1
|
Yang BF, Jin J, He XR, Yang ZH, Qian X, Tong YQ, Ke CX, Li ZH, Li ZX, Zhong LF, Gan ZH, Zhang XF, Zeng Y. Immunogenicity and safety of SARS-CoV-2 recombinant protein vaccine (CHO cell) LYB001 as a heterologous booster following two- or three-dose inactivated COVID-19 vaccine in adults aged ≥18 years: interim results of a randomized, active-controlled, double-blinded, phase 3 trial. Expert Rev Vaccines 2025; 24:81-90. [PMID: 39720838 DOI: 10.1080/14760584.2024.2446288] [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: 06/12/2024] [Revised: 10/13/2024] [Accepted: 12/20/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND LYB001 is a recombinant protein COVID-19 vaccine displaying a receptor-binding domain (RBD) in a highly immunogenic array on virus-like particles (VLPs). This study assessed the immunogenicity and safety of LYB001 as a booster. RESEARCH DESIGN AND METHODS In this randomized, active-controlled, double-blinded, phase 3 trial, participants aged ≥ 18 years received a booster with LYB001 or ZF2001 (Recombinant COVID-19 Vaccine). The primary endpoint was to compare the geometric mean titer (GMT) of neutralizing antibodies against Omicron BA.4/5 at 14 days after the booster. RESULTS Overall, 1,200 participants aged ≥ 18 years were enrolled, 599 received LYB001, and 601 received ZF2001. Based on similar baseline level, the 14-day GMT ratio (LYB001/ZF2001) against Omicron BA.4/5 was 1.39 (95% CI: 1.25, 1.56), demonstrating superiority (95% CI lower limit > 1) of LYB001. The spike protein-binding IgG concentrations induced by LYB001 were significantly higher than those induced by ZF2001 on day 14 and day 28 after the booster (p-value <0.0001). LYB001 recipients reported more adverse reactions than ZF2001 recipients (21.4% vs. 15.0%); however, all adverse reactions in the LYB001 group were mild-to-moderate. CONCLUSIONS LYB001 is highly immunogenic and retains a well-characterized safety profile in adults aged ≥ 18 years. CLINICAL TRIAL REGISTRATION www.clinicaltrials.gov, identifier is NCT05664932.
Collapse
Affiliation(s)
- Bei-Fang Yang
- Center for Clinical Trial of Vaccines, Hubei Institute for Infectious Disease, Control and Prevention, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Jing Jin
- Center for Clinical Trial of Vaccines, Hubei Institute for Infectious Disease, Control and Prevention, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
- Guangzhou Patronus Biotech Co., Ltd., Guangzhou, China
| | - Xi-Ran He
- Center for Clinical Trial of Vaccines, Hubei Institute for Infectious Disease, Control and Prevention, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
- Guangzhou Patronus Biotech Co., Ltd., Guangzhou, China
| | - Zhong-Hua Yang
- Center for Clinical Trial of Vaccines, Hubei Institute for Infectious Disease, Control and Prevention, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
- Guangzhou Patronus Biotech Co., Ltd., Guangzhou, China
| | - Xiao'ai Qian
- Yantai Patronus Biotech Co., Ltd., Yantai, China
| | - Ye-Qing Tong
- Hubei Institute for Infectious Disease Control and Prevention, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Chang-Xian Ke
- Immunization Prevention Department Shiyan City Yunyang District Center for Disease Control and Prevention, Shiyan, China
| | - Zhao-Hong Li
- Immunization Prevention Department Shiyan City Yunyang District Center for Disease Control and Prevention, Shiyan, China
| | - Zhao-Xia Li
- Immunization Prevention Department, Gucheng Center for Disease Control and Prevention, Xiangyang, China
| | - Lin-Feng Zhong
- Immunization Prevention Department, Wuxue City Center for Disease Control and Prevention, Wuxue, China
| | - Ze-Hong Gan
- Immunization Prevention Department, Wuxue City Center for Disease Control and Prevention, Wuxue, China
| | - Xian-Feng Zhang
- Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
| | - Ying Zeng
- Center for Clinical Trial of Vaccines, Hubei Institute for Infectious Disease, Control and Prevention, Hubei Provincial Center for Disease Control and Prevention, Wuhan, China
- Guangzhou Patronus Biotech Co., Ltd., Guangzhou, China
| |
Collapse
|
2
|
Edeling MA, Earnest L, Carrera Montoya J, Yap AHY, Mumford J, Roberts J, Wong CY, Hans D, Grima J, Bisset N, Bodle J, Rockman S, Torresi J. Development of Methods to Produce SARS CoV-2 Virus-Like Particles at Scale. Biotechnol Bioeng 2025; 122:1118-1129. [PMID: 39936889 PMCID: PMC11975197 DOI: 10.1002/bit.28937] [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: 05/14/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 02/13/2025]
Abstract
The devastating global toll precipitated by the SARS CoV-2 outbreak and the profound impact of vaccines in stemming that outbreak has established the need for molecular platforms capable of rapidly delivering effective, safe and accessible medical interventions in pandemic preparedness. We describe a simple, efficient and adaptable process to produce SARS CoV-2 virus-like particles (VLPs) that can be readily scaled for manufacturing. A rapid but gentle method of tangential flow filtration using a 100 kDa semi-permeable membrane concentrates and buffer exchanges 0.5 L of SARS CoV-2 VLP containing supernatant into low salt and optimal pH for anion exchange chromatography. VLPs are washed, eluted under high salt, dialyzed into physiological buffer, sterile filtered and aliquoted for storage at -80°C. Purification is completed in less than 2 days. A simple quality control process includes Western blot for coincident detection of Spike, Membrane and Envelope protein as a proxy for intact VLP, ELISA to detect conformationally sensitive Spike using readily available anti-Spike and/or anti-RBD antibodies, and negative stain and immunogold electron microscopy to validate particulate, Spike crowned VLPs. This process to produce SARS CoV-2 VLPs for preclinical studies serves as a roadmap for preparation of more distantly related VLPs for pandemic preparedness.
Collapse
Affiliation(s)
- Melissa A. Edeling
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Linda Earnest
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Julio Carrera Montoya
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Ashley Huey Yiing Yap
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Jamie Mumford
- Victorian Infectious Diseases Reference laboratoryRoyal Melbourne Hospital at the Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Jason Roberts
- Victorian Infectious Diseases Reference laboratoryRoyal Melbourne Hospital at the Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
- Department of Infectious DiseasesThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Chinn Yi Wong
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| | - Dhiraj Hans
- Research, Innovation & Commercialisation, Faculty of Medicine, Dentistry & Health SciencesThe University of MelbourneParkvilleVictoriaAustralia
| | - Joseph Grima
- Seqirus, Vaccine Innovation UnitParkvilleVictoriaAustralia
| | - Nicole Bisset
- Seqirus, Vaccine Innovation UnitParkvilleVictoriaAustralia
| | - Jesse Bodle
- Seqirus, Vaccine Innovation UnitParkvilleVictoriaAustralia
| | - Steven Rockman
- Seqirus, Vaccine Innovation UnitParkvilleVictoriaAustralia
| | - Joseph Torresi
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| |
Collapse
|
3
|
Monschein T, Zrzavy T, Rommer PS, Meuth SG, Chan A, Berger T, Hartung HP. SARS-CoV-2 Vaccines and Multiple Sclerosis: An Update. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200393. [PMID: 40279527 DOI: 10.1212/nxi.0000000000200393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/18/2025] [Indexed: 04/27/2025]
Abstract
The highly contagious zoonosis coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was declared a pandemic by the World Health Organization on March 11, 2020, and has led to a global health crisis with nearly 777 million confirmed infections and over 7 million deaths worldwide by November 10, 2024.1-3 Over time, various variants emerged, with Omicron and its sublines dominating the world over the past 3 years.4 In addition, there is increasing evidence regarding the immune response of SARS-CoV-2 vaccines, especially for people with multiple sclerosis (MS) receiving disease-modifying therapies. Hence, with this review, we aim to provide an updated overview and recommendations for clinical practice regarding MS and SARS-CoV-2 vaccines, including efficacy and safety, SARS-CoV-2 variants, vaccine hesitancy, and the immune response under treatment with respective disease-modifying therapies.
Collapse
Affiliation(s)
- Tobias Monschein
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Tobias Zrzavy
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria
| | - Paulus S Rommer
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria
| | - Sven G Meuth
- Department of Neurology, Medical Faculty, Universitätsklinikum Düsseldorf, Heinrich-Heine-University, Germany
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Thomas Berger
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria
| | - Hans-Peter Hartung
- Department of Neurology, Medical University of Vienna, Austria
- Comprehensive Center for Clinical Neurosciences and Mental Health, Medical University of Vienna, Austria
- Department of Neurology, Medical Faculty, Universitätsklinikum Düsseldorf, Heinrich-Heine-University, Germany
- Brain and Mind Center, University of Sydney, Australia; and
- Department of Neurology, Palacky University, Olomouc, Czech Republic
| |
Collapse
|
4
|
Atarashi Y, Kim J, Irino Y, Amano M, Tsuchiya K, Maeda K, Terada M, Iwamoto N, Shimada S, Mitsuya H, Yanagida M, Takamatsu Y. A high-throughput, fully automated competition assay to evaluate SARS-CoV-2 neutralizing responses and epitope specificity in clinical samples. Sci Rep 2025; 15:11589. [PMID: 40185856 PMCID: PMC11971398 DOI: 10.1038/s41598-025-94317-2] [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: 02/04/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025] Open
Abstract
Coronavirus disease-2019 (COVID-19) remains a critical global health concern. We developed a fully automated, high-throughput competition immunoassay to elucidate how epitope recognition on the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike receptor-binding domain (RBD) correlates with neutralizing activity. Analysis of clinical samples from both SARS-CoV-2-infected and vaccinated individuals revealed that vaccination elicits significantly higher antibody titers across multiple S1 subunit epitopes compared to natural infection. Notably, median antibody levels against the receptor-binding motif (RBM) exceeded 50% in both cohorts, highlighting the RBM as a key target for antibody induction irrespective of immune origin. Furthermore, the strongest correlation with neutralizing activity was observed for antibodies directed against the broader S1 subunit, indicating that epitopes outside the RBM also contribute to neutralization. These findings underscore the importance of both RBM- and non-RBM-directed antibodies in effective immune defense against SARS-CoV-2. Our assay enables large-scale, reliable quantification of neutralizing antibodies and provides critical insights for developing improved diagnostic antigens and vaccine strategies aimed at eliciting robust, multi-epitope immune responses.
Collapse
Affiliation(s)
- Yusuke Atarashi
- Central Research Laboratories, Sysmex Corporation, Kobe, 651-2271, Japan
| | - Jeeeun Kim
- Central Research Laboratories, Sysmex Corporation, Kobe, 651-2271, Japan
| | - Yasuhiro Irino
- Central Research Laboratories, Sysmex Corporation, Kobe, 651-2271, Japan
| | - Masayuki Amano
- Department of Clinical Retrovirology, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-8556, Japan
| | - Kiyoto Tsuchiya
- AIDS Clinical Center, Center hospital of the National Center for Global Health and Medicine, Tokyo, 162-8655, Japan
| | - Kenji Maeda
- Refractory Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, 162-8655, Japan
- Division of Antiviral Therapy, Joint Research Center for Human Retrovirus Infection, Kagoshima University, Kagoshima, Japan, 890-8544
| | - Mari Terada
- Department of Disease Control Center, Center Hospital of the National Center for Global Health and Medicine, Tokyo, 162-8655, Japan
| | - Noriko Iwamoto
- Department of Disease Control Center, Center Hospital of the National Center for Global Health and Medicine, Tokyo, 162-8655, Japan
| | - Shinya Shimada
- Japan Community Healthcare Organization, Kumamoto General Hospital, 866- 8660, Kumamoto, Japan
| | - Hiroaki Mitsuya
- Refractory Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, 162-8655, Japan
- Experimental Retrovirology Section, HIV and AIDS Malignancy Branch, National Cancer Institute, National Institutes of Health, 20892-1868, Bethesda, MD, USA
- Division of Clinical Sciences, Kumamoto University Hospital, 860-8556, Kumamoto, Japan
| | - Masatoshi Yanagida
- Central Research Laboratories, Sysmex Corporation, Kobe, 651-2271, Japan
| | - Yuki Takamatsu
- Refractory Viral Diseases, National Center for Global Health and Medicine Research Institute, Tokyo, 162-8655, Japan.
| |
Collapse
|
5
|
Liu W, Li Y, Li X, Wang F, Qi R, Zhu T, Li J. Pooled Analysis of the Effect of Pre-Existing Ad5 Neutralizing Antibodies on the Immunogenicity of Adenovirus Type 5 Vector-Based COVID-19 Vaccine from Eight Clinical Trials. Vaccines (Basel) 2025; 13:333. [PMID: 40266233 PMCID: PMC11945733 DOI: 10.3390/vaccines13030333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 03/19/2025] [Indexed: 04/24/2025] Open
Abstract
Background: Pre-existing adenovirus immunity restricts the utilization of adenovirus-vectored vaccines. The current study aims to conduct a pooled analysis of eight clinical trials to evaluate the influence of pre-existing Ad5 neutralizing antibodies on immunogenicity of Ad5-nCoV. Methods: The primary outcome indicator of this pooled analysis is the geometric mean titers (GMTs) of live SARS-CoV-2 NAbs against the wild-type strain on day 28 post-vaccination. Participants were divided into two cohorts: an adolescent cohort comprising individuals aged 6-17 years and an adult cohort with individuals aged 18 years and older. Within each cohort, individuals were further categorized into three subgroups based on their Ad5-nCoV vaccination schedules: one subgroup received a single intramuscular dose as the primary regimen (Ad5-IM-prime), another received an intramuscular dose as the heterologous prime-boost regimen (Ad5-IM-boost), and the last subgroup received an aerosolized dose as the heterologous prime-boost regimen (Ad5-IH-boost). Results: A total of 3512 participants were included in this pooled analysis. In the Ad5-IM-prime subgroup, there were 1001 adolescents and 1450 adults; in the Ad5-IM-boost subgroup, there were 65 adolescents and 396 adults; and in the Ad5-IH-boost subgroup, there were 207 adolescents and 393 adults. In the adult cohort, the GMTs of NAbs against wild-type SARS-CoV-2 on day 28 post-vaccination for the Ad5-IM-prime, Ad5-IM-boost, and Ad5-IH-boost subgroups were 35.6 (95% CI: 32.0, 39.7), 358.3 (95% CI: 267.6, 479.6), and 2414.1 (95% CI: 2006.9, 2904.0), respectively, with negative (less than 1:12) pre-existing NAb titers compared to 10.7 (95% CI: 9.1, 12.6), 116.9 (95% CI: 84.9, 161.1), and 762.7 (95% CI: 596.2, 975.8), respectively, with high (greater than 1:1000) pre-existing NAb titers. A similar trend was observed in the adolescent cohort, where pre-existing immunity was found to reduce the peak of live SARS-CoV-2 Nabs post-vaccination. Conclusions: Regardless of whether Ad5-nCoV is administered as a primary vaccination regimen or as a heterologous prime-boost strategy, a negative impact on immunogenicity can still be observed in the presence of high pre-existing immunity. However, when primary immunization is achieved with inactivated COVID-19 vaccines, aerosol inhalation can significantly enhance the immunogenicity of Ad5-nCoV compared to intramuscular injections of Ad5-nCoV as a booster.
Collapse
Affiliation(s)
- Wenqing Liu
- School of Public Health, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China; (W.L.); (Y.L.); (R.Q.)
| | - Yuqing Li
- School of Public Health, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China; (W.L.); (Y.L.); (R.Q.)
| | - Xiaolong Li
- CanSino Biologics Inc., Tianjin 300457, China; (X.L.); (F.W.)
| | - Feiyu Wang
- CanSino Biologics Inc., Tianjin 300457, China; (X.L.); (F.W.)
| | - Runjie Qi
- School of Public Health, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China; (W.L.); (Y.L.); (R.Q.)
| | - Tao Zhu
- CanSino Biologics Inc., Tianjin 300457, China; (X.L.); (F.W.)
| | - Jingxin Li
- School of Public Health, National Vaccine Innovation Platform, Nanjing Medical University, Nanjing 211166, China; (W.L.); (Y.L.); (R.Q.)
