concerning mandatory vaccination policies, emphasizing the need for cautious implementation of this strategy as a last resort for public health benefit.
Collapse
Affiliation(s)
- Samiyah Alshehri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Malik Sallam
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
- Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| |
Collapse
|
33
|
Zurlo M, Gasparello J, Verona M, Papi C, Cosenza LC, Finotti A, Marzaro G, Gambari R. The anti-SARS-CoV-2 BNT162b2 vaccine suppresses mithramycin-induced erythroid differentiation and expression of embryo-fetal globin genes in human erythroleukemia K562 cells. Exp Cell Res 2023; 433:113853. [PMID: 37944576 DOI: 10.1016/j.yexcr.2023.113853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 11/04/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023]
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is causative of the ongoing coronavirus disease 2019 (COVID-19) pandemic. The SARS-CoV-2 Spike protein (S-protein) plays an important role in the early phase of SARS-CoV-2 infection through efficient interaction with ACE2. The S-protein is produced by RNA-based COVID-19 vaccines, that were fundamental for the reduction of the viral spread within the population and the clinical severity of COVID-19. However, the S-protein has been hypothesized to be responsible for damaging cells of several tissues and for some important side effects of RNA-based COVID-19 vaccines. Considering the impact of COVID-19 and SARS-CoV-2 infection on the hematopoietic system, the aim of this study was to verify the effect of the BNT162b2 vaccine on erythroid differentiation of the human K562 cell line, that has been in the past intensively studied as a model system mimicking some steps of erythropoiesis. In this context, we focused on hemoglobin production and induced expression of embryo-fetal globin genes, that are among the most important features of K562 erythroid differentiation. We found that the BNT162b2 vaccine suppresses mithramycin-induced erythroid differentiation of K562 cells. Reverse-transcription-qPCR and Western blotting assays demonstrated that suppression of erythroid differentiation was associated with sharp inhibition of the expression of α-globin and γ-globin mRNA accumulation. Inhibition of accumulation of ζ-globin and ε-globin mRNAs was also observed. In addition, we provide in silico studies suggesting a direct interaction between SARS-CoV-2 Spike protein and Hb Portland, that is the major hemoglobin produced by K562 cells. This study thus provides information suggesting the need of great attention on possible alteration of hematopoietic parameters following SARS-CoV-2 infection and/or COVID-19 vaccination.
Collapse
Affiliation(s)
- Matteo Zurlo
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy.
| | - Jessica Gasparello
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy
| | - Marco Verona
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Chiara Papi
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy
| | - Lucia Carmela Cosenza
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy; Center 'Chiara Gemmo and Elio Zago' for the Research on Thalassemia, University of Ferrara, 44121 Ferrara, Italy
| | - Giovanni Marzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, Section of Biochemistry and Molecular Biology, University of Ferrara, 44121 Ferrara, Italy; Center 'Chiara Gemmo and Elio Zago' for the Research on Thalassemia, University of Ferrara, 44121 Ferrara, Italy.
| |
Collapse
|
34
|
Sallam M, Abbasi H, Obeidat RJ, Badayneh R, Alkhashman F, Obeidat A, Oudeh D, Uqba Z, Mahafzah A. Unraveling the association between vaccine attitude, vaccine conspiracies and self-reported side effects following COVID-19 vaccination among nurses and physicians in Jordan. Vaccine X 2023; 15:100405. [PMID: 38161986 PMCID: PMC10755110 DOI: 10.1016/j.jvacx.2023.100405] [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: 08/08/2023] [Revised: 10/25/2023] [Accepted: 11/01/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND The negative impact of vaccine conspiracies is linked with negative health behavior. The aim of the current study was to examine the association between attitudes toward booster COVID-19, influenza, and monkeypox (mpox) vaccinations with post-COVID-19 vaccine side effects, vaccine conspiracies, and attitude towards mandatory vaccination among nurses and physicians in Jordan. METHODS A structured closed-ended questionnaire was used to collect data on demographics, COVID-19 history, COVID-19 vaccine type and doses received, self-reported side effects post-COVID-19 vaccination, acceptance of booster COVID-19, seasonal influenza, and mpox vaccinations, attitudes towards mandatory vaccination, and beliefs in vaccine conspiracies. RESULTS The study sample comprised a total of 341 participants. Acceptance of yearly booster COVID-19 vaccination was expressed by 46.6% of the sample, while 73.3% accepted seasonal influenza vaccination, and only 37.0% accepted mpox vaccination. A higher frequency of self-reported side effects following the first COVID-19 vaccine dose was associated with embrace of vaccine conspiracies and vaccine type. For the second vaccine dose, a higher frequency of self-reported side effects was associated with the embrace of vaccine conspiracies, older age, and affiliation to private sector. In multinomial logistic regression analyses, the lower embrace of vaccine conspiracies was associated with lower odds of reporting side effects post-COVID-19 vaccination. The lower embrace of vaccine conspiracies and favorable attitude towards mandatory vaccination were associated with the willingness to get COVID-19, influenza, and mpox vaccinations. CONCLUSION The study findings highlighted the negative impact of embracing vaccine conspiracies on health-seeking behavior among nurses and physicians. The findings indicated that the willingness to get vaccinated was associated with lower endorsement of vaccine conspiracies. Additionally, the lower embrace of vaccine conspiracies was associated with a lower frequency of self-reported side effects following COVID-19 vaccination. These results emphasize the importance of addressing vaccine misinformation and promoting accurate information to ensure optimal vaccine uptake and public health outcomes.
Collapse
Affiliation(s)
- Malik Sallam
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| | - Hiba Abbasi
- Department of Internal Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Internal Medicine, Jordan University Hospital, Amman, Jordan
| | - Rawan J. Obeidat
- The Office of Infection Prevention and Control, Jordan University Hospital, Amman, Jordan
| | - Reham Badayneh
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Farah Alkhashman
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Aseel Obeidat
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Dana Oudeh
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Zena Uqba
- School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Azmi Mahafzah
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
| |
Collapse
|
35
|
Janekrongtham C, Salazar M, Doung-ngern P. Sex Differences in Serious Adverse Events Reported Following Booster Doses of COVID-19 Vaccination in Thailand: A Countrywide Nested Unmatched Case-Control Study. Vaccines (Basel) 2023; 11:1772. [PMID: 38140176 PMCID: PMC10747632 DOI: 10.3390/vaccines11121772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/17/2023] [Accepted: 10/25/2023] [Indexed: 12/24/2023] Open
Abstract
A booster dose of a COVID-19 vaccine has been proven effective in restoring vaccine effectiveness and is currently recommended for use in some populations at risk of severe COVID-19 infection. Since sex differences in adverse events are significant in response to the vaccines, the safety of booster selection must be studied to avoid serious adverse events (SAE), such as life-threatening diseases. First, this study aimed to identify sex differences in SAE incidences using a prospective cohort design. Second, a nested unmatched case-control study was used to identify factors associated with reported SAE within 30 days after the booster shot. Multivariable logistic regression indicated the adjusted odds ratio by accounting for host and vaccine variables, thus, policy effects. The findings confirmed that SAE was rare and that age-sex-dominated disease classifications differed. Specific to SAE following the booster dose, we found that females aged 12-40 had a higher risk of being reported with SAE than males of the same age, while males over 50 had a higher risk than females. Other risk factors identified were the presence of metabolic syndrome and the use of certain vaccine brands. Mechanisms could be explained by individual host responses rather than the vaccines' direct effect. Therefore, SAE could be preventable by age-sex-specific vaccine selection, post-vaccination precautions, and early symptom detection. Future vaccine development should aim to limit host-specific reactogenicity for safety concerns.
Collapse
Affiliation(s)
- Chawisar Janekrongtham
- Division of AIDS and STIs, Department of Disease Control, Ministry of Public Health, 88/21 Tiwanon Rd., Nonthaburi 11000, Thailand
| | - Mariano Salazar
- Department of Global Public Health, Karolinska Institutet, Widerströmska huset Tomtebodavägen 18 A, Plan 3, 17165 Stockholm, Sweden;
| | - Pawinee Doung-ngern
- Division of Epidemiology, Department of Disease Control, Ministry of Public Health, 88/21 Tiwanon Rd., Nonthaburi 11000, Thailand;
| |
Collapse
|
36
|
González-Celestino A, González-Osorio Y, García-Iglesias C, Echavarría-Iñiguez A, Sierra-Mencía A, Recio-García A, Trigo-López J, Planchuelo-Gómez A, Hurtado ML, Sierra-Martínez L, Ruiz M, Rojas-Hernández M, Pérez-Almendro C, Paniagua M, Núñez G, Mora M, Montilla C, Martínez-Badillo C, Lozano AG, Gil A, Cubero M, Cornejo A, Calcerrada I, Blanco M, Alberdí-Iglesias A, Fernández-de-Las-Peñas C, Guerrero-Peral AL, García-Azorín D. Differences and similarities between COVID-19 related-headache and COVID-19 vaccine related-headache. A case-control study. Rev Neurol 2023; 77:229-239. [PMID: 37962534 PMCID: PMC10831766 DOI: 10.33588/rn.7710.2023063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Indexed: 11/15/2023]
Abstract
INTRODUCTION Headache is a frequent symptom at the acute phase of coronavirus disease 2019 (COVID-19) and also one of the most frequent adverse effects following vaccination. In both cases, headache pathophysiology seems linked to the host immune response and could have similarities. We aimed to compare the clinical phenotype and the frequency and associated onset symptoms in patients with COVID-19 related-headache and COVID-19 vaccine related-headache. SUBJECTS AND METHODS A case-control study was conducted. Patients with confirmed COVID-19 infection and COVID-19-vaccine recipients who experienced new-onset headache were included. A standardised questionnaire was administered, including demographic variables, prior history of headaches, associated symptoms and headache-related variables. Both groups were matched for age, sex, and prior history of headache. A multivariate regression analysis was performed. RESULTS A total of 238 patients fulfilled eligibility criteria (143 patients with COVID-19 related-headache and 95 subjects experiencing COVID-19 vaccine related-headache). Patients with COVID-19 related-headache exhibited a higher frequency of arthralgia, diarrhoea, dyspnoea, chest pain, expectoration, anosmia, myalgia, odynophagia, rhinorrhoea, cough, and dysgeusia. Further, patients with COVID-19 related-headache had a more prolonged daily duration of headache and described the headache as the worst headache ever experienced. Patients with COVID-19 vaccine-related headache, experienced more frequently pain in the parietal region, phonophobia, and worsening of the headache by head movements or eye movements. CONCLUSION Headache caused by SARS-CoV-2 infection and COVID-19 vaccination related-headache have more similarities than differences, supporting a shared pathophysiology, and the activation of the innate immune response. The main differences were related to associated symptoms.