- Jiangsu Provincial Medical Innovation Center, National Health Commission Key Laboratory of Enteric Pathogenic Microbiology, Jiangsu Provincial Center for Disease Control and Prevention, Jiangsu Provincial Academy of Preventive Medicine, Nanjing 210009, China
| |
Collapse
|
6
|
Luo S, Yin L, Liu X, Wang X. Advances in Virus Biorecognition and Detection Techniques for the Surveillance and Prevention of Infectious Diseases. BIOSENSORS 2025; 15:198. [PMID: 40136995 PMCID: PMC11940537 DOI: 10.3390/bios15030198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Viral infectious diseases pose a serious threat to global public health due to their high transmissibility, rapid mutation rates, and limited treatment options. Recent outbreaks of diseases such as plague, monkeypox, avian influenza, and coronavirus disease 2019 (COVID-19) have underscored the urgent need for efficient diagnostic and surveillance technologies. Focusing on viral infectious diseases that seriously threaten human health, this review summarizes and analyzes detection techniques from the perspective of combining viral surveillance and prevention advice, and discusses applications in improving diagnostic sensitivity and specificity. One of the major innovations of this review is the systematic integration of advanced biorecognition and detection technologies, such as bionanosensors, rapid detection test strips, and microfluidic platforms, along with the exploration of artificial intelligence in virus detection. These technologies address the limitations of traditional methods and enable the real-time monitoring and early warning of viral outbreaks. By analyzing the application of these technologies in the detection of pathogens, new insights are provided for the development of next-generation diagnostic tools to address emerging and re-emerging viral threats. In addition, we analyze the current progress of developed vaccines, combining virus surveillance with vaccine research to provide new ideas for future viral disease prevention and control and vaccine development, and call for global attention and the development of new disease prevention and detection technologies.
Collapse
Affiliation(s)
- Shuwen Luo
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China;
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China;
| | - Lihong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China;
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China;
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China;
| |
Collapse
|
7
|
Adilović M. COVID-19 related complications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:259-314. [PMID: 40246346 DOI: 10.1016/bs.pmbts.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The COVID-19 pandemic has significantly impacted global healthcare systems, revealed vulnerabilities and prompted a re-evaluation of medical practices. Acute complications from the virus, including cardiovascular and neurological issues, have underscored the necessity for timely medical interventions. Advances in diagnostic methods and personalized therapies have been pivotal in mitigating severe outcomes. Additionally, Long COVID has emerged as a complex challenge, affecting various body systems and leading to respiratory, cardiovascular, neurological, psychological, and musculoskeletal problems. This broad spectrum of complications highlights the importance of multidisciplinary management approaches that prioritize therapy, rehabilitation, and patient-centered care. Vulnerable populations such as paediatric patients, pregnant women, and immunocompromised individuals face unique risks and complications, necessitating continuous monitoring and tailored management strategies to reduce morbidity and mortality associated with COVID-19.
Collapse
Affiliation(s)
- Muhamed Adilović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnička cesta, Sarajevo, Bosnia and Herzegovina.
| |
Collapse
|
8
|
Lu B, Chaudhary O, Banoth B, Nadkarni J, Zong W, Mausser E, Danz H, Motwani M, Ruiz S, Zhang D, Nageshwaran G, Rokbi B, Warren W, DeRosa F, Chivukula S. Impact of Extended Dosing Intervals and Ipsilateral Versus Contralateral Boosting on mRNA Vaccine Immunogenicity in Mice. Vaccines (Basel) 2025; 13:263. [PMID: 40266125 PMCID: PMC11946721 DOI: 10.3390/vaccines13030263] [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: 01/13/2025] [Revised: 02/14/2025] [Accepted: 02/26/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Although mRNA vaccines have the potential to be developed and deployed rapidly to combat infectious diseases, the ideal method of administration and boosting schedule strategy for generating optimal immunogenicity is an area of active research. We compared the immune responses resulting from different schedules for prime-boost and boosting either ipsilaterally or contralaterally in relation to the initial vaccine dose. METHODS Influenza hemagglutinin (HA) was used as a model antigen for different vaccination regimens in mice using both mRNA lipid nanoparticles (mRNA-LNP) and AF03-adjuvanted recombinant protein (rHA-AF03) vaccines. RESULTS Increasing the prime-boost interval resulted in higher levels of serum anti-HA IgG and functional antibody hemagglutination inhibition (HAI) responses in mRNA-LNP-vaccinated animals, which correlated with an induction of germinal center (GC) B cells and follicular helper T (Tfh) cells in lymph nodes. In addition, longer prime-boost intervals resulted in higher levels of IL-2 and TNF-α producing CD4+ T cells two weeks after boosting. The number of Ig-secreting long-lived plasma cells increased with the length of prime-boost intervals. Contralateral boosting resulted in an increase in HAI titers and GC B cells compared to an ipsilateral boost. However, significantly higher numbers of GC B cells were induced in the draining lymph nodes following ipsilateral boosting than in the non-draining lymph nodes. CONCLUSIONS Overall, our data provides insights into the immune mechanisms of action of mRNA-LNP to develop the optimal vaccine regimen for mRNA vaccine platforms.
Collapse
Affiliation(s)
- Bin Lu
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Omkar Chaudhary
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Balaji Banoth
- Former Employee of Sanofi, 200 West St., Waltham, MA 02451, USA
| | - Janhavi Nadkarni
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Wei Zong
- Translational and Early Development Biostatistics, Sanofi, 200 West St., Waltham, MA 02451, USA (D.Z.)
| | - Emilie Mausser
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Hillary Danz
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Mona Motwani
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Sophie Ruiz
- mRNA Center of Excellence, Sanofi, 1541 Avenue Marcel Mérieux, 69280 MarcyL’Etoile, France;
| | - Donghui Zhang
- Translational and Early Development Biostatistics, Sanofi, 200 West St., Waltham, MA 02451, USA (D.Z.)
| | - Gopinath Nageshwaran
- Global Antigen Design, Sanofi, 200 West St., Waltham, MA 02451, USA; (G.N.); (W.W.)
| | - Bachra Rokbi
- Global Antigen Design, Sanofi, 1541 Avenue Marcel Mérieux, 69280 Marcy L’Etoile, France;
| | - William Warren
- Global Antigen Design, Sanofi, 200 West St., Waltham, MA 02451, USA; (G.N.); (W.W.)
| | - Frank DeRosa
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| | - Sudha Chivukula
- mRNA Center of Excellence, Sanofi, 200 West St., Waltham, MA 02451, USA; (B.L.); (O.C.); (E.M.); (H.D.); (M.M.); (F.D.)
| |
Collapse
|
9
|
Chan JCH, Chan EYH, Cheng SMS, Leung D, Ho FTW, Tong PC, Lai WM, Lee MHL, Chim S, Tsang LCH, Kwan TC, Cheuk YC, Wang M, Wong HHW, Lee AMT, Li WY, Chan SM, Tam IYS, Lam JHY, Zhang K, Tu W, Peiris M, Rosa Duque JS, Lau YL, Ma ALT. Humoral and cellular immunogenicity of a fourth dose BNT162b2 in children with chronic kidney diseases. Clin Kidney J 2025; 18:sfaf052. [PMID: 40123970 PMCID: PMC11926591 DOI: 10.1093/ckj/sfaf052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Indexed: 03/25/2025] Open
Abstract
Background Children with chronic kidney disease (CKD) are at risk of severe complications after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and are recommended to receive vaccine boosters. Although coronavirus disease 2019 (COVID-19) boosters are effective in providing immune responses among healthy children, data on the use of a fourth dose among children with CKD are limited. Methods We prospectively investigated the immunogenicity and safety of a fourth dose of BNT162b2 in children with CKD. Dosages were 0.1 mL and 0.3 mL for children aged 5-11 years and 11-18 years, respectively. Humoral and cellular immunogenicity was assessed at pre-dose 4, and at 1 and 6 months post-dose 4. Results Twenty-one children, with a median age of 14.0 years, were included for evaluation. A fourth dose of BNT162b2 elicited significant increases in humoral spike receptor-binding domain immunoglobulin G levels and T-cell responses. Antibody responses were significantly lower among kidney transplant recipients or children receiving calcineurin inhibitors than other CKD children at 1 month post-dose 4. Breakthrough COVID-19 occurred in three children after the fourth dose, and one was hospitalized. One child developed mild gross hematuria 1 day after the fourth dose, which spontaneously resolved. The overall safety profile was acceptable. Conclusions A fourth dose of BNT162b2 was immunogenic and safe in children with CKD.
Collapse
Affiliation(s)
- Jeffery C H Chan
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Eugene Yu-Hin Chan
- Paediatric Nephrology Centre, Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Hong Kong
| | | | - Daniel Leung
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Fanny Tsz-Wai Ho
- Paediatric Nephrology Centre, Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Hong Kong
| | - Pak-Chiu Tong
- Paediatric Nephrology Centre, Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Hong Kong
| | - Wai-Ming Lai
- Paediatric Nephrology Centre, Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Hong Kong
| | - Matthew H L Lee
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, Hong Kong
| | - Stella Chim
- Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, Hong Kong
| | - Leo C H Tsang
- School of Public Health, The University of Hong Kong, Hong Kong
| | - Tsz-Chun Kwan
- School of Public Health, The University of Hong Kong, Hong Kong
| | - Yin Celeste Cheuk
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Manni Wang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Howard H W Wong
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Amos M T Lee
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Wing Yan Li
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Sau Man Chan
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Issan Y S Tam
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Jennifer H Y Lam
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Kaiyue Zhang
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Wenwei Tu
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Malik Peiris
- School of Public Health, The University of Hong Kong, Hong Kong
- Centre for Immunology & Infection C2i, Hong Kong
| | - Jaime S Rosa Duque
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Yu Lung Lau
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong
| | - Alison Lap-Tak Ma
- Paediatric Nephrology Centre, Department of Paediatrics and Adolescent Medicine, Hong Kong Children's Hospital, Hong Kong
| |
Collapse
|
10
|
Mandal S, Chanu WP, Natarajaseenivasan K. Development of a multi-epitope vaccine candidate to combat SARS-CoV-2 and dengue virus co-infection through an immunoinformatic approach. Front Immunol 2025; 16:1442101. [PMID: 40079004 PMCID: PMC11897530 DOI: 10.3389/fimmu.2025.1442101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Background Although the SARS-CoV-2 and dengue viruses seriously endanger human health, there is presently no vaccine that can stop a person from contracting both viruses at the same time. In this study, four antigens from SARS-CoV-2 and dengue virus were tested for immunogenicity, antigenicity, allergenicity, and toxicity and chosen to predict dominant T- and B-cell epitopes. Methods For designing a multi-epitope vaccine, the sequences were retrieved, and using bioinformatics and immunoinformatics, the physicochemical and immunological properties, as well as secondary structures, of the vaccine were predicted and studied. Additionally, the three-dimensional structure was estimated, improved upon, and confirmed using bioinformatics methods before being docked with TLR-2 and TLR-4. Eight helper T-cell lymphocyte (HTL) epitopes, ten cytotoxic T-cell lymphocyte (CTL) epitopes, nine B-cell epitopes, and TLR agonists were used to create a new multi-epitope vaccine. Furthermore, according to the immunological stimulation hypothesis, the vaccine could stimulate T and B cells to create large quantities of Th1 cytokines and antibodies. Results The study indicates that the developed vaccine is a favorable vaccine candidate with antigenicity, immunogenicity, non-toxicity, and non-allergenicity properties. The vaccine construct was made up of 460 amino acids, had an MW of 49391.51 Da, a theoretical pI of 9.86, and the formula C2203H3433N643O618S18, a lipid index of 39.84, a GRAVY of -0.473, an aliphatic index of 63.80, and an instability index of 39.84, which classifies the protein to be stable. Conclusion The acquired data showed that both vaccine designs had a considerable chance of preventing the co-infection of SARS-CoV-2 and dengue virus and that they demonstrate good results following in-silico testing. Furthermore, the vaccine may be an effective strategy in preventing SARS-CoV-2 and dengue since it can cause noticeably high levels of Th1 cytokines and antibodies.
Collapse
Affiliation(s)
- Saurav Mandal
- Division of Metabolomics, Proteomics & Imaging facility, Regional Medical Research Centre, Indian Council of Medical Research (ICMR), Dibrugarh, Assam, India
| | - Waribam Pratibha Chanu
- Department of Applied Physics, School of Vocational Studies and Applied Sciences (SoVSAS), Gautam Buddha University, Greater Noida, Uttar Pradesh, India
| | - Kalimuthusamy Natarajaseenivasan
- Division of Metabolomics, Proteomics & Imaging facility, Regional Medical Research Centre, Indian Council of Medical Research (ICMR), Dibrugarh, Assam, India
| |
Collapse
|
11
|
Conlin K, Jenkin D, de Whalley P, Weckx LY, Folegatti PM, Bibi S, Lambe T, Aley PK, Pollard AJ, Voysey M, Costa Clemens SA. Predictors of severity of SARS-CoV-2 infections in Brazil: Post hoc analyses of a randomised controlled trial. Vaccine 2025; 45:126582. [PMID: 39675209 DOI: 10.1016/j.vaccine.2024.126582] [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: 08/16/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVES To identify demographic, clinical and immunological factors associated with adverse COVID-19 outcomes. METHODS A large randomised controlled trial of ChAdOx1 nCoV-19 was undertaken in Brazil. Participants were randomised 1:1 either to receive ChAdOx1 nCov-19 or to a control group. COVID-19 infections were confirmed by nucleic acid amplification test (NAAT) and classified using the WHO clinical progression scale. Anti-spike antibody responses and serum neutralising activity were measured 28 days after second vaccination in some participants. Exploratory analyses were conducted into factors associated with COVID-19 infection severity and hospitalisation, using logistic regression models adjusted for demographic and clinical factors. RESULTS 10,416 participants were enrolled; 1790 had NAAT-positive COVID-19 infection; 63 cases required hospitalisation. More severe infection was associated with greater body-mass index (BMI) (odds ratio [OR] = 1.06 [95 %CI: 1.01-1.10], p = 0.01) and diabetes (OR = 3.67 [1.59-8.07], p = 0.003). Hospitalisation risk increased with greater age (OR = 1.06 [1.03-1.08], p < 0.001) and BMI (OR = 1.10 [1.05-1.16], p < 0.001). More severe infection and hospitalisation risks increased >180 days after last vaccination. In the fully vaccinated subgroup (n = 841), only greater age predicted hospitalisation (OR = 1.07 [1.03-1.12], p < 0.001). Serological responses to two vaccine doses diminished with age. CONCLUSIONS Unvaccinated individuals with high BMI and diabetes risked more severe COVID-19 outcomes. Vaccination mitigated this risk. CLINICAL TRIAL REGISTRATION NUMBER NCT04536051.
Collapse
Affiliation(s)
- Kerry Conlin
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Daniel Jenkin
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Philip de Whalley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Lily Yin Weckx
- Department of Pediatrics, Universidade Federal de São Paulo, São Paulo, Brazil.
| | - Pedro M Folegatti
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, UK.
| | - Parvinder K Aley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Merryn Voysey
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Sue Ann Costa Clemens
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Institute of Global Health, University of Siena, Siena, Brazil.