Collapse
Affiliation(s)
| | | | - C García-Iglesias
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | | | - A Sierra-Mencía
- Hospital Clínico Universitario de Valladolid, Valladolid, España
| | - A Recio-García
- Hospital Clínico Universitario de Valladolid, Valladolid, España
| | - J Trigo-López
- Hospital Clínico Universitario de Valladolid, Valladolid, España
| | | | - M L Hurtado
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - L Sierra-Martínez
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - M Ruiz
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - M Rojas-Hernández
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - C Pérez-Almendro
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - M Paniagua
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - G Núñez
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - M Mora
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - C Montilla
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - C Martínez-Badillo
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - A G Lozano
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - A Gil
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - M Cubero
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - A Cornejo
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - I Calcerrada
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - M Blanco
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | - A Alberdí-Iglesias
- Área Básica de Salud de Atención Primaria Valladolid Este, Valladolid, España
| | | | | | - D García-Azorín
- Universidad de Valladolid, Valladolid, España
- Hospital Clínico Universitario de Valladolid, Valladolid, España
| |
Collapse
|
37
|
Krumholz HM, Wu Y, Sawano M, Shah R, Zhou T, Arun AS, Khosla P, Kaleem S, Vashist A, Bhattacharjee B, Ding Q, Lu Y, Caraballo C, Warner F, Huang C, Herrin J, Putrino D, Hertz D, Dressen B, Iwasaki A. Post-Vaccination Syndrome: A Descriptive Analysis of Reported Symptoms and Patient Experiences After Covid-19 Immunization. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.09.23298266. [PMID: 37986769 PMCID: PMC10659483 DOI: 10.1101/2023.11.09.23298266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Introduction A chronic post-vaccination syndrome (PVS) after covid-19 vaccination has been reported but has yet to be well characterized. Methods We included 241 individuals aged 18 and older who self-reported PVS after covid-19 vaccination and who joined the online Yale Listen to Immune, Symptom and Treatment Experiences Now (LISTEN) Study from May 2022 to July 2023. We summarized their demographics, health status, symptoms, treatments tried, and overall experience. Results The median age of participants was 46 years (interquartile range [IQR]: 38 to 56), with 192 (80%) identifying as female, 209 (87%) as non-Hispanic White, and 211 (88%) from the United States. Among these participants with PVS, 127 (55%) had received the BNT162b2 [Pfizer-BioNTech] vaccine, and 86 (37%) received the mRNA-1273 [Moderna] vaccine. The median time from the day of index vaccination to symptom onset was three days (IQR: 1 day to 8 days). The time from vaccination to symptom survey completion was 595 days (IQR: 417 to 661 days). The median Euro-QoL visual analogue scale score was 50 (IQR: 39 to 70). The five most common symptoms were exercise intolerance (71%), excessive fatigue (69%), numbness (63%), brain fog (63%), and neuropathy (63%). In the week before survey completion, participants reported feeling unease (93%), fearfulness (82%), and overwhelmed by worries (81%), as well as feelings of helplessness (80%), anxiety (76%), depression (76%), hopelessness (72%), and worthlessness (49%) at least once. Participants reported a median of 20 (IQR: 13 to 30) interventions to treat their condition. Conclusions In this study, individuals who reported PVS after covid-19 vaccination had low health status, high symptom burden, and high psychosocial stress despite trying many treatments. There is a need for continued investigation to understand and treat this condition.
Collapse
Affiliation(s)
- Harlan M. Krumholz
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
- Center for Infection and Immunity, Yale School of Medicine, New Haven, Connecticut
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
- Department of Health Policy and Management, Yale School of Public Health, New Haven, Connecticut
| | - Yilun Wu
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
- Department of Biostatistics, Yale School of Public Health, New Haven, Connecticut
| | - Mitsuaki Sawano
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Rishi Shah
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
- Department of Applied Mathematics, Yale College, New Haven, Connecticut
| | - Tianna Zhou
- Yale School of Medicine, New Haven, Connecticut
| | | | | | - Shayaan Kaleem
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Anushree Vashist
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
- The College at the University of Chicago, Chicago, Illinois
| | - Bornali Bhattacharjee
- Center for Infection and Immunity, Yale School of Medicine, New Haven, Connecticut
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut
| | - Qinglan Ding
- College of Health and Human Sciences, Purdue University, West Lafayette, Indiana
| | - Yuan Lu
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - César Caraballo
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
- Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Frederick Warner
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Chenxi Huang
- Center for Outcomes Research and Evaluation, Yale New Haven Hospital, New Haven, Connecticut
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - Jeph Herrin
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| | - David Putrino
- Rehabilitation and Human Performance, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | - Akiko Iwasaki
- Center for Infection and Immunity, Yale School of Medicine, New Haven, Connecticut
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut
- Howard Hughes Medical Institute, Chevy Chase, Maryland
| |
Collapse
|
38
|
Abdulkader MA, Merza MA. Immediate and Long-Term Adverse Events of COVID-19 Vaccines: A One-Year Follow-Up Study From the Kurdistan Region of Iraq. Cureus 2023; 15:e47670. [PMID: 38021955 PMCID: PMC10671599 DOI: 10.7759/cureus.47670] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
Background The administration of COVID-19 vaccines has been critical in controlling the spread of the virus. However, understanding the potential adverse events (AEs) associated with these vaccines is crucial for public health. While most previous studies observed only short-term AEs, this study aimed to investigate the immediate and long-term AEs following the first and second doses of Pfizer, AstraZeneca, and Sinopharm vaccines, providing valuable long-term insights. Methodology A prospective, one-year, follow-up study was conducted by tracking 922 vaccinated individuals to assess short-term and long-term AEs. Demographics, clinical characteristics, vaccine types, and dose effects were taken into consideration. AEs were classified based on severity and duration. Statistical analyses were performed to compare differences among the vaccine groups, with p-values <0.05 considered significant. Bowker's and chi-square tests were performed using JMP Pro 14.3.0. Results Of the 922 participants, 55.53% (n = 512) were vaccinated with Pfizer, and 23.32% (n = 215) and 21.15% (n = 195) were vaccinated with Sinopharm and AstraZeneca, respectively. Overall, 72.34% of participants (n = 667) were suffering from AEs after the first dose, with a lower prevalence of AEs after the second dose (52.71%, n = 486). Pfizer exhibited the highest percentage and severity of AEs, followed by AstraZeneca and Sinopharm. Most AEs reported in this study were mild and resolved within 72 hours, with females experiencing more frequent AEs. The common short-term AEs observed were fever, injection-site pain, myalgia, fatigue, and headache. Notably, there were no chronic AEs, and only one case of myocarditis was associated with AstraZeneca. Conclusions Despite the variation in the prevalence of AEs among the three vaccines, the vaccination process proved to be safe with no serious short-term AEs. However, the long-term AEs associated with AstraZeneca and the decrease in the prevalence of AEs after the second dose of the COVID-19 vaccines warrant further investigations and priority for future research.