| |
Collapse
|
12
|
Thiem VD, Anh DD, Ha VH, Van Thom N, Thang TC, Mateus J, Carreño JM, Raghunandan R, Huong NM, Mercer LD, Flores J, Escarrega EA, Raskin A, Thai DH, Van Be L, Sette A, Innis BL, Krammer F, Weiskopf D. Safety and immunogenicity of an inactivated recombinant Newcastle disease virus vaccine expressing SARS-CoV-2 spike: A randomised, comparator-controlled, phase 2 trial. Vaccine 2025; 44:126542. [PMID: 39615342 PMCID: PMC11672239 DOI: 10.1016/j.vaccine.2024.126542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 11/11/2024] [Accepted: 11/16/2024] [Indexed: 12/20/2024]
Abstract
Production of affordable coronavirus disease 2019 (COVID-19) vaccines in low- and lower-middle-income countries is needed. NDV-HXP-S is an inactivated egg-based recombinant Newcastle disease virus vaccine expressing the spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). A public sector manufacturer in Vietnam assessed the immunogenicity of NDV-HXP-S (COVIVAC) relative to an authorized vaccine. This phase 2 stage of a randomised, observer-blind, controlled, phase 1/2 trial was conducted at three community health centers in Thai Binh Province, Vietnam. Healthy males and non-pregnant females, 18 years of age and older, were eligible. Participants were randomised by age (18-59, ≥60 years) to receive one of three treatments by intramuscular injection twice, 28 days apart: COVIVAC at 3 μg or 6 μg, or AstraZeneca COVID-19 vaccine VAXZEVRIA™. Participants and personnel assessing outcomes were masked to treatment. The vaccine dose was selected based on Phase 1 results. A 6 μg dose was chosen to explore the immunogenicity gain over the 3-μg dose. The study's aim is to evaluate the safety and immunogenicity of COVIVAC at two dose levels compared to VAXZEVRIA, the most commonly used COVID-19 vaccine in Vietnam. The main outcome was the induction of 50% neutralising antibody titers against vaccine-homologous pseudotyped virus 14 days (day 43) and 6 months (day 197) after the second vaccination by age group. The primary immunogenicity and safety analyses included all participants who received one dose of the vaccine. ClinicalTrials.govNCT05940194. During August 10-23, 2021, 737 individuals were screened, and 374 were randomised (124-125 per group); all subjects received vaccine dose one and all but three received doses two four weeks later. Subjects 18-59 years of age achieved the following geometric mean titers of PNA 14 days after vaccine dose two: 153⋅28 (95 % CI 124·2-189⋅15) for COVIVAC 3 μg, 176⋅2 (95 % CI 141⋅45-220.27) for COVIVAC 6 μg, and 99⋅92(95 % CI 80.80-123⋅56) for VAXZEVRIA. Subjects ≥60 years of age also achieved potent geometric mean titers of PNA at the same timepoint: 183⋅57 (95 % CI 133.4-252⋅61) for COVIVAC 3 μg, 257⋅87 (95 % CI 181⋅6-367⋅18) for COVIVAC 6 μg, and 79⋅49(95 % CI 55⋅68-113⋅4) for VAXZEVRIA. On day 43, the geometric mean fold rise of 50 % neutralising antibody titers for subjects age 18-59 years was 31·20 (COVIVAC 3 μg N = 82, 95 % CI 25·14-38·74), 35·80 (COVIVAC 6 μg; N = 83, 95 % CI 29·03-44·15), 18·85 (VAXZEVRIA; N = 82, 95 % CI 15·10-23·54), and for subjects age ≥ 60 years was 37·27 (COVIVAC 3 μg; N = 42, 95 % CI 27·43-50·63), 50·10 (COVIVAC 6 μg; N = 40, 95 % CI 35·46-70·76), 16·11 (VAXZEVRIA; N = 40, 95 % CI 11·73-22·13). Among subjects seronegative for anti-S IgG at baseline, the day 43 geometric mean titer ratio of neutralising antibody (COVIVC 6 μg/VAXZEVRIA) was 1·77 (95 % CI 1·30-2·40) for subjects age 18-59 years and 3·24 (95 % CI 1·98-5·32) for subjects age ≥ 60 years. On day 197, the age-specific ratios were 1·11 (95 % CI 0·51-2·43) and 2·32 (0·69-7·85). Vaccines were well tolerated; reactogenicity was predominantly mild and transient. The percentage of subjects with unsolicited adverse events (AEs) during 28 days after vaccinations was similar among treatments (COVIVAC 3 μg 29·0 %, COVIVAC 6 μg 23·2 %, VAXZEVRIA 31·2 %); no vaccine-related AE was reported. Considering that induction of neutralising antibodies against SARS-CoV-2 has been correlated with the efficacy of COVID-19 vaccines, including VAXZEVRIA, our results suggest that vaccination with COVIVAC may afford clinical benefit matching or exceeding that of the VAXZEVRIA vaccine. ClinicalTrials.govNCT05940194.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Female
- Humans
- Male
- Middle Aged
- Young Adult
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- COVID-19/prevention & control
- COVID-19/immunology
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- COVID-19 Vaccines/adverse effects
- Immunogenicity, Vaccine
- Newcastle disease virus/immunology
- Newcastle disease virus/genetics
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/immunology
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
- Vaccines, Inactivated/adverse effects
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/adverse effects
- Vaccines, Synthetic/administration & dosage
- Vietnam
Collapse
Affiliation(s)
- Vu Dinh Thiem
- National Institute of Hygiene and Epidemiology, 1 Yersin Street, Hai Ba Trung District, Hanoi, Viet Nam
| | - Dang Duc Anh
- National Institute of Hygiene and Epidemiology, 1 Yersin Street, Hai Ba Trung District, Hanoi, Viet Nam
| | - Vu Hai Ha
- National Institute of Hygiene and Epidemiology, 1 Yersin Street, Hai Ba Trung District, Hanoi, Viet Nam
| | - Nguyen Van Thom
- Center for Disease Control, Thai Binh Province, 10 Hoàng Công Chất street, Quang Trung ward, Thai Binh, Viet Nam
| | - Tran Cong Thang
- PATH Vietnam, Hanoi Towers, 49 Hai Ba Trung Street, Hoan Kiem District, Hanoi, Viet Nam
| | - Jose Mateus
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Juan Manuel Carreño
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP) at Mount Sinai New York, New York, NY, USA
| | - Rama Raghunandan
- Center for Vaccine Innovation and Access, PATH, 2201 Westlake Avenue, Suite 200, Seattle, WA 98121, USA.
| | - Nguyen Mai Huong
- PATH Vietnam, Hanoi Towers, 49 Hai Ba Trung Street, Hoan Kiem District, Hanoi, Viet Nam
| | - Laina D Mercer
- Center for Vaccine Innovation and Access, PATH, 2201 Westlake Avenue, Suite 200, Seattle, WA 98121, USA
| | - Jorge Flores
- Center for Vaccine Innovation and Access, PATH, 2201 Westlake Avenue, Suite 200, Seattle, WA 98121, USA
| | - E Alexandar Escarrega
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
| | - Ariel Raskin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP) at Mount Sinai New York, New York, NY, USA
| | - Duong Huu Thai
- Institute of Vaccines and Medical Biologicals, 9 Pasteur, Xuong Huan, Nha Trang City, Khanh Hoa, Viet Nam
| | - Le Van Be
- Institute of Vaccines and Medical Biologicals, 9 Pasteur, Xuong Huan, Nha Trang City, Khanh Hoa, Viet Nam
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA; Department of Medicine, University of California, San Diego, (UCSD), La Jolla, CA, 92037, USA
| | - Bruce L Innis
- Center for Vaccine Innovation and Access, PATH, 2201 Westlake Avenue, Suite 200, Seattle, WA 98121, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai New York, New York, NY, USA; Center for Vaccine Research and Pandemic Preparedness (C-VaRPP) at Mount Sinai New York, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, NY, New York, USA.
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA; Department of Medicine, University of California, San Diego, (UCSD), La Jolla, CA, 92037, USA.
| |
Collapse
|
13
|
Riccò M, Cascio A, Costantino C, Corrado S, Zanella I, Giuri PG, Esposito S. Risk of Nipah Virus Seroprevalence in Healthcare Workers: A Systematic Review with Meta-Analysis. Viruses 2025; 17:81. [PMID: 39861870 PMCID: PMC11768436 DOI: 10.3390/v17010081] [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: 11/02/2024] [Revised: 12/22/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Nipah virus (NiV) is a zoonotic pathogen with the potential to cause human outbreaks with a high case fatality ratio. In this systematic review and meta-analysis, available evidence on NiV infections occurring in healthcare workers (HCWs) was collected and critically appraised. According to the PRISMA statement, four medical databases (PubMed, CINAHL, EMBASE, and Scopus) and the preprint repository medRixv were inquired through a specifically designed searching strategy. A total of 2593 entries were identified; of them, 16 studies were included in qualitative and quantitative analysis detailing the outcome of NiV infection on HCWs and estimates of seroprevalence among healthcare professions. All studies reported data from Asian countries: Malaysia, Singapore, Bangladesh, India (States of Kerala and Bengal), and Philippines. Seroprevalence was estimated from seven studies in 0.00% (95%CI 0.00 to 0.10) for IgM-class antibodies and 0.08% (95%CI 0.00 to 0.72) for IgG class-antibodies, but four of the sampled studies did not report any seropositive cases. A case fatality ratio of 73.52% (95%CI 34.01 to 99.74) was calculated from 10 studies. In conclusion, the present study shows that NiV may result in a possible occupational infection among HCWs involved in managing incident cases. As most NiV outbreaks occur in limited resources settings, it is reasonable that even basic preventive measures (i.e., mandatory use of PPE and appropriate isolation of incident cases with physical distancing) may be quite effective in avoiding the occurrence of new infections among HCWs.
Collapse
Affiliation(s)
- Matteo Riccò
- AUSL–IRCCS di Reggio Emilia, Servizio di Prevenzione e Sicurezza Negli Ambienti di Lavoro (SPSAL), Local Health Unit of Reggio Emilia, 42122 Reggio Emilia, Italy
| | - Antonio Cascio
- Infectious and Tropical Diseases Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, “G. D’Alessandro”, University of Palermo, AOUP P. Giaccone, 90127 Palermo, Italy;
| | - Claudio Costantino
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE) “G. D’Alessandro”, University of Palermo, 90127 Palermo, Italy;
| | - Silvia Corrado
- ASST Rhodense, Dipartimento della Donna e Area Materno-Infantile, UOC Pediatria, 20024 Milan, Italy;
| | - Ilaria Zanella
- AUSL–IRCCS di Reggio Emilia, Servizio di Prevenzione e Sicurezza Negli Ambienti di Lavoro (SPSAL), Local Health Unit of Reggio Emilia, 42122 Reggio Emilia, Italy
| | | | - Susanna Esposito
- Pediatric Clinic, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy;
| |
Collapse
|
14
|
Maine CJ, Miyake-Stoner SJ, Spasova DS, Picarda G, Chou AC, Brand ED, Olesiuk MD, Domingo CC, Little HJ, Goodman TT, Posy JL, Gonzalez J, Bayone TL, Sparks J, Gary EN, Xiang Z, Tursi NJ, Hojecki CE, Ertl HCJ, Weiner DB, Casmil IC, Blakney AK, Essink B, Somodevilla G, Wang NS, Geall AJ, Goldberg Z, Aliahmad P. Safety and immunogenicity of an optimized self-replicating RNA platform for low dose or single dose vaccine applications: a randomized, open label Phase I study in healthy volunteers. Nat Commun 2025; 16:456. [PMID: 39774967 PMCID: PMC11707033 DOI: 10.1038/s41467-025-55843-9] [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: 11/05/2024] [Accepted: 01/02/2025] [Indexed: 01/11/2025] Open
Abstract
Self-replicating RNA (srRNA) technology, in comparison to mRNA vaccines, has shown dose-sparing by approximately 10-fold and more durable immune responses. However, no improvements are observed in the adverse events profile. Here, we develop an srRNA vaccine platform with optimized non-coding regions and demonstrate immunogenicity and safety in preclinical and clinical development. Optimized srRNA vaccines generate protective immunity (according to the WHO defined thresholds) at doses up to 1,000,000-fold lower than mRNA in female mouse models of influenza and rabies. Clinically, safety and immunogenicity of RBI-4000, an srRNA vector encoding the rabies glycoprotein, was evaluated in a Phase I study (NCT06048770). RBI-4000 was able to elicit de novo protective immunity in the majority of healthy participants when administered at a dose of 0.1, 1, or 10 microgram (71%, 94%, 100%, respectively) in a prime-boost schedule. Similarly, we observe immunity above the WHO benchmark of protection following a single administration in most participants at both 1 and 10 microgram doses. There are no serious adverse events reported across all cohorts. These data establish the high therapeutic index of optimized srRNA vectors, demonstrating feasibility of both low dose and single dose approaches for vaccine applications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ebony N Gary
- The Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Zhi Xiang
- The Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Nicholas J Tursi
- The Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Casey E Hojecki
- The Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Hildegund C J Ertl
- The Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - David B Weiner
- The Vaccine & Immunotherapy Center, The Wistar Institute, Philadelphia, PA, USA
| | - Irafasha C Casmil
- Michael Smith Laboratories, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Anna K Blakney
- Michael Smith Laboratories, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | | | | | | | | | | | | |
Collapse
|
15
|
Rai S, Tripathi S. Comparative efficacy of leading COVID-19 vaccines: A network meta-analysis. Indian J Med Res 2025; 161:9-20. [PMID: 40036106 PMCID: PMC11878698 DOI: 10.25259/ijmr_750_2024] [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: 06/10/2024] [Accepted: 01/10/2025] [Indexed: 03/06/2025] Open
Abstract
In the fight against the COVID-19 virus, various vaccines using different technologies such as mRNA, viral vectors, protein subunits, and inactivated whole viruses have become primary defence strategies. This study aims to compare their effectiveness in controlling the spread of the pandemic. Using the comprehensive resources from three major databases-PubMed, EMBASE, and the Cochrane Library-we conducted an extensive literature review up to April 30, 2023. By employing a frequentist network meta-analysis, we analysed both direct and indirect estimates of vaccine efficacy, providing a clear comparison of the leading candidates in the global fight against COVID-19. Fifteen vaccines from 26 articles were used in our network meta-analysis. The statistically significant direct estimates were obtained by Spikevax [VE: 93.29 (91.31, 95.27); P<0.05], Pfizer BioNTech [VE: 92.07 (90.03, 94.12); P<0.05], Sputnik [VE: 91.60 (85.60, 97.60); P<0.05], Novavax [VE: 88.99 (83.55, 94.42); P<0.05], Sinovac [VE: 83.50 (65.40, 101.60); P<0.05], Covifenz [VE: 77.27 (68.48, 86.06); P<0.05], Zifivax [VE: 75.94 (70.86, 81.02); P<0.05], Covishield [VE: 72.34 (67.12, 77.56); P<0.05], S-Trimer [VE: 71.61 (56.23, 86.98); P<0.05], Covaxin [VE: 70.81 (65.33, 76.29); P<0.05], Soberna [VE: 69.70 (56.50, 82.90); P<0.05], Zydus Cadila [VE: 66.60 (47.60, 85.60); P<0.05], CVnCoV [VE: 63.70 (52.20, 75.20); P<0.05], Convidecia [VE: 57.50 (39.70, 75.30); P <0.05], and Jcovden [VE : 52.42 (47.28, 57.57); P<0.05]. Spikevax emerged triumphant with an unparalleled P score of 0.95, solidifying its status as a top ranking prevention tool against the COVID-19 in our investigation. Our analysis reveals a ranking of vaccine efficacy, with Spikevax emerging as the most effective, followed closely by Comirnaty, Sputnik, and others, collectively providing strong protection against the ongoing threat of COVID-19.
Collapse
Affiliation(s)
- Sanjay Rai
- Department of Centre of Community Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Shashank Tripathi
- Department of Biostatistics and Medical Informatics, University College of Medical Sciences and GTB Hospital, New Delhi, India
| |
Collapse
|
16
|
Kulkarni PS, Padmapriyadarsini C, Vekemans J, Bavdekar A, Gupta M, Kulkarni P, Garg B, Gogtay NJ, Tambe M, Lalwani S, Singh K, Munshi R, Meshram S, Selvavinayagam T, Pandey K, Bhimarasetty DM, Ramakrishnan S, Bhamare C, Dharmadhikari A, Budhawant C, Bonhomme CJ, Thakar M, Kurle SN, Kelly EJ, Gautam M, Gupta N, Panda S, Bhargava B, Poonawalla CS, Shaligram U, Kapse D, Gunale B. Seropersistence of SII-ChAdOx1 nCoV-19 (COVID-19 vaccine): 6-month follow-up of a randomized, controlled, observer-blind, phase 2/3 immuno-bridging study in Indian adults. Hum Vaccin Immunother 2024; 20:2304974. [PMID: 38512394 PMCID: PMC10962622 DOI: 10.1080/21645515.2024.2304974] [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: 08/24/2023] [Accepted: 01/10/2024] [Indexed: 03/23/2024] Open
Abstract
AZD1222 (ChAdOx1 nCoV-19) is a replication-deficient adenoviral vectored coronavirus disease-19 (COVID-19) vaccine that is manufactured as SII-ChAdOx1 nCoV-19 by the Serum Institute of India Pvt Ltd following technology transfer from Oxford University/AstraZeneca. The non-inferiority of SII-ChAdOx1 nCoV-19 with AZD1222 was previously demonstrated in an observer-blind, phase 2/3 immuno-bridging study (trial registration: CTRI/2020/08/027170). In this analysis of immunogenicity and safety data 6 months post first vaccination (Day 180), 1,601 participants were randomized 3:1 to SII-ChAdOx1 nCoV-19 or AZD1222 (immunogenicity/reactogenicity cohort n = 401) and 3:1 to SII-ChAdOx1 nCoV-19 or placebo (safety cohort n = 1,200). Immunogenicity was measured by anti-severe acute respiratory syndrome coronavirus 2 spike (anti-S) binding immunoglobulin G and neutralizing antibody (nAb) titers. A decline in anti-S titers was observed in both vaccine groups, albeit with a greater decline in SII-ChAdOx1 nCoV-19 vaccinees (geometric mean titer [GMT] ratio [95% confidence interval (CI) of SII-ChAdOx1 nCoV-19 to AZD1222]: 0.60 [0.41-0.87]). Consistent similar decreases in nAb titers were observed between vaccine groups (GMT ratio [95% CI]: 0.88 [0.44-1.73]). No cases of severe COVID-19 were reported following vaccination, while one case was observed in the placebo group. No causally related serious adverse events were reported through 180 days. No thromboembolic or autoimmune adverse events of special interest were reported. Collectively, these data illustrate that SII-ChAdOx1 nCoV-19 maintained a high level of immunogenicity 6 months post-vaccination. SII-ChAdOx1 nCoV-19 was safe and well tolerated.
Collapse
Affiliation(s)
| | | | - Johan Vekemans
- Formerly of: Clinical Development, Infection, Late-stage Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Madhu Gupta
- Department of Community Medicine and School of Public Health, Post Graduate Institute of Medical Education & Research, Chandigarh, India
| | - Praveen Kulkarni
- Department of Community Medicine, JSS Academy of Higher Education and Research, Mysore, India
| | - B.S. Garg
- Department of Community Medicine and Dr Sushila Nayar School of Public Health, Mahatma Gandhi Institute of Medical Sciences, Wardha, India
| | - Nithya J. Gogtay
- Department of Clinical Pharmacology, Seth G S Medical College & KEM Hospital, Mumbai, India
| | - Muralidhar Tambe
- Department of Community Medicine, B J Government Medical College and Sassoon General Hospitals, Pune, India
| | - Sanjay Lalwani
- Department of Pediatrics, Bharati Vidyapeeth Deemed University Medical College and Hospital, Pune, India
| | - Kiranjit Singh
- Jehangir Clinical Development Centre Pvt Ltd, Pune, India
| | - Renuka Munshi
- Department of Clinical Pharmacology, TN Medical College & BYL Nair Hospital, Mumbai, India
| | - Sushant Meshram
- Department of Pulmonary Medicine, Government Medical College, Nagpur, India
| | | | - Krishna Pandey
- Rajendra Memorial Research Institute of Medical Sciences, Patna, India
| | | | - S.R. Ramakrishnan
- Department of Clinical Research, Sri Ramchandra Institute of Higher Education and Research, Chennai, India
| | | | | | | | - Cyrille J. Bonhomme
- Laboratory Services, Vaccines Sciences Lab, Clinical Research, PPD, Part of Thermo Fisher Scientific, Richmond, VA, USA
| | | | | | - Elizabeth J. Kelly
- Formerly of: Translational Medicine, Vaccines and Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Follmann D, Wang X, Baden LR, El Sahly HM, Essink B, Gilbert P, Janes HE, Kelley CF, Berman MA, Frank I, Chu E, Deng W, Priddy F, Dixit A, Tomassini JE, Das R, Miller J, Zhou H. Who to Boost When: The Effect of Age and Dosing Interval on Delta and Omicron COVID-19 Incidence in the Open-label Phase of the COVE Trial. Open Forum Infect Dis 2024; 11:ofae689. [PMID: 39679349 PMCID: PMC11639572 DOI: 10.1093/ofid/ofae689] [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: 06/20/2024] [Accepted: 11/22/2024] [Indexed: 12/17/2024] Open
Abstract
Background To help inform COVID-19 vaccination recommendations, we evaluated the impact of age and dosing interval on clinical benefit of a third dose of mRNA-1273. Methods Approximately 17 000 participants from the phase 3 Coronavirus Efficacy trial who previously received 2 doses of 100 µg mRNA-1273 were evaluated for COVID-19 between September 2021 and April 2022 during uptake of a third booster dose of 50 µg of mRNA-1273. Cox models assessed booster relative efficacy of a third dose. Results Initial booster relative efficacy against Delta COVID-19 was 83% (95% confidence interval, 60-93) 14 days postdose and 83% (67-91) 60 days later. Initial booster efficacy against Omicron COVID-19 was 56% (44-65) at 14 days postdose and 4% (-27 to 28) 120 days later. For those aged ≥65 years, initial booster efficacy against Omicron COVID-19 was 86% (69-93) compared with 50% (36-61) for those <65 years. Placebo crossover to 2 doses of mRNA-1273 induced a median 5-month difference from the second to third dose between the original randomized arms. Postboost, the mRNA-1273 arm had a 24% (16%, 32%) lower risk of Omicron COVID-19 compared to the placebo-mRNA-1273 arm. Modeling predicted a 41% postboost reduction in Omicron COVID-19 for a 15- versus 7-month interval between the second and third doses. Conclusions Boosting reduced Delta COVID-19 risk by 83% through 2 months and reduced Omicron COVID-19 risk by 56% but declined by 4 months. A 15- versus 7-month dosing interval predicted a 41% postboost reduction in Omicron COVID-19 but increased preboost risk. Primary Funding Source The National Institutes of Health/National Institute of Allergy and Infectious Diseases. Registration for the COVE Trial. ClinicalTrials.gov ID# NCT04470427.
Collapse
Affiliation(s)
- Dean Follmann
- National Institute of Allergy and Infectious Diseases, Biostatistics Research Branch, Bethesda, Maryland, USA
| | | | - Lindsey R Baden
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hana M El Sahly
- Departments of Molecular Virology and Microbiology and Medicine, Baylor College of Medicine, Houston, Texas, USA
| | | | - Peter Gilbert
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Holly E Janes
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Colleen F Kelley
- Division of Infectious Diseases, Emory University School of Medicine Grady Health System, Atlanta, Georgia, USA
| | - Megan A Berman
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, USA
| | - Ian Frank
- Deparment of Infectious Diseases, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eric Chu
- National Institute of Allergy and Infectious Diseases, Biostatistics Research Branch, Bethesda, Maryland, USA
| | | | | | - Avika Dixit
- Moderna, Inc., Cambridge, Massachusetts, USA
| | | | | | | | | |
Collapse
|
18
|
Xu Y, Kong X, Huang W, Liang Z, Huang J, Li Y, Zhang N, Liu D, Guo W, Mei J. Efficacy of the COVID-19 vaccination in patients with asymptomatic or mild illness during the Omicron epidemic in Guangzhou: a multi-centre retrospective cohort study. Ann Med 2024; 56:2307504. [PMID: 38465636 PMCID: PMC10930140 DOI: 10.1080/07853890.2024.2307504] [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: 08/13/2023] [Accepted: 01/15/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Despite the widespread administration of coronavirus disease 2019 (COVID-19) vaccines, the impact on patients with asymptomatic to mild illness remains unclear. Here, we aimed to assess the efficacy of various vaccine doses and types on the duration of isolation duration and discharge rates, the viral shedding duration, and negative rates in asymptomatic to mild COVID-19 patients. METHODS We included adult patients at the Fangcang isolation centres in Pazhou or Yongning between November and December 2022. We analysed data on basic demographics, admission details, laboratory indicators and vaccination information. RESULTS A total of 6560 infected patients were included (3584 from Pazhou and 2976 from Yongning). Of these, 90.6% received inactivated vaccines, 3.66% received recombinant SARS-CoV-2 spike protein subunit vaccines and 0.91% received adenovirus vaccines. Among the 6173 vaccinated individuals, 71.9% received a booster dose. By day 9, the isolation rate reached 50% among vaccinated patients. On day 7.5, the positive rate among vaccinated individuals reached 50%. CONCLUSIONS Full vaccination was effective, with heterologous vaccines showing greater efficacy than inactivated vaccines alone. However, there was no significant difference in the vaccine protective effect 12 months after vaccination.
Collapse
Affiliation(s)
- Yuanda Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Xuetao Kong
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
- Department of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Weiqing Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Zijing Liang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Jinkun Huang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Yimin Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Nuofu Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Dan Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Wenwei Guo
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Jiang Mei
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
19
|
González-Parra G, Mahmud MS, Kadelka C. Learning from the COVID-19 pandemic: A systematic review of mathematical vaccine prioritization models. Infect Dis Model 2024; 9:1057-1080. [PMID: 38988830 PMCID: PMC11233876 DOI: 10.1016/j.idm.2024.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/26/2024] [Accepted: 05/10/2024] [Indexed: 07/12/2024] Open
Abstract
As the world becomes ever more connected, the chance of pandemics increases as well. The recent COVID-19 pandemic and the concurrent global mass vaccine roll-out provides an ideal setting to learn from and refine our understanding of infectious disease models for better future preparedness. In this review, we systematically analyze and categorize mathematical models that have been developed to design optimal vaccine prioritization strategies of an initially limited vaccine. As older individuals are disproportionately affected by COVID-19, the focus is on models that take age explicitly into account. The lower mobility and activity level of older individuals gives rise to non-trivial trade-offs. Secondary research questions concern the optimal time interval between vaccine doses and spatial vaccine distribution. This review showcases the effect of various modeling assumptions on model outcomes. A solid understanding of these relationships yields better infectious disease models and thus public health decisions during the next pandemic.
Collapse
Affiliation(s)
- Gilberto González-Parra
- Instituto de Matemática Multidisciplinar, Universitat Politècnica de València, València, Spain
- Department of Mathematics, New Mexico Tech, 801 Leroy Place, Socorro, 87801, NM, USA
| | - Md Shahriar Mahmud
- Department of Mathematics, Iowa State University, 411 Morrill Rd, Ames, 50011, IA, USA
| | - Claus Kadelka
- Department of Mathematics, Iowa State University, 411 Morrill Rd, Ames, 50011, IA, USA
| |
Collapse
|
20
|
Zhou P, Qiu T, Wang X, Yang X, Shi H, Zhu C, Dai W, Xing M, Zhang X, Xu J, Zhou D. One HA stalk topping multiple heads as a novel influenza vaccine. Emerg Microbes Infect 2024; 13:2290838. [PMID: 38044872 PMCID: PMC10810646 DOI: 10.1080/22221751.2023.2290838] [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: 09/04/2023] [Accepted: 11/29/2023] [Indexed: 12/05/2023]
Abstract
Classic chimeric hemagglutinin (cHA) was designed to induce immune responses against the conserved stalk domain of HA. However, it is unclear whether combining more than one HA head domain onto one stalk domain is immunogenic and further induce immune responses against influenza viruses. Here, we constructed numerous novel cHAs comprising two or three fuzed head domains from different subtypes grafted onto one stalk domain, designated as cH1-H3, cH1-H7, cH1-H3-H7, and cH1-H7-H3. The three-dimensional structures of these novel cHAs were modelled using bioinformatics simulations. Structural analysis showed that the intact neutralizing epitopes were exposed in cH1-H7 and were predicted to be immunogenic. The immunogenicity of the cHAs constructs was evaluated in mice using a chimpanzee adenoviral vector (AdC68) vaccine platform. The results demonstrated that cH1-H7 expressed by AdC68 (AdC68-cH1-H7) induced the production of high levels of binding antibodies, neutralizing antibodies, and hemagglutinin inhibition antibodies against homologous pandemic H1N1, drifted seasonal H1N1, and H7N9 virus. Moreover, vaccinated mice were fully protected from a lethal challenge with the aforementioned influenza viruses. Hence, cH1-H7 cHAs with potent immunogenicity might be a potential novel vaccine to provide protection against different subtypes of influenza virus.
Collapse
Affiliation(s)
- Ping Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People’s Republic of China
- Chinese Academy of Sciences, Institut Pasteur of Shanghai, Shanghai, People’s Republic of China
| | - Tianyi Qiu
- Institute of Clinical Science, ZhongShan Hospital, Fudan University, Shanghai, People’s Republic of China
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, People’s Republic of China
| | - Xiang Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People’s Republic of China
| | - Xi Yang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People’s Republic of China
| | - Hongyang Shi
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Caihong Zhu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People’s Republic of China
| | - Weiqian Dai
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Man Xing
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People’s Republic of China
| | - Jianqing Xu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People’s Republic of China
| | - Dongming Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, People’s Republic of China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, People’s Republic of China
| |
Collapse
|
21
|
Musango L, Mandrosovololona V, Randriatsarafara FM, Ranarison VM, Kirigia JM, Ratsimbasoa CA. The present value of human life losses associated with COVID-19 and likely productivity losses averted through COVID-19 vaccination in Madagascar. BMC Public Health 2024; 24:3296. [PMID: 39604940 PMCID: PMC11600903 DOI: 10.1186/s12889-024-20786-1] [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: 10/02/2023] [Accepted: 11/18/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND As of 3 March 2023, Madagascar had reported 1,422 deaths from COVID-19. Up to now, there hasn't been a study to estimate the Total Present Value of Human Life lostTPVHL MADAGASCAR , productivity losses, and potential productivity losses averted through COVID-19 vaccination for use in advocacy. The study reported in this paper aimed to fill these information gaps. METHODS The Human Capital Model (HCM) was used to estimate theTPVHL MADAGASCAR , which is the sum of the discounted value of human life losses among individuals in seven different age groups. The Present Value of Human Life for each age groupPVHL i was calculated by multiplying the discount factor, the undiscounted years of life, the non-health gross domestic product (GDP) per capita, and the number of COVID-19 deaths in that age group. To test the robustness of the results, the HCM was rerun five times, assuming (i) a 5% discount rate, (ii) a 10% discount rate, (iii) Africa's highest average life expectancy at birth of 78.76 years, (iv) the world's highest life expectancy of 88.17 years, (v) projected excess COVID-19 mortality of 11,418.66 deaths as of 3 March 2023 in Madagascar, and assuming different levels of vaccine coverage: 100%, 70%, 60.93%, and 8.266%. RESULTS The 1,422 human lives lost due to COVID-19 had aTPVHL MADAGASCAR of Int$ 46,331,412; and an average of Int$ 32,582 per human life. Re-estimation of the HCM, using (i) discount rates of 5% and 10% reducedTPVHL MADAGASCAR by 23% and 53%, respectively; (ii) average life expectancies of 78.76 years and 88.17 years increasedTPVHL MADAGASCAR by 23.7% and 39.5%, respectively; (iii) projected excess COVID-19 mortality of 11,418.66 augmentedTPVHL MADAGASCAR by 703%. Furthermore, it is estimated that vaccinating 70% of the target population could potentially save the country Int$ 1.1 billion, equivalent to 1.94% of the GDP. CONCLUSIONS The COVID-19 pandemic has resulted in significant health and productivity losses for Madagascar. Optimizing COVID-19 vaccination coverage for the target population could substantially reduce these losses.
Collapse
Affiliation(s)
- Laurent Musango
- World Health Organization, Country Office, BP 362 Maison Commune des Nations-Unies, Enceinte Galaxy, Andraharo, Antananarivo, Madagascar.
| | - Vatsiharizandry Mandrosovololona
- World Health Organization, Country Office, BP 362 Maison Commune des Nations-Unies, Enceinte Galaxy, Andraharo, Antananarivo, Madagascar
| | | | - Volahanta Malala Ranarison
- World Health Organization, Country Office, BP 362 Maison Commune des Nations-Unies, Enceinte Galaxy, Andraharo, Antananarivo, Madagascar
| | | | - Claude Arsène Ratsimbasoa
- Faculte de Medecine de Fianarantsoa, Centre National d'Application de La Reherche Pharmaceutique, Antananarivo, Madagascar
| |
Collapse
|
22
|
Berber E, Ross TM. Factors Predicting COVID-19 Vaccine Effectiveness and Longevity of Humoral Immune Responses. Vaccines (Basel) 2024; 12:1284. [PMID: 39591186 PMCID: PMC11598945 DOI: 10.3390/vaccines12111284] [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: 10/16/2024] [Revised: 11/09/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, prompted global efforts to develop vaccines to control the disease. Various vaccines, including mRNA (BNT162b2, mRNA-1273), adenoviral vector (ChAdOx1, Ad26.COV2.S), and inactivated virus platforms (BBIBP-CorV, CoronaVac), elicit high-titer, protective antibodies against the virus, but long-term antibody durability and effectiveness vary. The objective of this study is to elucidate the factors that influence vaccine effectiveness (VE) and the longevity of humoral immune responses to COVID-19 vaccines through a review of the relevant literature, including clinical and real-world studies. Here, we discuss the humoral immune response to different COVID-19 vaccines and identify factors influencing VE and antibody longevity. Despite initial robust immune responses, vaccine-induced immunity wanes over time, particularly with the emergence of variants, such as Delta and Omicron, that exhibit immune escape mechanisms. Additionally, the durability of the humoral immune responses elicited by different vaccine platforms, along with the identification of essential determinants of long-term protection-like pre-existing immunity, booster doses, hybrid immunity, and demographic factors-are critical for protecting against severe COVID-19. Booster vaccinations substantially restore neutralizing antibody levels, especially against immune-evasive variants, while individuals with hybrid immunity have a more durable and potent immune response. Importantly, comorbidities such as diabetes, cardiovascular disease, chronic kidney disease, and cancer significantly reduce the magnitude and longevity of vaccine-induced protection. Immunocompromised individuals, particularly those undergoing chemotherapy and those with hematologic malignancies, have diminished humoral responses and benefit disproportionately from booster vaccinations. Age and sex also influence immune responses, with older adults experiencing accelerated antibody decline and females generally exhibiting stronger humoral responses compared to males. Understanding the variables affecting immune protection is crucial to improving vaccine strategies and predicting VE and protection against COVID-19.
Collapse
Affiliation(s)
- Engin Berber
- Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
| | - Ted M. Ross
- Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA;
- Florida Research and Innovation Center, Cleveland Clinic, Florida, FL 34986, USA
| |
Collapse
|
23
|
Huang Y, Wang W, Liu Y, Wang Z, Cao B. COVID-19 vaccine updates for people under different conditions. SCIENCE CHINA. LIFE SCIENCES 2024; 67:2323-2343. [PMID: 39083202 DOI: 10.1007/s11427-024-2643-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 06/06/2024] [Indexed: 10/22/2024]
Abstract
SARS-CoV-2 has caused global waves of infection since December 2019 and continues to persist today. The emergence of SARS-CoV-2 variants with strong immune evasion capabilities has compromised the effectiveness of existing vaccines against breakthrough infections. Therefore, it is important to determine the best utilization strategies for different demographic groups given the variety of vaccine options available. In this review, we will discuss the protective efficacy of vaccines during different stages of the epidemic and emphasize the importance of timely updates to target prevalent variants, which can significantly improve immune protection. While it is recognized that vaccine effectiveness may be lower in certain populations such as the elderly, individuals with chronic comorbidities (e.g., diabetes with poor blood glucose control, those on maintenance dialysis), or those who are immunocompromised compared to the general population, administering multiple doses can result in a strong protective immune response that outweighs potential risks. However, caution should be exercised when considering vaccines that might trigger an intense immune response in populations prone to inflammatory flare or other complications. In conclusion, individuals with special conditions require enhanced and more effective immunization strategies to prevent infection or reinfection, as well as to avoid the potential development of long COVID.