Collapse
Affiliation(s)
- Manhal A Abdulkader
- Department of Clinical Pharmacy, College of Pharmacy, University of Duhok, Duhok, IRQ
| | - Muayad A Merza
- Department of Internal Medicine, University of Duhok, Duhok, IRQ
| |
Collapse
|
39
|
Zhao T, Yang Z, Wu Y, Yang J. Immunogenicity and safety of COVID-19 vaccines among people living with HIV: A systematic review and meta-analysis. Epidemiol Infect 2023; 151:e176. [PMID: 37704371 PMCID: PMC10600909 DOI: 10.1017/s095026882300153x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 08/20/2023] [Accepted: 09/07/2023] [Indexed: 09/15/2023] Open
Abstract
Available data suggest that the immunogenicity of COVID-19 vaccines might decrease in the immunocompromised population, but data on vaccine immunogenicity and safety among people living with HIV (PLWH) are still lacking. The purpose of this meta-analysis is to compare the immunogenicity and safety of COVID-19 vaccines in PLWH with healthy controls. We comprehensively searched the following databases: PubMed, Cochrane Library, and EMBASE. The risk ratio (RR) of seroconversion after the first and second doses of a COVID-19 vaccine was separately pooled using random-effects meta-analysis. Seroconversion rate was lower among PLWH compared with healthy individuals after the first (RR = 0.77, 95% confident interval (CI) 0.64-0.92) and second doses (RR = 0.97, 95%CI 0.95-0.99). The risk of total adverse reactions among PLWH is similar to the risk in the healthy group, after the first (RR = 0.87, 95%CI 0.70-1.10) and second (RR = 0.83, 95%CI 0.65-1.07) doses. This study demonstrates that the immunogenicity and safety of SARS-CoV-2 vaccine in fully vaccinated HIV-infected patients were generally satisfactory. A second dose was related to seroconversion enhancement. Therefore, we considered that a booster dose may provide better seroprotection for PLWH. On the basis of a conventional two-dose regimen for COVID-19 vaccines, the booster dose is very necessary.
Collapse
Affiliation(s)
- Tianyu Zhao
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, China
| | - Zongxing Yang
- The Second Department of Infectious Disease, Xixi Hospital of Hangzhou, The Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuxia Wu
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, China
| | - Jin Yang
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou, China
- Department of Translational Medicine Center, Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
40
|
Niu D, Wu Y, Lian J. Circular RNA vaccine in disease prevention and treatment. Signal Transduct Target Ther 2023; 8:341. [PMID: 37691066 PMCID: PMC10493228 DOI: 10.1038/s41392-023-01561-x] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 06/02/2023] [Accepted: 07/09/2023] [Indexed: 09/12/2023] Open
Abstract
CircRNAs are a class of single-stranded RNAs with covalently linked head-to-tail topology. In the decades since its initial discovery, their biogenesis, regulation, and function have rapidly disclosed, permitting a better understanding and adoption of them as new tools for medical applications. With the development of biotechnology and molecular medicine, artificial circRNAs have been engineered as a novel class of vaccines for disease treatment and prevention. Unlike the linear mRNA vaccine which applications were limited by its instability, inefficiency, and innate immunogenicity, circRNA vaccine which incorporate internal ribosome entry sites (IRESs) and open reading frame (ORF) provides an improved approach to RNA-based vaccination with safety, stability, simplicity of manufacture, and scalability. However, circRNA vaccines are at an early stage, and their optimization, delivery and applications require further development and evaluation. In this review, we comprehensively describe circRNA vaccine, including their history and superiority. We also summarize and discuss the current methodological research for circRNA vaccine preparation, including their design, synthesis, and purification. Finally, we highlight the delivery options of circRNA vaccine and its potential applications in diseases treatment and prevention. Considering their unique high stability, low immunogenicity, protein/peptide-coding capacity and special closed-loop construction, circRNA vaccine, and circRNA-based therapeutic platforms may have superior application prospects in a broad range of diseases.
Collapse
Affiliation(s)
- Dun Niu
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 400038, Chongqing, China
- Department of Clinical Biochemistry, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Yaran Wu
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 400038, Chongqing, China
- Department of Clinical Biochemistry, Army Medical University (Third Military Medical University), 400038, Chongqing, China
| | - Jiqin Lian
- Department of Clinical Laboratory Medicine, Southwest Hospital, Army Medical University (Third Military Medical University), 400038, Chongqing, China.
- Department of Clinical Biochemistry, Army Medical University (Third Military Medical University), 400038, Chongqing, China.
| |
Collapse
|
41
|
Fan X, Zhao Y, Wu F, Yuan Y, Lang B, Yin D, Xu Z, Jiang S, Zou H, Yuan J, Sun C. Safety and Immunogenicity of SARS-CoV-2 Vaccines in People Living With HIV (PLWH): A Systematic Review and Meta-analysis of Real-World Studies. INFECTIOUS MICROBES AND DISEASES 2023. [DOI: 10.1097/im9.0000000000000129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2025]
Abstract
Abstract
The safety and immunogenicity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines in people living with HIV (PLWH) in real world studies remain controversial. Thus, we conducted a comprehensive systematic review and meta-analysis to address this issue. Data search were conducted from PubMed, Web of science, and Embase. Adverse events following vaccination, the risk ratio (RR) of SARS-CoV-2-specific IgG seroconversion, and the level of anti-SARS-CoV-2 neutralizing antibodies were compared between the PLWH group and a healthy control group. A total of 10,582 PLWH from 22 studies were included. In our analysis, the incidence of local or systemic adverse events after the first SARS-CoV-2 vaccine dose was not statistically different between PLWH and healthy controls. However, there was a statistical difference after the second dose (RR = 0.83, 95% CI: 0.71 – 0.98). The seroconversion rate of SARS-CoV-2 IgG antibodies in PLWH was significantly lower than that in the healthy control group (RR = 0.94, 95% CI: 0.89-0.98; I2
= 80%, P < 0.01). The anti-SARS-CoV-2 neutralizing antibody titers in PLWH after full immunization were also significantly lower than that in the healthy control group (RR = 0.91, 95% CI: 0.85-0.98; I2
= 81%, P < 0.01). The safety and tolerance of COVID-19 vaccines in PLWH are acceptable. However, their immunogenicity may be impaired to a certain extent, characterized by a lower IgG seroconversion rate and neutralizing antibody titers compared with healthy individuals. These findings should provide guidance for optimizing future COVID-19 vaccination strategies among PLWH.