Collapse
Affiliation(s)
- Yijiao Huang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
- School of Basic Medical Sciences, Tsinghua Medicine, Tsinghua University, Beijing, 100084, China
- Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Weiyang Wang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yan Liu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
- Department of Infectious Disease, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, 264000, China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Bin Cao
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Beijing, 100029, China.
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.
- Tsinghua University-Peking University Joint Centre for Life Sciences, Tsinghua University, Beijing, 100084, China.
- Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Changping Laboratory, Beijing, 102200, China.
- Department of Respiratory Medicine, Capital Medical University, Beijing, 100069, China.
- New Cornerstone Science Laboratory, China-Japan Friendship Hospital, Beijing, 100029, China.
| |
Collapse
|
24
|
Paranilam J, Arcioni F, Franco A, Lai KZH, Brown J, Kimball-Carroll S. Delphi Panel Consensus Statement Generation: COVID-19 Vaccination Recommendations for Immunocompromised Populations in the European Union. Infect Dis Ther 2024; 13:2227-2253. [PMID: 39382830 PMCID: PMC11499477 DOI: 10.1007/s40121-024-01051-9] [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: 06/17/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
INTRODUCTION The coronavirus disease 2019 (COVID-19) pandemic has caused unprecedented pressure on healthcare systems globally. The lack of quality guidelines on the management of COVID-19 in rheumatologic disease, renal disease, hematological malignancy, and solid organ transplant recipients has resulted in a wide variation in clinical practice. METHODS Using a Delphi process, a panel of 16 key opinion leaders developed clinical practice statements regarding vaccine recommendations in areas where standards are absent or limited. Agreement among practicing physicians with consensus statements was also assessed via an online physician survey. The strength of the consensus was determined by the following rating system: a strong rating was defined as all four key opinion leaders (KOLs) rating the statement ≥ 8, a moderate rating was defined as three out of four KOLs rating the statement ≥ 8, and no consensus was defined as less than three out of four KOLs provided a rating of ≤ 8. Specialists voted on agreement with each consensus statement for their disease area using the same ten-point scoring system. RESULTS Key opinion leaders in rheumatology, nephrology, and hematology achieved consensuses for all nine statements pertaining to the primary and booster series with transplant physicians reaching consensus on eight of nine statements. Experts agreed that COVID-19 vaccines are safe, effective, and well tolerated by patients with rheumatological conditions, renal disease, hematologic malignancy, and recipients of solid organ transplants. The Delphi process yielded strong to moderate suggestions for the use of COVID-19 messenger ribonucleic acid (mRNA) vaccines and the necessity of the COVID-19 booster for the immunocompromised population. The expert panel had mixed feelings concerning the measurement of antibody titers, higher-dose mRNA vaccines, and the development of disease-specific COVID-19 guidance. CONCLUSIONS These results confirmed the necessity of COVID-19 vaccines and boosters in immunocompromised patients with rheumatologic disease, renal disease, hematological malignancy, and solid organ transplant recipients. Statements where consensus was not achieved were due to absent or limited evidence.
Collapse
Affiliation(s)
| | - Francesco Arcioni
- Pediatric Onco-Hematology with Bone Marrow Transplantation, Azienda Ospedaliera Di Perugia, Piazza Menghini 1, 06132, Perugia, Italy
| | - Antonio Franco
- Department of Nephrology, Hospital Dr Balmis, 03010, Alicante, Spain
| | | | | | | |
Collapse
|
25
|
Kuddus MA, Mohiuddin M, Paul AK, Rahman A. Insights from qualitative and bifurcation analysis of COVID-19 vaccination model in Bangladesh. PLoS One 2024; 19:e0312780. [PMID: 39485754 PMCID: PMC11530044 DOI: 10.1371/journal.pone.0312780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 10/13/2024] [Indexed: 11/03/2024] Open
Abstract
The unprecedented global impact of the 2019 coronavirus disease (COVID-19) has necessitated a comprehensive understanding of its transmission dynamics and control measures. In this study, we present a detailed analysis of a COVID-19 vaccination model tailored to the context of Bangladesh, incorporating dual-dose vaccination strategies. By employing qualitative and bifurcation analysis techniques, we investigate the equilibrium points, effective reproduction number (R0), and critical thresholds that influence the prevalence and control of COVID-19 in the region. Our findings reveal insights into the effectiveness of vaccination programs and provide a framework for developing targeted control plans. Through a rigorous examination of model parameters and sensitivity analysis, we identify key factors driving COVID-19 transmission dynamics, emphasizing the significance of vaccination rates and other critical parameters. The validation of our model against real-world data underscores its utility in informing evidence-based decision-making for managing the COVID-19 pandemic in Bangladesh and beyond.
Collapse
Affiliation(s)
- Md Abdul Kuddus
- Department of Mathematics, University of Rajshahi, Rajshahi, Bangladesh
| | - M. Mohiuddin
- Department of Mathematics, Comilla University, Cumilla, Bangladesh
| | - Anip Kumar Paul
- Department of Mathematics, University of Rajshahi, Rajshahi, Bangladesh
| | - Azizur Rahman
- School of Computing, Mathematics and Engineering, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| |
Collapse
|
26
|
Fryer HA, Geers D, Gommers L, Zaeck LM, Tan NH, Jones-Freeman B, Goorhuis A, Postma DF, Visser LG, Hogarth PM, Koopmans MPG, GeurtsvanKessel CH, O'Hehir RE, van der Kuy PHM, de Vries RD, van Zelm MC. Fourth dose bivalent COVID-19 vaccines outperform monovalent boosters in eliciting cross-reactive memory B cells to Omicron subvariants. J Infect 2024; 89:106246. [PMID: 39127451 DOI: 10.1016/j.jinf.2024.106246] [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: 05/15/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Bivalent COVID-19 vaccines comprising ancestral Wuhan-Hu-1 (WH1) and the Omicron BA.1 or BA.5 subvariant elicit enhanced serum antibody responses to emerging Omicron subvariants. Here, we characterized the RBD-specific memory B cell (Bmem) response following a fourth dose with a BA.1 or BA.5 bivalent vaccine, in direct comparison with a WH1 monovalent fourth dose. Healthcare workers previously immunized with mRNA or adenoviral vector monovalent vaccines were sampled before and one month after a fourth dose with a monovalent or a BA.1 or BA.5 bivalent vaccine. Serum neutralizing antibodies (NAb) were quantified, as well as RBD-specific Bmem with an in-depth spectral flow cytometry panel including recombinant RBD proteins of the WH1, BA.1, BA.5, BQ.1.1, and XBB.1.5 variants. Both bivalent vaccines elicited higher NAb titers against Omicron subvariants compared to the monovalent vaccine. Following either vaccine type, recipients had slightly increased WH1 RBD-specific Bmem numbers. Both bivalent vaccines significantly increased WH1 RBD-specific Bmem binding of all Omicron subvariants tested by flow cytometry, while recognition of Omicron subvariants was not enhanced following monovalent vaccination. IgG1+ Bmem dominated the response, with substantial IgG4+ Bmem only detected in recipients of an mRNA vaccine for their primary dose. Thus, Omicron-based bivalent vaccines can significantly boost NAb and Bmem specific for ancestral WH1 and Omicron variants and improve recognition of descendent subvariants by pre-existing, WH1-specific Bmem beyond that of a monovalent vaccine. This provides new insights into the capacity of variant-based mRNA booster vaccines to improve immune memory against emerging SARS-CoV-2 variants and potentially protect against severe disease. ONE-SENTENCE SUMMARY: Omicron BA.1 and BA.5 bivalent COVID-19 boosters, used as a fourth dose, increase RBD-specific Bmem cross-recognition of Omicron subvariants, both those encoded by the vaccines and antigenically distinct subvariants, further than a monovalent booster.
Collapse
Affiliation(s)
- Holly A Fryer
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Daryl Geers
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Lennert Gommers
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Luca M Zaeck
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Ngoc H Tan
- Dept. Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Bernadette Jones-Freeman
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Abraham Goorhuis
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Centers, Amsterdam, the Netherlands; Infection and Immunity, Amsterdam Public Health, University of Amsterdam, Amsterdam, the Netherlands
| | - Douwe F Postma
- Department of Internal Medicine and Infectious Diseases, University Medical Center Groningen, Groningen, the Netherlands
| | - Leo G Visser
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - P Mark Hogarth
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Immune Therapies Group, Burnet Institute, Melbourne, Victoria, Australia
| | - Marion P G Koopmans
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | | | - Robyn E O'Hehir
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, Victoria, Australia
| | - P Hugo M van der Kuy
- Dept. Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Rory D de Vries
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Menno C van Zelm
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, Victoria, Australia; Dept. Immunology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
27
|
Patel A, Mathias A, Baghel A, Kumari A, Kujur S. Clinical characterization, molecular and genomic sequencing analysis of SARS-Cov-2 during second wave at Raigarh, Chhattisgarh, India. Bioinformation 2024; 20:1059-1064. [PMID: 39917233 PMCID: PMC11795479 DOI: 10.6026/9732063002001059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 02/09/2025] Open
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-Cov-2) has been changing continuously. This study was conducted to evaluate clinical characteristics, Molecular analysis & Genomic sequencing of SARS-Cov-2 during second wave in Raigarh district, Chhattisgarh, India. This study evaluated 13402 breakthrough cases of COVID -19. The laboratory obtained the nasopharyngeal/oropharyngeal swabs (NPS/OPS) of SARS-CoV-2 patients who tested positive by real-time RT-PCR, together with clinical and demographic information. Next generation sequencing (NGS) was used to sequence these clinical specimens in order to identify nucleotide changes in the SARS-CoV-2 genome from these strains. In the study population, variants of concern (VOCs) and other variations were looked for. Clinical severity was mild in 47.05% patients with mutational variants; while 52.94% patient's clinical severity was moderate. Delta (B.1.617.2) was the most common VOC detected. Among non VOC variants, AY.4 and AY.12 variants were most commonly detected. Envelope (E) gene and RNA-dependent RNA polymerase (RdRp) mutation were most commonly observed.
Collapse
Affiliation(s)
- Anubha Patel
- Department of Microbiology, Government Medical College, Mahasamund, Chhattisgarh, India
| | - Anuniti Mathias
- Department of Microbiology, BRLSABVM Medical College, Rajnandgaon, Chhattisgarh, India
| | - Ashish Baghel
- Department of Community Medicine, Government Medical College, Mahasamund, Chhattisgarh, India
| | - Ankita Kumari
- Department of Microbiology, LSLAMG Medical College, Raigarh, Chhattisgarh, India
| | - Swati Kujur
- Scientific Officer (Biology), State Forensic Science Laboratory, Raipur, Chhattisgarh, India
| |
Collapse
|
28
|
Kim Y, Han K, Kim JH. Retinal Vascular Occlusions After COVID-19 Vaccination in South Korea: A Nation-Wide Population-Based Study. Ophthalmic Epidemiol 2024:1-9. [PMID: 39288331 DOI: 10.1080/09286586.2024.2399345] [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: 02/18/2024] [Revised: 08/09/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE To investigate the association between the retinal vascular occlusion and vaccination against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). METHODS This nationwide population-based cohort study included 2,742,065 individuals aged ≥ 20 years who were vaccinated against SARS-CoV-2 from March 1, 2021, to December 31, 2021, and unvaccinated individuals matched at a ratio of approximately 1:10 by gender and age, all without a history of retinal vascular occlusion. The occurrence of retinal vascular occlusion was observed up to 60 days after the 1st vaccination date in the vaccination group, while 60 days from January 1, 2021, in the non-vaccination group. The risk of developing retinal vascular occlusion was compared between vaccinated and unvaccinated subjects. Risks were also compared among the different types of vaccines. RESULTS Vaccination lowered the risk of retinal vascular occlusion, with an odds ratio (OR) of 0.80 (95% confidence interval (CI), 0.64-0.99; p = 0.039). For individuals aged < 40 years, the vaccination lowered the risk of retinal vascular occlusion occurrence significantly compared with those over the age of 40 (OR, 0.35 for age 20-39, 0.83 for age 40-64, 0.81 for age ≥ 65; P for interaction = 0.028). There was a significant difference in the ORs for retinal vascular occlusion among the four vaccine types (p < 0.001). CONCLUSIONS SARS-CoV-2 vaccination did not increase the risk of retinal vascular occlusion. However, the risk levels differed depending on the type of vaccine used. Considering the ongoing evolution of SARS-CoV-2 vaccines, it is imperative to conduct additional assessments of the recently introduced vaccines.
Collapse
Affiliation(s)
- Yeji Kim
- Department of Ophthalmology, Kim's Eye Hospital, Seoul, South Korea
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, South Korea
| | - Jae Hui Kim
- Department of Ophthalmology, Kim's Eye Hospital, Seoul, South Korea
| |
Collapse
|
29
|
Wang S, Li W, Wang Z, Yang W, Li E, Xia X, Yan F, Chiu S. Emerging and reemerging infectious diseases: global trends and new strategies for their prevention and control. Signal Transduct Target Ther 2024; 9:223. [PMID: 39256346 PMCID: PMC11412324 DOI: 10.1038/s41392-024-01917-x] [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: 02/22/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 09/12/2024] Open
Abstract
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhenshan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Wanying Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
- Department of Laboratory Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
30
|
Mahroum N, Habra M, Alrifaai MA, Shoenfeld Y. Antiphospholipid syndrome in the era of COVID-19 - Two sides of a coin. Autoimmun Rev 2024; 23:103543. [PMID: 38604461 DOI: 10.1016/j.autrev.2024.103543] [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: 01/31/2024] [Revised: 04/08/2024] [Accepted: 04/08/2024] [Indexed: 04/13/2024]
Abstract
In addition to the respiratory symptoms associated with COVID-19, the disease has consistently been linked to many autoimmune diseases such as systemic lupus erythematous and antiphospholipid syndrome (APS). APS in particular was of paramount significance due to its devastating clinical sequela. In fact, the hypercoagulable state seen in patients with acute COVID-19 and the critical role of anticoagulant treatment in affected individuals shed light on the possible relatedness between APS and COVID-19. Moreover, the role of autoimmunity in the assumed association is not less important especially with the accumulated data available regarding the autoimmunity-triggering effect of SARS-CoV-2 infection. This is furtherly strengthened at the time patients with COVID-19 manifested antiphospholipid antibodies of different types following infection. Additionally, the severe form of the APS spectrum, catastrophic APS (CAPS), was shown to have overlapping characteristics with severe COVID-19 such as cytokine storm and multi-organ failure. Interestingly, COVID vaccine-induced autoimmune phenomena described in the medical literature have pointed to an association with APS. Whether the antiphospholipid antibodies were present or de novo, COVID vaccine-induced vascular thrombosis in certain individuals necessitates further investigations regarding the possible mechanisms involved. In our current paper, we aimed to focus on the associations mentioned, their implications, importance, and consequences.
Collapse
Affiliation(s)
- Naim Mahroum
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey.
| | - Mona Habra
- International School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | | | - Yehuda Shoenfeld
- Zabludowicz Center for autoimmune diseases, Sheba Medical Center, Ramat-Gan, Israel; Reichman University, Herzliya, Israel
| |
Collapse
|
31
|
Zhou P, Watt J, Mai J, Cao H, Li Z, Chen Z, Duan R, Quan Y, Gingras AC, Rini JM, Hu J, Liu J. Intranasal HD-Ad-FS vaccine induces systemic and airway mucosal immunities against SARS-CoV-2 and systemic immunity against SARS-CoV-2 variants in mice and hamsters. Front Immunol 2024; 15:1430928. [PMID: 39281669 PMCID: PMC11392758 DOI: 10.3389/fimmu.2024.1430928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/08/2024] [Indexed: 09/18/2024] Open
Abstract
The outbreak of coronavirus disease 19 (COVID-19) has highlighted the demand for vaccines that are safe and effective in inducing systemic and airway mucosal immunity against the aerosol transmission of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this study, we developed a novel helper-dependent adenoviral vector-based COVID-19 mucosal vaccine encoding a full-length SARS-CoV-2 spike protein (HD-Ad-FS). Through intranasal immunization (single-dose and prime-boost regimens), we demonstrated that the HD-Ad-FS was immunogenic and elicited potent systemic and airway mucosal protection in BALB/c mice, transgenic ACE2 (hACE2) mice, and hamsters. We detected high titers of neutralizing antibodies (NAbs) in sera and bronchoalveolar lavages (BALs) in the vaccinated animals. High levels of spike-specific secretory IgA (sIgA) and IgG were induced in the airway of the vaccinated animals. The single-dose HD-Ad-FS elicited a strong immune response and protected animals from SARS-CoV-2 infection. In addition, the prime-boost vaccination induced cross-reactive serum NAbs against variants of concern (VOCs; Beta, Delta, and Omicron). After challenge, VOC infectious viral particles were at undetectable or minimal levels in the lower airway. Our findings highlight the potential of airway delivery of HD-Ad-FS as a safe and effective vaccine platform for generating mucosal protection against SARS-CoV-2 and its VOCs.
Collapse
MESH Headings
- Animals
- SARS-CoV-2/immunology
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
- COVID-19/prevention & control
- COVID-19/immunology
- Immunity, Mucosal
- Mice
- Administration, Intranasal
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Mice, Inbred BALB C
- Cricetinae
- Female
- Humans
- Mice, Transgenic
- Adenoviridae/genetics
- Adenoviridae/immunology
- Angiotensin-Converting Enzyme 2/immunology
- Angiotensin-Converting Enzyme 2/genetics
- Angiotensin-Converting Enzyme 2/metabolism
- Mesocricetus
Collapse
Affiliation(s)
- Peter Zhou
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jacqueline Watt
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Juntao Mai
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Huibi Cao
- Translational Medicine Program, Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Zhijie Li
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ziyan Chen
- Translational Medicine Program, Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Rongqi Duan
- Translational Medicine Program, Hospital for Sick Children Research Institute, Toronto, ON, Canada
| | - Ying Quan
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Anne-Claude Gingras
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - James M Rini
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jim Hu
- Translational Medicine Program, Hospital for Sick Children Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jun Liu
- Department of Molecular Genetics, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
32
|
Baziboroun M, Hosseinzadeh S, Gholinia H, Sadeghi F, Yahyapour Y. COVID-19 infection after vaccination. CASPIAN JOURNAL OF INTERNAL MEDICINE 2024; 15:644-650. [PMID: 39359433 PMCID: PMC11444105 DOI: 10.22088/cjim.15.4.644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 11/18/2023] [Indexed: 10/04/2024]
Abstract
Background Although vaccination is the most effective and specific approach for prevention of infectious diseases, but in a small percentage of vaccinated person's breakthrough infections can occur. This study aimed to determine the effectiveness of different common coronavirus vaccines in this area. Methods 109 COVID-19 vaccinated patients were enrolled, with different types of vaccines (Sinopharm, AstraZeneca, Sputnic, Bharath, CovIran Barkat and Pasto-CoV) and time of administration in 2021 in Babol, Iran. Patients after 14 days of administration of the final dose of corona vaccines with positive COVID-19 RT-PCR test entered to study. Patients' data such as RT-PCR, type of vaccine, age, sex and outcome were collected using electronic medical records. Results 47 patients were not fully vaccinated, 62 had two vaccine doses and 51 were fully vaccinated and considered to a breakthrough infection. Although, most of the patients with SARS-CoV-2 infection were either mild (n=18 [16.56%]), or moderate (n=86 [78.9%]), 5 (4.6%) patients had severe or critical illness, of whom 3 admitted in intensive care unit, 3 intubated, and 4 died. The average age of the participants with COVID-19 infections was 61.23 ± 19.91 years. Conclusion Based on our results, the COVID-19 breakthrough occurring with two doses of current vaccines were mild and moderate.
Collapse
Affiliation(s)
- Mana Baziboroun
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Sayareh Hosseinzadeh
- Part of Infectious Control, Ayatollah Rouhani Hospital, Babol University of Medical Sciences, Babol, Iran
| | - Hemmat Gholinia
- Social Determinants of Health Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Farzin Sadeghi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Yousef Yahyapour
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
33
|
Morciglio A, Zia RKP, Hyman JM, Jiang Y. Understanding the oscillations of an epidemic due to vaccine hesitancy. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:6829-6846. [PMID: 39483095 DOI: 10.3934/mbe.2024299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Vaccine hesitancy threatens to reverse the progress in tackling vaccine-preventable diseases. We used an $ SIS $ model with a game theory model for vaccination and parameters from the COVID-19 pandemic to study how vaccine hesitancy impacts epidemic dynamics. The system showed three asymptotic behaviors: total rejection of vaccinations, complete acceptance, and oscillations. With increasing fear of infection, stable endemic states become periodic oscillations. Our results suggest that managing fear of infection relative to vaccination is vital to successful mass vaccinations.
Collapse
Affiliation(s)
- Anthony Morciglio
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA 30303, USA
| | - R K P Zia
- Center for Soft Matter and Biological Physics, Department of Physics, Virginia Tech, Blacksburg, VA 24061, USA
- Department of Physics, University of Houston, Houston, Texas 77204, USA
| | - James M Hyman
- Department of Mathematics, Tulane University, New Orleans, LA 70118, USA
| | - Yi Jiang
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
34
|
Shoemaker K, Soboleva K, Branche A, Shankaran S, Theodore DA, Bari M, Ezeh V, Green J, Kelly E, Lan D, Olsson U, Saminathan S, Shankar NK, Villegas B, Villafana T, Falsey AR, Sobieszczyk ME. Long-Term Safety and Immunogenicity of AZD1222 (ChAdOx1 nCoV-19): 2-Year Follow-Up from a Phase 3 Study. Vaccines (Basel) 2024; 12:883. [PMID: 39204009 PMCID: PMC11359581 DOI: 10.3390/vaccines12080883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
A better understanding of the long-term safety, efficacy, and immunogenicity of COVID-19 vaccines is needed. This phase 3, randomized, placebo-controlled study for AZD1222 (ChAdOx1 nCoV-19) primary-series vaccination enrolled 32,450 participants in the USA, Chile, and Peru between August 2020 and January 2021 (NCT04516746). Endpoints included the 2-year follow-up assessment of safety, efficacy, and immunogenicity. After 2 years, no emergent safety signals were observed for AZD1222, and no cases of thrombotic thrombocytopenia syndrome were reported. The assessment of anti-SARS-CoV-2 nucleocapsid antibody titers confirmed the durability of AZD1222 efficacy for up to 6 months, after which infection rates in the AZD1222 group increased over time. Despite this, all-cause and COVID-19-related mortality remained low through the study end, potentially reflecting the post-Omicron decoupling of SARS-CoV-2 infection rates and severe COVID-19 outcomes. Geometric mean titers were elevated for anti-SARS-CoV-2 neutralizing antibodies at the 1-year study visit and the anti-spike antibodies were elevated at year 2, providing further evidence of increasing SARS-CoV-2 infections over long-term follow-up. Overall, this 2-year follow-up of the AZD1222 phase 3 study confirms that the long-term safety profile remains consistent with previous findings and supports the continued need for COVID-19 booster vaccinations due to waning efficacy and humoral immunity.
Collapse
Affiliation(s)
- Kathryn Shoemaker
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA; (K.S.); (D.L.)
| | - Karina Soboleva
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA; (K.S.); (V.E.)
| | - Angela Branche
- Division of Infectious Diseases, Department of Medicine, University of Rochester, Rochester, NY 14627, USA;
| | - Shivanjali Shankaran
- Division of Infectious Diseases, Rush University Medical Center, Chicago, IL 60612, USA;
| | - Deborah A. Theodore
- Division of Infectious Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, New York-Presbyterian/Columbia University Irving Medical Center, New York, NY 10032, USA; (D.A.T.)
| | - Muhammad Bari
- Formerly Patient Safety, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK;
| | - Victor Ezeh
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA; (K.S.); (V.E.)
| | - Justin Green
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - Elizabeth Kelly
- Formerly Translational Medicine, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA;
| | - Dongmei Lan
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA; (K.S.); (D.L.)
| | - Urban Olsson
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, 431 83 Gothenburg, Sweden;
| | - Senthilkumar Saminathan
- Patient Safety, Chief Medical Office, R&D, AstraZeneca, Bangalore 560045, India; (S.S.); (N.K.S.)
| | - Nirmal Kumar Shankar
- Patient Safety, Chief Medical Office, R&D, AstraZeneca, Bangalore 560045, India; (S.S.); (N.K.S.)
| | - Berta Villegas
- Clinical Operations, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Mississauga, ON L4Y 1M4, Canada;
| | - Tonya Villafana
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD 20878, USA; (K.S.); (V.E.)
| | - Ann R. Falsey
- Department of Medicine, Infectious Diseases, University of Rochester School of Medicine and Dentistry, Rochester, New York, NY 14642, USA;
- Infectious Disease, Rochester Regional Health, Rochester, New York, NY 14617, USA
| | - Magdalena E. Sobieszczyk
- Division of Infectious Diseases, Department of Medicine, Vagelos College of Physicians and Surgeons, New York-Presbyterian/Columbia University Irving Medical Center, New York, NY 10032, USA; (D.A.T.)
| |
Collapse
|
35
|
Yamaguchi D, Shimizu R, Kubota R. Development of a SARS-CoV-2 viral dynamic model for patients with COVID-19 based on the amount of viral RNA and viral titer. CPT Pharmacometrics Syst Pharmacol 2024; 13:1354-1365. [PMID: 38783551 PMCID: PMC11330184 DOI: 10.1002/psp4.13164] [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: 12/27/2023] [Revised: 04/17/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
The target-cell limited model, which is one of the mathematical modeling approaches providing a quantitative understanding of viral dynamics, has been applied to describe viral RNA profiles of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in previous studies. However, these models have been developed mainly using patient data from the early phase of the pandemic. Furthermore, no reports focused on the profiles of the viral titer. In this study, the dynamics of both viral RNA and viral titer were characterized using data reflecting the current clinical situation in which the Omicron variant has become epidemic and vaccines for SARS-CoV-2 have become available. Consecutive data for 5212 viral RNA levels and 5216 viral titers were obtained from 720 patients with coronavirus disease 2019 (COVID-19) in a phase II/III study for ensitrelvir. Our model assumed that productively infected cells would produce only infectious viruses, which could be transformed into non-infectious viruses, and has been used to describe the dynamics of both viral RNA levels and viral titer. The time from infection to symptom onset (tinf) of unvaccinated patients was estimated to be 3.0 days, which was shorter than that of the vaccinated patients. The immune-related parameter as a power function for the vaccinated patients was 1.1 times stronger than that for the unvaccinated patients. Our model allows the prediction of the viral dynamics in patients with COVID-19 from the time of infection to symptom onset. Vaccination status was identified as a factor influencing tinf and the immune function.
Collapse
Affiliation(s)
- Daichi Yamaguchi
- Clinical Pharmacology & PharmacokineticsShionogi & Co., Ltd.OsakaJapan
| | - Ryosuke Shimizu
- Clinical Pharmacology & PharmacokineticsShionogi & Co., Ltd.OsakaJapan
| | - Ryuji Kubota
- Clinical Pharmacology & PharmacokineticsShionogi & Co., Ltd.OsakaJapan
| |
Collapse
|
36
|
Costa Clemens SA, Jepson B, Bhorat QE, Ahmad A, Akhund T, Aley PK, Bansal H, Bibi S, Kelly EJ, Khan M, Lambe T, Lombaard JJ, Matthews S, Pipolo Milan E, Olsson U, Ramasamy MN, Moura de Oliveira Paiva MS, Seegobin S, Shoemaker K, Szylak A, Villafana T, Pollard AJ, Green JA. Immunogenicity and safety of beta variant COVID-19 vaccine AZD2816 and AZD1222 (ChAdOx1 nCoV-19) as primary-series vaccination for previously unvaccinated adults in Brazil, South Africa, Poland, and the UK: a randomised, partly double-blinded, phase 2/3 non-inferiority immunobridging study. THE LANCET. MICROBE 2024; 5:100863. [PMID: 38878794 DOI: 10.1016/s2666-5247(24)00078-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 02/12/2024] [Accepted: 03/12/2024] [Indexed: 08/10/2024]
Abstract
BACKGROUND AZD2816 is a variant-adapted COVID-19 vaccine that expresses the full-length SARS-CoV-2 beta variant spike protein but is otherwise similar to AZD1222 (ChAdOx1 nCoV-19). This study aimed to evaluate the safety and immunogenicity of AZD1222 or AZD2816 (or both) primary-series vaccination in a cohort of adult participants who were previously unvaccinated. METHODS In this phase 2/3, randomised, multinational, active-controlled, non-inferiority, immunobridging study, adult participants previously unvaccinated for COVID-19 were enrolled at 16 study sites in Brazil, South Africa, Poland, and the UK. Participants were stratified by age, sex, and comorbidity and randomly assigned 5:5:5:2 to receive a primary series of AZD1222 (AZD1222 group), AZD2816 (AZD2816 [4-week] group), or AZD1222-AZD2816 (AZD1222-AZD2816 group) at 4-week dosing intervals, or AZD2816 at a 12-week interval (AZD2816 [12-week] group) and evaluated for safety and immunogenicity through 180 days after dose 2. Primary outcomes were safety (rates of solicited adverse events occurring during 7 days and unsolicited adverse events occurring during 28 days after each dose) and immunogenicity (non-inferiority of pseudovirus neutralising antibody geometric mean titre [GMT], GMT ratio margin of 0·67, and seroresponse rate, rate difference margin of -10%, recorded 28 days after dose 2 with AZD2816 [4-week interval] against beta vs AZD1222 against ancestral SARS-CoV-2) in participants who were seronegative at baseline. This trial is registered with ClinicalTrials.gov, NCT04973449, and is completed. FINDINGS Between July 7 and Nov 12, 2021, 1449 participants were assigned to the AZD1222 group (n=413), the AZD2816 (4-week) group (n=415), the AZD1222-AZD2816 group (n=412), and the AZD2816 (12-week) group (n=209). Ten (2·6%) of 378 participants who were seronegative at baseline in the AZD1222 group, nine (2·4%) of 379 in the AZD2816 (4-week) group, eight (2·1%) of 380 in the AZD1222-AZD2816 group, and 11 (5·8%) of 191 in the AZD2816 (12-week) group had vaccine-related unsolicited adverse events. Serious adverse events were recorded in one (0·3%) participant in the AZD1222 group, one (0·3%) in the AZD2816 (4-week) group, two (0·5%) in the AZD1222-AZD2816 group, and none in the AZD2816 (12-week) group. Co-primary immunogenicity endpoints were met: neutralising antibody GMT (ratio 1·19 [95% CI 1·08-1·32]; lower bound greater than 0·67) and seroresponse rate (difference 1·7% [-3·1 to 6·5]; lower bound greater than -10%) at 28 days after dose 2 were non-inferior in the AZD2816 (4-week) group against beta versus in the AZD1222 group against ancestral SARS-CoV-2. Seroresponse rates were highest with AZD2816 against beta (12-week interval 94·3% [95% CI 89·4-97·3]; 4-week interval 85·7% [81·5-89·2]) and with AZD1222 (84·6% [80·3-88·2]) against ancestral SARS-CoV-2. INTERPRETATION Primary series of AZD1222 and AZD2816 were well tolerated, with no emergent safety concerns. Both vaccines elicited robust immunogenicity against beta and ancestral SARS-CoV-2 with greater responses demonstrated when testing against SARS-CoV-2 strains that matched those targeted by the respective vaccine. These findings demonstrate the continued importance of ancestral COVID-19 vaccines in protecting against severe COVID-19 and highlight the feasibility of using the ChAdOx1 platform to develop COVID-19 vaccines against future SARS-CoV-2 variants. FUNDING AstraZeneca.