Collapse
Affiliation(s)
- Xueying Fan
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Yangguo Zhao
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Fan Wu
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Yue Yuan
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Bing Lang
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Di Yin
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Zhongliang Xu
- Nanshan District Center for Disease Control and Prevention, Shenzhen, China
| | - Shiqiang Jiang
- Nanshan District Center for Disease Control and Prevention, Shenzhen, China
| | | | - Jianhui Yuan
- Nanshan District Center for Disease Control and Prevention, Shenzhen, China
| | | |
Collapse
|
42
|
Zasada AA, Darlińska A, Wiatrzyk A, Woźnica K, Formińska K, Czajka U, Główka M, Lis K, Górska P. COVID-19 Vaccines over Three Years after the Outbreak of the COVID-19 Epidemic. Viruses 2023; 15:1786. [PMID: 37766194 PMCID: PMC10536649 DOI: 10.3390/v15091786] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 09/29/2023] Open
Abstract
The outbreak of COVID-19 started in December 2019 and spread rapidly all over the world. It became clear that the development of an effective vaccine was the only way to stop the pandemic. It was the first time in the history of infectious diseases that the process of the development of a new vaccine was conducted on such a large scale and accelerated so rapidly. At the end of 2020, the first COVID-19 vaccines were approved for marketing. At the end of March 2023, over three years after the outbreak of the COVID-19 pandemic, 199 vaccines were in pre-clinical development and 183 in clinical development. The candidate vaccines in the clinical phase are based on the following platforms: protein subunit, DNA, RNA, non-replication viral vector, replicating viral vector, inactivated virus, virus-like particles, live attenuated virus, replicating viral vector combined with an antigen-presenting cell, non-replication viral vector combined with an antigen-presenting cell, and bacterial antigen-spore expression vector. Some of the new vaccine platforms have been approved for the first time for human application. This review presents COVID-19 vaccines currently available in the world, procedures for assurance of the quality and safety of the vaccines, the vaccinated population, as well as future perspectives for the new vaccine platforms in drug and therapy development for infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Aleksandra Anna Zasada
- Department of Sera and Vaccines Evaluation, National Institute of Public Health NIH—National Research Institute, 00-791 Warsaw, Poland; (A.D.); (A.W.); (K.W.); (K.F.); (U.C.); (M.G.); (K.L.); (P.G.)
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kim JW, Jung JY, Suh CH, Ye YM, Kim HA. Effects of COVID-19 and Influenza Vaccination on Rheumatic Diseases: Results From a Survey of Patient-Reported Outcomes After Vaccination. J Korean Med Sci 2023; 38:e247. [PMID: 37582497 PMCID: PMC10427213 DOI: 10.3346/jkms.2023.38.e247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 04/05/2023] [Indexed: 08/17/2023] Open
Abstract
BACKGROUND This study aimed to compare the occurrence of adverse events (AEs) and disease flares after vaccination against coronavirus disease 2019 (COVID-19) and influenza in patients with autoimmune rheumatic diseases (ARDs). METHODS Between November 2021 and March 2022, a survey was conducted among patients with ARD who received COVID-19 and influenza vaccinations. The questionnaire included 11 mandatory and closed-ended questions, and the following items were collected: medical history, immunization history, type of vaccine, patient-reported AEs, flare-up of the underlying disease after vaccination, and a confirmed diagnosis of COVID-19 or influenza. We compared the occurrence of vaccine-related adverse reactions to the COVID-19 and influenza vaccines based on the survey results. Multivariate logistic regression analysis was used to identify the factors affecting AEs or disease flares and to compare the post-vaccine response to mixed and matched vaccines. RESULTS We analyzed 601 adults with ARD who received the COVID-19 vaccine, with a mean age of 49.6 years (80.5% female). A total of 255 participants (42.4%) received a complete course of primary vaccination, 342 (56.9%) completed the booster dose, and 132 (38.6%) received a mixed vaccine. The frequencies of AEs (188 [52.2%] vs. 21 [5.8%]; P < 0.001) and disease flares (58 [16.2%] vs. 5 [1.4%]; P < 0.001) after COVID-19 vaccination were significantly higher than those after influenza vaccination. In the risk factor analysis, previous allergic reaction to other vaccines (odds ratio, 1.95; confidence interval, 1.07-3.70; P = 0.034) was the only factor associated with the occurrence of AEs. There was no difference in the post-vaccine responses between the mixed and matched vaccines. CONCLUSION The results of the survey of patients with ARD revealed that patient-reported AEs and underlying disease flares after receiving the COVID-19 vaccine were significantly higher than those after the influenza vaccine.
Collapse
Affiliation(s)
- Ji-Won Kim
- Department of Rheumatology, Ajou University School of Medicine, Suwon, Korea
| | - Ju-Yang Jung
- Department of Rheumatology, Ajou University School of Medicine, Suwon, Korea
| | - Chang-Hee Suh
- Department of Rheumatology, Ajou University School of Medicine, Suwon, Korea
| | - Young-Min Ye
- Department of Allergy & Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Hyoun-Ah Kim
- Department of Rheumatology, Ajou University School of Medicine, Suwon, Korea.
| |
Collapse
|
44
|
Alsudais AS, Alkanani RS, Fathi AB, Almuntashiri SS, Jamjoom JN, Alzhrani MA, Althubaiti A, Radi S. Autoimmune diabetes mellitus after COVID-19 vaccination in adult population: a systematic review of case reports. BMC Endocr Disord 2023; 23:164. [PMID: 37542216 PMCID: PMC10403898 DOI: 10.1186/s12902-023-01424-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 07/24/2023] [Indexed: 08/06/2023] Open
Abstract
BACKGROUND Autoimmune/type 1 diabetes mellitus (T1DM) is a recently described rare occurrence following the administration of adjuvants such as coronavirus disease 2019 (COVID-19) vaccines. This systematic review aimed to review all available literature on the potential association between COVID-19 vaccines and T1DM. METHODS The Directory of Open Access Journals, MEDLINE, Google Scholar, and Scopus were systematically searched for all published studies from inception to July 2022. Articles reporting T1DM development within 8 weeks of administration of COVID-19 vaccine were included. Two reviewers independently performed the risk of bias assessment following the Joanna Briggs Institute (JBI) Critical Appraisal Checklist for Case Reports. RESULTS Eight eligible studies were retrieved, comprising 12 patients diagnosed with T1DM after being vaccinated with a COVID-19 vaccine. Six patients (50%) reported T1DM after receiving the second dose. Five patients (41.7%) presented with diabetic ketoacidosis, of which four presented within the first eight days after vaccination. Five patients (41.7%) had genetic susceptibility, with RNA binding motif protein 45 (RBM45/DRB1) and major histocompatibility complex, class II, DQ beta 1 (HLA-DQB1) mutations being prominent. INTERPRETATION In this review, we have shown a small number of new-onset diabetes cases coincidently occurring soon after the COVID-19 vaccine, especially in those with genetic susceptibility. Despite being older, these patients had a similar phenotype to T1DM. While there might be a causal relationship between COVID-19 vaccines and T1DM development, this should not influence decisions regarding vaccination since the overall benefit outweighs the risk. Further larger prospective trials are needed to assess causal relationship and to clarify the potential roles of COVID-19 vaccine-derived antigens in autoimmune disease development. PROTOCOL REGISTRATION PROSPERO-CRD42022342093.