Collapse
Affiliation(s)
- Sue Ann Costa Clemens
- Department of Paediatrics, University of Oxford, Oxford, UK; Institute for Global Health, Siena University, Siena, Italy
| | - Brett Jepson
- Biometrics, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Qasim E Bhorat
- Soweto Clinical Trials Centre, Soweto, Gauteng, South Africa
| | - Abdullahi Ahmad
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Tauseefullah Akhund
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Parvinder K Aley
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Himanshu Bansal
- Biometrics, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Sagida Bibi
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Elizabeth J Kelly
- Formerly Translational Medicine, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Mark Khan
- Clinical Development, BioPharmaceuticals R&D, AstraZeneca, Mississauga, ON, Canada
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; Chinese Academy of Medical Science, Oxford Institute, University of Oxford, Oxford, UK
| | | | - Sam Matthews
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Eveline Pipolo Milan
- Centro de Estudos e Pesquisas em Moléstias Infec, Centro de Pesquisas Clínicas de Natal, Natal, Rio Grande do Norte, Brazil
| | - Urban Olsson
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Maheshi N Ramasamy
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Seth Seegobin
- Biometrics, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Kathryn Shoemaker
- Biometrics, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA; Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Ameena Szylak
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Mississauga, ON, Canada
| | - Tonya Villafana
- Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK; NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Justin A Green
- Clinical Development, Vaccines & Immune Therapies, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
| |
Collapse
|
37
|
Chaudhary S, Aggarwal M, Kumari P, Vishwas G, Meshram GG, Dhaka R, Garg M. Safety Surveillance of Covishield Vaccine-Associated Adverse Events During the COVID-19 Pandemic: A Retrospective Longitudinal Study. Cureus 2024; 16:e67257. [PMID: 39301361 PMCID: PMC11411232 DOI: 10.7759/cureus.67257] [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: 08/19/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Adverse events following immunization (AEFI) must be reported and assessed to promote patient safety. This longitudinal study examined the nature and severity of adverse events reported after Covishield (Serum Institute of India, Pune, India) vaccine administration to North Indians in a tertiary care hospital. METHOD A retrospective evaluation of adverse drug reactions (ADRs) reported after Covishield vaccine administration in our hospital over 18 months was conducted. The assessment was carried out to analyze the pattern of ADRs reported by individuals receiving the Covishield vaccine from January 2021 to June 2022. Data such as age, gender, category, dose administered, type of ADR, duration of the event, medical history, and outcome of the reactions were collected. Each reported adverse event was assessed individually. Causality was determined using the WHO-UMC causality assessment scale. The data were analyzed and are expressed as mean ± standard deviation and percentage. RESULTS A total of 14,590 individuals were vaccinated at our study center from January 2021 to June 2022. During this period, 146 AEFIs (1.0%) were reported at our ADR monitoring center, Employees' State Insurance Corporation Medical College and Hospital (ESIC MCH), Faridabad, India. The majority of AEFIs were systemic, were reported after the first dose, and had an onset within 12 hours after vaccination. Fever, injection site pain, drowsiness, headache, vomiting, swelling, tenderness, and body aches were the most commonly reported adverse effects. No significant relationships were observed between the administered vaccine dose and sex, severity, duration of the event, or outcome. However, the incidence of adverse events was greater with the first vaccine dose than with the second dose. The possibility of serious or fatal adverse events was lowest in the general population and higher in the elderly with comorbidities. CONCLUSION The data suggest that the Covishield vaccine had mild to moderate adverse effects on the study population. This pharmacovigilance study will complement safety data and aid in the benefit-risk analysis of adverse effects associated with the Covishield vaccine. Additionally, healthcare professionals should be encouraged to conduct further safety studies by establishing robust vaccine safety monitoring systems in hospitals. Continuing medical education and workshops should also be conducted to educate healthcare workers about active surveillance.
Collapse
Affiliation(s)
- Shilpa Chaudhary
- Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, IND
- Pharmacology/Pharmacovigilance, Employees' State Insurance Corporation (ESIC) Medical College and Hospital, Faridabad, IND
| | - Monica Aggarwal
- Pharmacology, Employees' State Insurance Corporation (ESIC) Medical College and Hospital, Faridabad, IND
| | - Puja Kumari
- Pharmacology, Employees' State Insurance Corporation (ESIC) Medical College and Hospital, Faridabad, IND
| | - Gopal Vishwas
- Clinical Medicine, Indian Council of Medical Research - Rajendra Memorial Research Institute of Medical Sciences (ICMR-RMRIMS), Patna, IND
| | | | - Rohit Dhaka
- Community Medicine, Employees' State Insurance Corporation (ESIC) Medical College and Hospital, Faridabad, IND
| | - Minakshi Garg
- Drug Regulatory Affairs, Delhi Pharmaceutical Sciences and Research University, New Delhi, IND
| |
Collapse
|
38
|
Choi EML, Kasonia K, Kavunga-Membo H, Mukadi-Bamuleka D, Soumah A, Mossoko Z, Edwards T, Tetsa-Tata D, Makarimi R, Toure O, Mambula G, Brindle H, Camacho A, Connor NE, Mukadi P, McLean C, Keshinro B, Gaddah A, Robinson C, Luhn K, Foster J, Roberts CH, Johnson JE, Imbault N, Bausch DG, Grais RF, Watson-Jones D, Muyembe-Tamfum JJ. Immunogenicity of an Extended Dose Interval for the Ad26.ZEBOV, MVA-BN-Filo Ebola Vaccine Regimen in Adults and Children in the Democratic Republic of the Congo. Vaccines (Basel) 2024; 12:828. [PMID: 39203955 PMCID: PMC11359010 DOI: 10.3390/vaccines12080828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/21/2024] [Accepted: 07/18/2024] [Indexed: 09/03/2024] Open
Abstract
During the 2018-2020 Ebola virus disease outbreak in Democratic Republic of the Congo, a phase 3 trial of the Ad26.ZEBOV, MVA-BN-Filo Ebola vaccine (DRC-EB-001) commenced in Goma, with participants being offered the two-dose regimen given 56 days apart. Suspension of trial activities in 2020 due to the COVID-19 pandemic led to some participants receiving a late dose 2 outside the planned interval. Blood samples were collected from adults, adolescents, and children prior to their delayed dose 2 vaccination and 21 days after, and tested for IgG binding antibodies against Ebola virus glycoprotein using the Filovirus Animal Nonclinical Group (FANG) ELISA. Results from 133 participants showed a median two-dose interval of 9.3 months. The pre-dose 2 antibody geometric mean concentration (GMC) was 217 ELISA Units (EU)/mL (95% CI 157; 301) in adults, 378 EU/mL (281; 510) in adolescents, and 558 EU/mL (471; 661) in children. At 21 days post-dose 2, the GMC increased to 22,194 EU/mL (16,726; 29,449) in adults, 37,896 EU/mL (29,985; 47,893) in adolescents, and 34,652 EU/mL (27,906; 43,028) in children. Participants receiving a delayed dose 2 had a higher GMC at 21 days post-dose 2 than those who received a standard 56-day regimen in other African trials, but similar to those who received the regimen with an extended interval.
Collapse
Affiliation(s)
- Edward Man-Lik Choi
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK; (K.K.); (D.T.-T.); (H.B.); (N.E.C.); (C.h.R.); (D.G.B.); (D.W.-J.)
| | - Kambale Kasonia
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK; (K.K.); (D.T.-T.); (H.B.); (N.E.C.); (C.h.R.); (D.G.B.); (D.W.-J.)
| | - Hugo Kavunga-Membo
- Institut National de Recherche Biomédicale, Kinshasa P.O. Box 1192, Democratic Republic of the Congo; (H.K.-M.); (D.M.-B.); (Z.M.); (P.M.); (J.J.M.-T.)
| | - Daniel Mukadi-Bamuleka
- Institut National de Recherche Biomédicale, Kinshasa P.O. Box 1192, Democratic Republic of the Congo; (H.K.-M.); (D.M.-B.); (Z.M.); (P.M.); (J.J.M.-T.)
| | - Aboubacar Soumah
- Epicentre, 75019 Paris, France; (A.S.); (R.M.); (O.T.); (G.M.); (A.C.); (R.F.G.)
| | - Zephyrin Mossoko
- Institut National de Recherche Biomédicale, Kinshasa P.O. Box 1192, Democratic Republic of the Congo; (H.K.-M.); (D.M.-B.); (Z.M.); (P.M.); (J.J.M.-T.)
| | - Tansy Edwards
- MRC International Statistics and Epidemiology Group, Faculty of Epidemiology and Population Health, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK;
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki 852-8523, Japan
| | - Darius Tetsa-Tata
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK; (K.K.); (D.T.-T.); (H.B.); (N.E.C.); (C.h.R.); (D.G.B.); (D.W.-J.)
| | - Rockyath Makarimi
- Epicentre, 75019 Paris, France; (A.S.); (R.M.); (O.T.); (G.M.); (A.C.); (R.F.G.)
| | - Oumar Toure
- Epicentre, 75019 Paris, France; (A.S.); (R.M.); (O.T.); (G.M.); (A.C.); (R.F.G.)
| | - Grace Mambula
- Epicentre, 75019 Paris, France; (A.S.); (R.M.); (O.T.); (G.M.); (A.C.); (R.F.G.)
| | - Hannah Brindle
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK; (K.K.); (D.T.-T.); (H.B.); (N.E.C.); (C.h.R.); (D.G.B.); (D.W.-J.)
| | - Anton Camacho
- Epicentre, 75019 Paris, France; (A.S.); (R.M.); (O.T.); (G.M.); (A.C.); (R.F.G.)
| | - Nicholas E. Connor
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK; (K.K.); (D.T.-T.); (H.B.); (N.E.C.); (C.h.R.); (D.G.B.); (D.W.-J.)
| | - Pierre Mukadi
- Institut National de Recherche Biomédicale, Kinshasa P.O. Box 1192, Democratic Republic of the Congo; (H.K.-M.); (D.M.-B.); (Z.M.); (P.M.); (J.J.M.-T.)
| | - Chelsea McLean
- Janssen Vaccines and Prevention B.V., 2333 CN Leiden, The Netherlands; (C.M.); (B.K.); (C.R.); (K.L.)
| | - Babajide Keshinro
- Janssen Vaccines and Prevention B.V., 2333 CN Leiden, The Netherlands; (C.M.); (B.K.); (C.R.); (K.L.)
| | | | - Cynthia Robinson
- Janssen Vaccines and Prevention B.V., 2333 CN Leiden, The Netherlands; (C.M.); (B.K.); (C.R.); (K.L.)
| | - Kerstin Luhn
- Janssen Vaccines and Prevention B.V., 2333 CN Leiden, The Netherlands; (C.M.); (B.K.); (C.R.); (K.L.)
| | - Julie Foster
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK; (K.K.); (D.T.-T.); (H.B.); (N.E.C.); (C.h.R.); (D.G.B.); (D.W.-J.)
| | - Chrissy h. Roberts
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK; (K.K.); (D.T.-T.); (H.B.); (N.E.C.); (C.h.R.); (D.G.B.); (D.W.-J.)
| | | | - Nathalie Imbault
- Coalition for Epidemic Preparedness Innovations, 0191 Oslo, Norway;
| | - Daniel G. Bausch
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK; (K.K.); (D.T.-T.); (H.B.); (N.E.C.); (C.h.R.); (D.G.B.); (D.W.-J.)
- FIND, 1218 Geneva, Switzerland
| | - Rebecca F. Grais
- Epicentre, 75019 Paris, France; (A.S.); (R.M.); (O.T.); (G.M.); (A.C.); (R.F.G.)
| | - Deborah Watson-Jones
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK; (K.K.); (D.T.-T.); (H.B.); (N.E.C.); (C.h.R.); (D.G.B.); (D.W.-J.)
- Mwanza Intervention Trials Unit, National Institute for Medical Research, Mwanza P.O. Box 11936, Tanzania
| | - Jean Jacques Muyembe-Tamfum
- Institut National de Recherche Biomédicale, Kinshasa P.O. Box 1192, Democratic Republic of the Congo; (H.K.-M.); (D.M.-B.); (Z.M.); (P.M.); (J.J.M.-T.)
| |
Collapse
|
39
|
Tobias J, Steinberger P, Wilkinson J, Klais G, Kundi M, Wiedermann U. SARS-CoV-2 Vaccines: The Advantage of Mucosal Vaccine Delivery and Local Immunity. Vaccines (Basel) 2024; 12:795. [PMID: 39066432 PMCID: PMC11281395 DOI: 10.3390/vaccines12070795] [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: 06/12/2024] [Revised: 07/10/2024] [Accepted: 07/15/2024] [Indexed: 07/28/2024] Open
Abstract
Immunity against respiratory pathogens is often short-term, and, consequently, there is an unmet need for the effective prevention of such infections. One such infectious disease is coronavirus disease 19 (COVID-19), which is caused by the novel Beta coronavirus SARS-CoV-2 that emerged around the end of 2019. The World Health Organization declared the illness a pandemic on 11 March 2020, and since then it has killed or sickened millions of people globally. The development of COVID-19 systemic vaccines, which impressively led to a significant reduction in disease severity, hospitalization, and mortality, contained the pandemic's expansion. However, these vaccines have not been able to stop the virus from spreading because of the restricted development of mucosal immunity. As a result, breakthrough infections have frequently occurred, and new strains of the virus have been emerging. Furthermore, SARS-CoV-2 will likely continue to circulate and, like the influenza virus, co-exist with humans. The upper respiratory tract and nasal cavity are the primary sites of SARS-CoV-2 infection and, thus, a mucosal/nasal vaccination to induce a mucosal response and stop the virus' transmission is warranted. In this review, we present the status of the systemic vaccines, both the approved mucosal vaccines and those under evaluation in clinical trials. Furthermore, we present our approach of a B-cell peptide-based vaccination applied by a prime-boost schedule to elicit both systemic and mucosal immunity.
Collapse
Affiliation(s)
- Joshua Tobias
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Peter Steinberger
- Division of Immune Receptors and T Cell Activation, Institute of Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Joy Wilkinson
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Gloria Klais
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Michael Kundi
- Department of Environmental Health, Center for Public Health, Medical University of Vienna, 1090 Vienna, Austria;
| | - Ursula Wiedermann
- Institute of Specific Prophylaxis and Tropical Medicine, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
40
|
Edwards KR, Malhi H, Schmidt K, Davis AR, Homad LJ, Warner NL, Chhan CB, Scharffenberger SC, Gaffney K, Hinkley T, Potchen NB, Wang JY, Price J, McElrath MJ, Olson J, King NP, Lund JM, Moodie Z, Erasmus JH, McGuire AT. A gH/gL-encoding replicon vaccine elicits neutralizing antibodies that protect humanized mice against EBV challenge. NPJ Vaccines 2024; 9:120. [PMID: 38926438 PMCID: PMC11208421 DOI: 10.1038/s41541-024-00907-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Epstein-Barr virus (EBV) is associated with several malignancies, neurodegenerative disorders and is the causative agent of infectious mononucleosis. A vaccine that prevents EBV-driven morbidity and mortality remains an unmet need. EBV is orally transmitted, infecting both B cells and epithelial cells. Several virally encoded proteins are involved in entry. The gH/gL glycoprotein complex is essential for infectivity irrespective of cell type, while gp42 is essential for infection of B cells. gp350 promotes viral attachment by binding to CD21 or CD35 and is the most abundant glycoprotein on the virion. gH/gL, gp42 and gp350, are known targets of neutralizing antibodies and therefore relevant immunogens for vaccine development. Here, we developed and optimized the delivery of several alphavirus-derived replicon RNA (repRNA) vaccine candidates encoding gH/gL, gH/gL/gp42 or gp350 delivered by a cationic nanocarrier termed LION™. The lead candidate, encoding full-length gH/gL, elicited high titers of neutralizing antibodies that persisted for at least 8 months and a vaccine-specific CD8+ T cell response. Transfer of vaccine-elicited IgG protected humanized mice from EBV-driven tumor formation and death following high-dose viral challenge. These data demonstrate that LION/repRNA-gH/gL is an ideal candidate vaccine for preventing EBV infection and/or related malignancies in humans.
Collapse
Affiliation(s)
- Kristina R Edwards
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Harman Malhi
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Karina Schmidt
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Amelia R Davis
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Leah J Homad
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Crystal B Chhan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Samuel C Scharffenberger
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | | | - Nicole B Potchen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Jing Yang Wang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jason Price
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - James Olson
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA, USA
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jennifer M Lund
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Zoe Moodie
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Andrew T McGuire
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
41
|
Ison MG, Papi A, Athan E, Feldman RG, Langley JM, Lee DG, Leroux-Roels I, Martinon-Torres F, Schwarz TF, van Zyl-Smit RN, Verheust C, Dezutter N, Gruselle O, Fissette L, David MP, Kostanyan L, Hulstrøm V, Olivier A, Van der Wielen M, Descamps D. Efficacy and Safety of Respiratory Syncytial Virus (RSV) Prefusion F Protein Vaccine (RSVPreF3 OA) in Older Adults Over 2 RSV Seasons. Clin Infect Dis 2024; 78:1732-1744. [PMID: 38253338 PMCID: PMC11175669 DOI: 10.1093/cid/ciae010] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/15/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND The adjuvanted RSV prefusion F protein-based vaccine (RSVPreF3 OA) was efficacious against RSV-related lower respiratory tract disease (RSV-LRTD) in ≥60-years-olds over 1 RSV season. We evaluated efficacy and safety of 1 RSVPreF3 OA dose and of 2 RSVPreF3 OA doses given 1 year apart against RSV-LRTD over 2 RSV seasons post-dose 1. METHODS In this phase 3, blinded trial, ≥60-year-olds were randomized (1:1) to receive RSVPreF3 OA or placebo pre-season 1. RSVPreF3 OA recipients were re-randomized (1:1) to receive a second RSVPreF3 OA dose (RSV_revaccination group) or placebo (RSV_1dose group) pre-season 2; participants who received placebo pre-season 1 received placebo pre-season 2 (placebo group). Efficacy of both vaccine regimens against RSV-LRTD was evaluated over 2 seasons combined (confirmatory secondary objective, success criterion: lower limits of 2-sided CIs around efficacy estimates >20%). RESULTS The efficacy analysis comprised 24 967 participants (RSV_1dose: 6227; RSV_revaccination: 6242; placebo: 12 498). Median efficacy follow-up was 17.8 months. Efficacy over 2 seasons of 1 RSVPreF3 OA dose was 67.2% (97.5% CI: 48.2-80.0%) against RSV-LRTD and 78.8% (95% CI: 52.6-92.0%) against severe RSV-LRTD. Efficacy over 2 seasons of a first dose followed by revaccination was 67.1% (97.5% CI: 48.1-80.0%) against RSV-LRTD and 78.8% (95% CI: 52.5-92.0%) against severe RSV-LRTD. Reactogenicity/safety of the revaccination dose were similar to dose 1. CONCLUSIONS One RSVPreF3 OA dose was efficacious against RSV-LRTD over 2 RSV seasons in ≥60-year-olds. Revaccination 1 year post-dose 1 was well tolerated but did not seem to provide additional efficacy benefit in the overall study population. CLINICAL TRIALS REGISTRATION ClinicalTrials.gov: NCT04886596.