Collapse
Affiliation(s)
- Ali S Alsudais
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
| | - Raghad S Alkanani
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
| | - Abdulaziz B Fathi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
| | - Saleh S Almuntashiri
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
| | - Jafar N Jamjoom
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
| | - Mustafa A Alzhrani
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
| | - Alaa Althubaiti
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
| | - Suhaib Radi
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Jeddah, Saudi Arabia
- King Abdullah International Medical Research Centre, Jeddah, Saudi Arabia
- Department of Internal Medicine, Division of Endocrinology, Ministry of the National Guard-Health Affairs, Jeddah, Saudi Arabia
| |
Collapse
|
45
|
Mera-Gallego R, León-Rodríguez L, Barreiro Juncal M, Pérez Molina L, Guisado Barral B, Busto Domínguez I, Andrés-Rodríguez NF, Fornos-Pérez JA. [Pharmacovigilance of vaccines against COVID-19 in community pharmacies. Results after the second dose and comparison between both]. FARMACEUTICOS COMUNITARIOS 2023; 15:5-16. [PMID: 39157697 PMCID: PMC11328522 DOI: 10.33620/fc.2173-9218.(2023).21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/10/2023] [Indexed: 08/20/2024]
Abstract
Aim Detection and tracing of suspicious adverse reactions (ARs) in community pharmacies after the second of COVID-19 vaccine dose. Comparison between doses. Methods Design: prospective observational study. Subjects Vaccinated against COVID-19, of legal age, who consent to participate. Variables Number and percentage of participants with ARs. Number, type and frequency of ARs. Impact on their daily life. Relations between variables.Approved by the Galician Ethical Committee of Research with medicines. Results 693 participants with the 2nd dose, 63.6% women. Age 56.8 years. 312 (45.0%) vaccinated, 49.4% women and 37.3% men (p<0.0001), reported at least one AR: 43.9% with Comirnaty®, 37.7% with Vaxzevria®, 63.0% with Spikevax®.There were 972 ARs, 75.2% in women and 24.8% in men (p<0.0001). Mean 1.4/vaccinated (maximum 11). The most prevalent AR: pain at injection site 197 (28.4%), tiredness/fatigue 141 (20.3%), myalgia 112 (16.2%), headache 95 (13.7%), fever 84 (12.1%).51 participants with ARs needed professional help: 10 from the doctor, 6 in the emergency room, 3 in hospitals (1 referral), 33 in the pharmacy. 70 (15.1%) were prevented from their daily activity. 201 Ars from vaccinated persons were reported.Number of people vaccinated with ARs and the number of ARs were less with the 2nd dose (p<0.05).Inverse relationship (p<0.05) between "age" and "number of vaccinated with ARs", "need for professional care" and "prevented daily activity". Conclusions The number of vaccinated participants with ARs and their number was also high with the second dose, although lower than with the first. Women and younger people are predictive of increased risk of AR after vaccination against COVID-19.
Collapse
Affiliation(s)
- Rocío Mera-Gallego
- Farmacéutica comunitaria en VigoEspaña
- Grupo Berbés de Investigación y DocenciaGrupo Berbés de Investigación y DocenciaEspaña
| | - Laura León-Rodríguez
- Grupo Berbés de Investigación y DocenciaGrupo Berbés de Investigación y DocenciaEspaña
- Doctora en Farmacia. Farmacéutica comunitaria en Ourense.España
| | - Miriam Barreiro Juncal
- Farmacéutica comunitaria en VigoEspaña
- Grupo Berbés de Investigación y DocenciaGrupo Berbés de Investigación y DocenciaEspaña
| | - Laura Pérez Molina
- Farmacéutica comunitaria en VigoEspaña
- Grupo Berbés de Investigación y DocenciaGrupo Berbés de Investigación y DocenciaEspaña
| | - Bibiana Guisado Barral
- Grupo Berbés de Investigación y DocenciaGrupo Berbés de Investigación y DocenciaEspaña
- Farmacéutica comunitaria en Crecente (Pontevedra).CEspaña
| | - Iván Busto Domínguez
- Grupo Berbés de Investigación y DocenciaGrupo Berbés de Investigación y DocenciaEspaña
- Departamento técnico del Colegio Oficial de Farmacéuticos de PontevedraColegio Oficial de Farmacéuticos de PontevedraEspaña
| | - N. Floro Andrés-Rodríguez
- Grupo Berbés de Investigación y DocenciaGrupo Berbés de Investigación y DocenciaEspaña
- Doctor en FarmaciaFarmaciaEspaña
| | - José A. Fornos-Pérez
- Grupo Berbés de Investigación y DocenciaGrupo Berbés de Investigación y DocenciaEspaña
- Doctor en Farmacia. Farmacéutico comunitario en Cangas do Morrazo (Pontevedra)España
| |
Collapse
|
46
|
Park HJ, Bang YJ, Kwon SP, Kwak W, Park SI, Roh G, Bae SH, Kim JY, Kwak HW, Kim Y, Yoo S, Kim D, Keum G, Bang EK, Hong SH, Nam JH. Analyzing immune responses to varied mRNA and protein vaccine sequences. NPJ Vaccines 2023; 8:84. [PMID: 37271785 DOI: 10.1038/s41541-023-00684-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 05/25/2023] [Indexed: 06/06/2023] Open
Abstract
In response to the COVID-19 pandemic, different types of vaccines, such as inactive, live-attenuated, messenger RNA (mRNA), and protein subunit, have been developed against SARS-CoV-2. This has unintentionally created a unique scenario where heterologous prime-boost vaccination against a single virus has been administered to a large human population. Here, we aimed to analyze whether the immunization order of vaccine types influences the efficacy of heterologous prime-boost vaccination, especially mRNA and protein-based vaccines. We developed a new mRNA vaccine encoding the hemagglutinin (HA) glycoprotein of the influenza virus using the 3'-UTR and 5'-UTR of muscle cells (mRNA-HA) and tested its efficacy by heterologous immunization with an HA protein vaccine (protein-HA). The results demonstrated higher IgG2a levels and hemagglutination inhibition titers in the mRNA-HA priming/protein-HA boosting (R-P) regimen than those induced by reverse immunization (protein-HA priming/mRNA-HA boosting, P-R). After the viral challenge, the R-P group showed lower virus loads and less inflammation in the lungs than the P-R group did. Transcriptome analysis revealed that the heterologous prime-boost groups had differentially activated immune response pathways, according to the order of immunization. In summary, our results demonstrate that the sequence of vaccination is critical to direct desired immune responses. This study demonstrates the potential of a heterologous vaccination strategy using mRNA and protein vaccine platforms against viral infection.
Collapse
Affiliation(s)
- Hyeong-Jun Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
- BK Plus Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
- SML Biopharm, Gwangmyeong, 14353, Republic of Korea
| | - Yoo-Jin Bang
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
- BK Plus Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
- SML Biopharm, Gwangmyeong, 14353, Republic of Korea
| | - Sung Pil Kwon
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Woori Kwak
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Sang-In Park
- SML Biopharm, Gwangmyeong, 14353, Republic of Korea
| | - Gahyun Roh
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
- BK Plus Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Seo-Hyeon Bae
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
- BK Plus Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Jae-Yong Kim
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea
- BK Plus Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea
- SML Biopharm, Gwangmyeong, 14353, Republic of Korea
| | - Hye Won Kwak
- SML Biopharm, Gwangmyeong, 14353, Republic of Korea
| | - Yongkwan Kim
- SML Biopharm, Gwangmyeong, 14353, Republic of Korea
| | - Soyeon Yoo
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Daegeun Kim
- SML Biopharm, Gwangmyeong, 14353, Republic of Korea
| | - Gyochang Keum
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Eun-Kyoung Bang
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| | - So-Hee Hong
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, 07804, Republic of Korea.
| | - Jae-Hwan Nam
- Department of Medical and Biological Sciences, The Catholic University of Korea, Bucheon, Republic of Korea.
- BK Plus Department of Biotechnology, The Catholic University of Korea, Bucheon, Republic of Korea.
| |
Collapse
|
47
|
Haider SMS, Alvi SA, Khan H, Majeed R, Syed T, Anwar A, Hashmi AA. Common Side Effects of Pfizer COVID-19 Vaccine: An Experience From Pakistan. Cureus 2023; 15:e40878. [PMID: 37492805 PMCID: PMC10363686 DOI: 10.7759/cureus.40878] [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] [Accepted: 06/23/2023] [Indexed: 07/27/2023] Open
Abstract
Introduction The severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) epidemic spread quickly. Vaccines are now being distributed to stop the infectious spread and halt fatalities. The Pfizer-BioNTech vaccine was the first mRNA-based vaccine introduced to boost immunity against COVID-19; however, it could lead to various adverse reactions. Therefore, the aim of this study was to assess the prevalence of Pfizer vaccine side effects among participants. Methods This was a multicenter cross-sectional study that was performed using a non-probability sampling method. The study period was about six months from March 1, 2022, to August 31, 2022. A total of 1000 participants who received two doses of the Pfizer vaccine met the inclusion criteria. Demographic details of participants, for example, gender, age, comorbidities, Pfizer vaccine with both doses along with booster dose, previous exposure to coronavirus disease 2019 (COVID-19) infection, and the incidence of any local and systemic side effects following the first and second doses of vaccine, were reported. Results The study findings showed that out of 1000 participants, 644 (64.4%) were males and 356 (35.6%) were females; their mean age was 43.06±14.98 years. Among them, 280 (28.0%) had hypertension and 356 (35.6%) had diabetes. Following the first dose of the Pfizer vaccine, burning at the injection site and fever were the most commonly reported side effects in 704 (70.4%) and 700 (70.0%) participants, respectively. Following the second dose of the Pfizer vaccine, muscle pain was the most commonly reported side effect in 628 (62.8%) participants. Conclusion This study concluded that the most frequent adverse effects of the Pfizer vaccine were burning at the injection site, fever, pain at the injection site, muscle pain, swelling at the injection site, and joint pain. Moreover, the first dose was associated with more side effects than the second dose.
Collapse
Affiliation(s)
| | - Shaf Ali Alvi
- Internal Medicine, Hamdard College of Medicine and Dentistry, Karachi, PAK
| | - Hamza Khan
- General Surgery, Hamdard College of Medicine and Dentistry, Karachi, PAK
| | - Rameen Majeed
- Biochemistry, Jinnah Sindh Medical University, Karachi, PAK
| | - Tatheer Syed
- Public Health, Jinnah Sindh Medical University, Karachi, PAK
| | - Adnan Anwar
- Physiology, Hamdard College of Medicine and Dentistry, Karachi, PAK
- Internal Medicine, Essa General Hospital, Karachi, PAK
| | - Atif A Hashmi
- Pathology, Liaquat National Hospital and Medical College, Karachi, PAK
| |
Collapse
|
48
|
Abstract
The relationship between viral infection and onset of autoimmune diseases such as systemic lupus erythematosus remains uncertain. During the COVID-19 pandemic, organ-specific and multisystemic autoimmune phenomena temporally related to the viral infection have been described. Immune dysregulation triggered by the SARS-CoV-2 virus leading to hyperactivation of both the innate and adaptive immune systems contributes to the excessive production of pro-inflammatory cytokines, autoantibodies, and subsequent autoimmune manifestations. We report two patients without known autoimmune diseases who developed lupus nephritis shortly after a documented mild SARS-CoV-2 infection. Together with other similar cases in the literature, the observation supports a viral trigger of the development of systemic lupus erythematosus in susceptible individuals.
Collapse
Affiliation(s)
- Chi Chiu Mok
- Department of Medicine, Tuen Mun Hospital, Hong Kong, China
| | - Chiu Sum Chu
- Department of Medicine, Tuen Mun Hospital, Hong Kong, China
| | - Sau Mei Tse
- Department of Medicine, Tuen Mun Hospital, Hong Kong, China
| |
Collapse
|
49
|
Pranić SM, Vasanthan LT, Thompson JY, Mishra V, Kumar P, Ananda RA, Malih N, Chan KK. Protocol of a scoping review of systematic reviews and meta-analyses about COVID-19 vaccines and associated adverse events from vaccination. PLoS One 2023; 18:e0285442. [PMID: 37163502 PMCID: PMC10171674 DOI: 10.1371/journal.pone.0285442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 04/23/2023] [Indexed: 05/12/2023] Open
Abstract
The continuous dissemination of coronavirus disease of 2019 (COVID-19) literature can inform decision-makers and the public. Since the widespread use of COVID-19 vaccines, more systematic reviews have summarized the effectiveness and reported adverse events associated with vaccination. Previous systematic and scoping reviews on COVID-19 summarized various aspects surrounding COVID-19, however, a scoping review is needed to summarize the characteristics of COVID-19 vaccines and associated adverse events reported in systematic reviews and meta-analyses to provide comprehensive evidence for informed medical decision-making. We will conduct a scoping review concerning COVID-19 vaccines and adverse events from vaccines. We will search from December 2019 to present in Epistemonikos, Campbell Library, CINAHL (Ovid), MEDLINE (Ovid), Scopus, CENTRAL (Ovid), Web of Science, WHO COVID-19 database, Joanna Briggs Institute of Excellence, and COVID-19 Evidence Reviews resource. We will include systematic reviews, meta-analyses, or both of randomized controlled trials and observational studies and exclude individual randomized controlled trials and observational studies. Abstracts and full-texts will be screened prior to selection. Investigators will independently use a calibrated quantitative and qualitative data extraction sheet and rate the quality of articles with AMSTAR, resolving disagreements to aim for good agreement (≥80%). An updated scoping review of the characteristics and safety of COVID-19 vaccines would highlight the accuracy of the evidence to inform decision-making concerning COVID-19 vaccination.
Collapse
Affiliation(s)
- Shelly Melissa Pranić
- Department of Public Health, University of Split School of Medicine, Split, Croatia
- Cochrane Croatia, Split, Croatia
| | - Lenny T. Vasanthan
- Department of Physical Medicine and Rehabilitation, Christian Medical College, Vellore, India
| | - Jacqueline Y. Thompson
- Institute of Applied Health Research, University of Birmingham, University of Birmingham, Birmingham, United Kingdom
| | - Vinayak Mishra
- Department of Children’s Health, University of Liverpool, Liverpool, United Kingdom
| | | | | | - Narges Malih
- Global Health Research Group, University of the Balearic Islands (UIB), Palma, Spain
- Social Determinants of Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ka-King Chan
- Pamela Youde Nethersole Eastern Hospital, Hong Kong, China
| |
Collapse
|
50
|
Terada-Hirashima J, Takamatsu Y, Shimizu Y, Uemura Y, Takeuchi JS, Tomita N, Matsuda K, Maeda K, Yamamoto S, Fukunaga A, Ohmagari N, Mikami A, Sonoda K, Ujiie M, Mitsuya H, Sugiura W. Immunogenicity and safety of single booster dose of KD-414 inactivated COVID-19 vaccine in adults: An open-label, single-center, non-randomized, controlled study in Japan. Hum Vaccin Immunother 2023; 19:2193074. [PMID: 37052247 PMCID: PMC10114994 DOI: 10.1080/21645515.2023.2193074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023] Open
Abstract
Although vaccines for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) disease 2019 (COVID-19) induce effective immune responses, vaccination with booster doses is necessary because of waning immunity. We conducted an open-label, non-randomized, single-arm study in adults in Japan to assess the immunogenicity and safety of a single booster dose of the KD-414 purified whole-SARS-CoV-2-virion inactivated vaccine candidate after vaccination with a primary series of BNT162b2. The primary endpoint was serum neutralizing activity at 7 days after booster injection compared with the primary series of BNT162b2. The SARS-CoV-2-structural protein-binding antibody level and T cell response against SARS-CoV-2-Spike (S) peptides were also examined as secondary endpoints, and safety profile assessments were conducted. Twenty subjects who participated in a previous study declined an injection of KD-414 (non-KD-414 group) and received a booster dose of BNT162b2 instead. The non-KD-414 group was compared to the KD-414 group as a secondary outcome. A single dose of KD-414 induced lower serum neutralizing activity against the wild-type virus within 7 days compared to after the primary series of BNT162b2 but significantly induced anti-SARS-CoV-2-S1-receptor-binding domain-binding immunoglobulin G (IgG) antibodies and SARS-CoV-2-S peptide-specific CD4+ and CD8+ T cell responses. Local or systemic symptoms were significantly lower in the participants who received KD-414 than in those who received BNT162b2 as the third COVID-19 vaccine dose. The present data indicate that a single booster dose of KD-414 induces a substantial immune response in BNT162b2-primed individuals and has a good safety profile, thereby supporting further clinical trials to identify rational targets.
Collapse
Affiliation(s)
- Junko Terada-Hirashima
- Department of Clinical Research Promotion, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
- Department of Respiratory Medicine, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Yuki Takamatsu
- Department of Refractory Viral Diseases, National Center for Global Health and Medicine Research Institute, Shinjuku-ku, Tokyo, Japan
| | - Yosuke Shimizu
- Department of Data Science, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yukari Uemura
- Department of Data Science, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Junko S Takeuchi
- Department of Academic-Industrial Partnerships Promotion, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Noriko Tomita
- Department of Clinical Research Promotion, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kouki Matsuda
- Department of Refractory Viral Diseases, National Center for Global Health and Medicine Research Institute, Shinjuku-ku, Tokyo, Japan
| | - Kenji Maeda
- Department of Refractory Viral Diseases, National Center for Global Health and Medicine Research Institute, Shinjuku-ku, Tokyo, Japan
| | - Shohei Yamamoto
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Ami Fukunaga
- Department of Epidemiology and Prevention, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Ayako Mikami
- Department of Clinical Research Promotion, Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kengo Sonoda
- Development Department, KM Biologics Co. Ltd, Kumamoto, Japan
| | - Mugen Ujiie
- Disease Control and Prevention Center, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Hiroaki Mitsuya
- Department of Refractory Viral Diseases, National Center for Global Health and Medicine Research Institute, Shinjuku-ku, Tokyo, Japan
| | - Wataru Sugiura
- Center for Clinical Sciences, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|