Collapse
Affiliation(s)
| | - Alberto Papi
- Pulmonary Division, University of Ferrara, St. Anna University Hospital, Ferrara, Italy
| | - Eugene Athan
- Barwon Health, University Hospital Geelong, Geelong, Australia
- Centre for Innovation in Infectious Diseases and Immunology Research, Deakin University, Geelong, Australia
| | | | - Joanne M Langley
- Canadian Center for Vaccinology, Dalhousie University, IWK Health and Nova Scotia Health, Halifax, Canada
| | - Dong-Gun Lee
- Division of Infectious Diseases, Department of Internal Medicine, Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Isabel Leroux-Roels
- Center for Vaccinology, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Federico Martinon-Torres
- Translational Pediatrics and Infectious Diseases, Pediatrics Department, Hospital Clínico Universitario de Santiago, Santiago de Compostela, Spain
- Genetics, Vaccines, Infectious Diseases, and Pediatrics Research Group, Instituto de Investigación Sanitaria de Santiago, Universidad de Santiago de Compostela, Santiago de Compostela, Spain
- Consorcio Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Tino F Schwarz
- Institute of Laboratory Medicine and Vaccination Center, Klinikum Würzburg Mitte, Campus Juliusspital, Würzburg, Germany
| | - Richard N van Zyl-Smit
- Division of Pulmonology and University of Cape Town Lung Institute, Department of Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Rostad CA, Atmar RL, Walter EB, Frey S, Meier JL, Sherman AC, Lai L, Tsong R, Kao CM, Raabe V, El Sahly HM, Keitel WA, Whitaker JA, Smith MJ, Schmader KE, Swamy GK, Abate G, Winokur P, Buchanan W, Cross K, Wegel A, Xu Y, Yildirim I, Kamidani S, Rouphael N, Roberts PC, Mulligan MJ, Anderson EJ. A Phase 2 Clinical Trial to Evaluate the Safety, Reactogenicity, and Immunogenicity of Different Prime-Boost Vaccination Schedules of 2013 and 2017 A(H7N9) Inactivated Influenza Virus Vaccines Administered With and Without AS03 Adjuvant in Healthy US Adults. Clin Infect Dis 2024; 78:1757-1768. [PMID: 38537255 PMCID: PMC11175706 DOI: 10.1093/cid/ciae173] [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: 01/23/2024] [Indexed: 06/15/2024] Open
Abstract
INTRODUCTION A surge of human influenza A(H7N9) cases began in 2016 in China from an antigenically distinct lineage. Data are needed about the safety and immunogenicity of 2013 and 2017 A(H7N9) inactivated influenza vaccines (IIVs) and the effects of AS03 adjuvant, prime-boost interval, and priming effects of 2013 and 2017 A(H7N9) IIVs. METHODS Healthy adults (n = 180), ages 19-50 years, were enrolled into this partially blinded, randomized, multicenter phase 2 clinical trial. Participants were randomly assigned to 1 of 6 vaccination groups evaluating homologous versus heterologous prime-boost strategies with 2 different boost intervals (21 vs 120 days) and 2 dosages (3.75 or 15 μg of hemagglutinin) administered with or without AS03 adjuvant. Reactogenicity, safety, and immunogenicity measured by hemagglutination inhibition and neutralizing antibody titers were assessed. RESULTS Two doses of A(H7N9) IIV were well tolerated, and no safety issues were identified. Although most participants had injection site and systemic reactogenicity, these symptoms were mostly mild to moderate in severity; injection site reactogenicity was greater in vaccination groups receiving adjuvant. Immune responses were greater after an adjuvanted second dose, and with a longer interval between prime and boost. The highest hemagglutination inhibition geometric mean titer (95% confidence interval) observed against the 2017 A(H7N9) strain was 133.4 (83.6-212.6) among participants who received homologous, adjuvanted 3.75 µg + AS03/2017 doses with delayed boost interval. CONCLUSIONS Administering AS03 adjuvant with the second H7N9 IIV dose and extending the boost interval to 4 months resulted in higher peak antibody responses. These observations can broadly inform strategic approaches for pandemic preparedness. Clinical Trials Registration. NCT03589807.
Collapse
MESH Headings
- Humans
- Influenza Vaccines/immunology
- Influenza Vaccines/administration & dosage
- Influenza Vaccines/adverse effects
- Adult
- Male
- Female
- Middle Aged
- Influenza A Virus, H7N9 Subtype/immunology
- Vaccines, Inactivated/immunology
- Vaccines, Inactivated/administration & dosage
- Vaccines, Inactivated/adverse effects
- Antibodies, Viral/blood
- Influenza, Human/prevention & control
- Influenza, Human/immunology
- Young Adult
- Immunization, Secondary
- Immunization Schedule
- Hemagglutination Inhibition Tests
- United States
- Immunogenicity, Vaccine
- Antibodies, Neutralizing/blood
- Polysorbates/administration & dosage
- Polysorbates/adverse effects
- alpha-Tocopherol/administration & dosage
- alpha-Tocopherol/adverse effects
- Squalene/administration & dosage
- Squalene/adverse effects
- Squalene/immunology
- Healthy Volunteers
- Drug Combinations
- Adjuvants, Vaccine/administration & dosage
- Vaccination/methods
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/adverse effects
Collapse
Affiliation(s)
- Christina A Rostad
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Robert L Atmar
- Departments of Medicine and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Emmanuel B Walter
- Department of Pediatrics and Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Sharon Frey
- Center for Vaccine Development, Saint Louis University, St. Louis, Missouri, USA
| | - Jeffery L Meier
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Amy C Sherman
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Lilin Lai
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Carol M Kao
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Vanessa Raabe
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- New York University Langone Vaccine Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Hana M El Sahly
- Departments of Medicine and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Wendy A Keitel
- Departments of Medicine and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Jennifer A Whitaker
- Departments of Medicine and Molecular Virology & Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Michael J Smith
- Department of Pediatrics and Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Kenneth E Schmader
- Department of Medicine-Geriatrics, Duke University and GRECC, Durham VA Health Care System, Durham, North Carolina, USA
| | - Geeta K Swamy
- Department of Obstetrics and Gynecology and Duke Human Vaccine Institute, Duke University, Durham, North Carolina, USA
| | - Getahun Abate
- Center for Vaccine Development, Saint Louis University, St. Louis, Missouri, USA
| | - Patricia Winokur
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Wendy Buchanan
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | | | | | - Yongxian Xu
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Inci Yildirim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Satoshi Kamidani
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Nadine Rouphael
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Paul C Roberts
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Mark J Mulligan
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
- New York University Langone Vaccine Center, NYU Grossman School of Medicine, New York, New York, USA
| | - Evan J Anderson
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
- Hope Clinic, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
43
|
Dorjee K, Namdon T, Topgyal S, Gyatso U, Tsundue T, Dolma T, Kumar V, Lhadon D, Yangkyi T, Khachoe T, Dorjee S, Sadoff RC, Peters D, Gupta A, Paster Z, Chaisson RE, Phunkyi D, Sadutshang TD. Association between Covishield vaccine and menstrual disturbance. Findings from a cross-sectional study among participants of Zero TB cohort in India. Vaccine 2024; 42:3572-3577. [PMID: 38679512 PMCID: PMC11875020 DOI: 10.1016/j.vaccine.2024.04.063] [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: 12/04/2023] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND The association between covid-19 vaccine and menstrual disturbance is unclear. METHODS An in-person cross-sectional survey among female members ≥ 18 years enrolled in an ongoing Zero TB prospective cohort in Northern India who had received one or two doses of covid-19 vaccine was conducted to study the characteristics and association of menstrual disturbance within six months of receiving Covishield. RESULTS Between June 29 and September 5, 2021, 339 females ≥ 18 years of age were administered the survey. Median age was 30 (IQR: 22-39) years; 84 % were between 18 and 49 and 16 % were ≥ 50 years old. There were 152 college students, 27 healthcare workers, and 160 nuns. Forty-two women (12 %) had received one dose and 297 (88 %) had received two doses of Covishield. Overall, 66 (20 %) women reported experiencing menstrual disturbance after receiving Covishield vaccine. The problems included early menstruation: 6 % (n = 19/339); late menstruation: 4 % (n = 14/339); and heavier bleeding: 5 % (n = 17/339). Disturbances lasted for less than seven days and cycles normalized in 1-3 months. There was no post-menopausal bleeding. There was no significant difference in menstrual disturbance based on receiving one vs. two doses of Covishield (OR: 1.58; 95 % CI: 0.55-4.57; p = 0.381). History of SARS-CoV-2 infection was not associated with the development of menstrual disturbance among the vaccinees (OR: 0.63; 95 % CI: 0.24-1.73; p = 0.379). Presence of emotional disturbance at baseline (OR: 31; 95 % CI: 3.52-267; p = 0.002) or previous history of dysmenorrhea (OR: 41; 95 % CI: 8.7-196; p < 0.001) was associated with menstrual disturbance in the vaccinees, indicating their potential to confound or bias study results. CONCLUSION Menstrual problems were reported by Covishield vaccinees, but they were minor and reversible within three months and do not constitute a ground for vaccine hesitancy. Studies designed to assess causal link taking care to avoid selection bias or confounding are needed.
Collapse
Affiliation(s)
- Kunchok Dorjee
- Center for TB and AIDS Research, Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD, USA; Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Gupta Klinsky India Institute, Johns Hopkins University, Baltimore, MD, USA.
| | | | | | | | | | | | - Vipan Kumar
- Department of Orthopedics, Dr. Rajendra Prasad Govt. Medical College, Tanda, HP, India
| | | | | | | | - Sangyal Dorjee
- Center for TB and AIDS Research, Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD, USA; Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - R C Sadoff
- Center for TB and AIDS Research, Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD, USA
| | - David Peters
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Amita Gupta
- Center for TB and AIDS Research, Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD, USA; Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA; Gupta Klinsky India Institute, Johns Hopkins University, Baltimore, MD, USA
| | - Zorba Paster
- Department of Family Medicine, University of Wisconsin, Wisconsin, USA
| | - Richard E Chaisson
- Center for TB and AIDS Research, Division of Infectious Diseases, Johns Hopkins University, Baltimore, MD, USA; Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | | | | |
Collapse
|
44
|
Davis C, Singh D, Anderson K, Vardeu A, Kopycinski J, Bridges-Webb A, Trickett A, O’Brien S, Downs M, Kaur R, Kolenovska R, Bussey L, Rutkowski K, Sebastian S, Cargill T, Barnes E, Evans TG, Cicconi P. Effect of Prior ChAdOx1 COVID-19 Immunisation on T-Cell Responses to ChAdOx1-HBV. Vaccines (Basel) 2024; 12:644. [PMID: 38932373 PMCID: PMC11209196 DOI: 10.3390/vaccines12060644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 05/24/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
There are varying data concerning the effect of prior anti-vector immunity on the T-cell response induced by immunisation with an identical vectored vaccine containing a heterologous antigen insert. To determine whether prior exposure to ChAdOx1-SARS-CoV2 immunisation (Vaxzevria®) impacts magnitudes of antigen-specific T-cell responses elicited by subsequent administration of the same viral vector (encoding HBV antigens, ChAdOx1-HBV), healthy volunteers that had received Vaxzevria® (n = 15) or the Pfizer or Moderna mRNA COVID-19 vaccine (n = 11) between 10 and 18 weeks prior were recruited to receive a single intramuscular injection of ChAdOx1-HBV. Anti-ChAdOx1-neutralising antibody titers were determined, and vector or insert-specific T-cell responses were measured by a gamma-interferon ELISpot and intracellular cytokine staining (ICS) assay using multiparameter flow cytometry. Participants were followed for three months after the ChAdOx1-HBV injection, which was well-tolerated, and no dropouts occurred. The baseline ChAdOx1 neutralisation titers were higher in the Vaxzevria® cohort (median of 848) than in the mRNA cohort (median of 25). T-cell responses to HBV antigens, measured by ELISpot, were higher on day 28 in the mRNA group (p = 0.013) but were similar between groups on day 84 (p = 0.441). By ICS, these differences persisted at the last time point. There was no clear correlation between the baseline responses to the adenoviral hexon and the subsequent ELISpot responses. As vaccination within 3 months using the same viral vector backbone affected the insert-specific T-cell responses, a greater interval after prior adenoviral immunisation using heterologous antigens may be warranted in settings in which these cells play critical roles.
Collapse
Affiliation(s)
- Charlotte Davis
- Barinthus Biotherapeutics, Harwell, Didcot OX11 0DF, UK (A.V.); (M.D.)
| | - Dave Singh
- Medicines Evaluation Unit Ltd., Manchester M23 9QZ, UK;
| | - Katie Anderson
- Barinthus Biotherapeutics, Harwell, Didcot OX11 0DF, UK (A.V.); (M.D.)
| | - Antonella Vardeu
- Barinthus Biotherapeutics, Harwell, Didcot OX11 0DF, UK (A.V.); (M.D.)
| | - Jakub Kopycinski
- Barinthus Biotherapeutics, Harwell, Didcot OX11 0DF, UK (A.V.); (M.D.)
| | | | - Alice Trickett
- Barinthus Biotherapeutics, Harwell, Didcot OX11 0DF, UK (A.V.); (M.D.)
| | - Susanne O’Brien
- Barinthus Biotherapeutics, Harwell, Didcot OX11 0DF, UK (A.V.); (M.D.)
| | - Matthew Downs
- Barinthus Biotherapeutics, Harwell, Didcot OX11 0DF, UK (A.V.); (M.D.)
| | - Randip Kaur
- Barinthus Biotherapeutics, Harwell, Didcot OX11 0DF, UK (A.V.); (M.D.)
| | - Radka Kolenovska
- Barinthus Biotherapeutics, Harwell, Didcot OX11 0DF, UK (A.V.); (M.D.)
| | - Louise Bussey
- Barinthus Biotherapeutics, Harwell, Didcot OX11 0DF, UK (A.V.); (M.D.)
| | - Kathryn Rutkowski
- Barinthus Biotherapeutics, Harwell, Didcot OX11 0DF, UK (A.V.); (M.D.)
| | - Sarah Sebastian
- Barinthus Biotherapeutics, Harwell, Didcot OX11 0DF, UK (A.V.); (M.D.)
| | - Tamsin Cargill
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 2JD, UK (E.B.)
| | - Eleanor Barnes
- Nuffield Department of Medicine, University of Oxford, Oxford OX1 2JD, UK (E.B.)
| | - Thomas G. Evans
- Barinthus Biotherapeutics, Harwell, Didcot OX11 0DF, UK (A.V.); (M.D.)
| | - Paola Cicconi
- Centre for Clinical Vaccinology and Tropical Medicine (CCVTM), University of Oxford, Oxford OX1 2JD, UK;
| |
Collapse
|
45
|
Ward T, Fyles M, Glaser A, Paton RS, Ferguson W, Overton CE. The real-time infection hospitalisation and fatality risk across the COVID-19 pandemic in England. Nat Commun 2024; 15:4633. [PMID: 38821930 PMCID: PMC11143367 DOI: 10.1038/s41467-024-47199-3] [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: 02/14/2023] [Accepted: 03/22/2024] [Indexed: 06/02/2024] Open
Abstract
The COVID-19 pandemic led to 231,841 deaths and 940,243 hospitalisations in England, by the end of March 2023. This paper calculates the real-time infection hospitalisation risk (IHR) and infection fatality risk (IFR) using the Office for National Statistics Coronavirus Infection Survey (ONS CIS) and the Real-time Assessment of Community Transmission Survey between November 2020 to March 2023. The IHR and the IFR in England peaked in January 2021 at 3.39% (95% Credible Intervals (CrI): 2.79, 3.97) and 0.97% (95% CrI: 0.62, 1.36), respectively. After this time, there was a rapid decline in the severity from infection, with the lowest estimated IHR of 0.32% (95% CrI: 0.27, 0.39) in December 2022 and IFR of 0.06% (95% CrI: 0.04, 0.08) in April 2022. We found infection severity to vary more markedly between regions early in the pandemic however, the absolute heterogeneity has since reduced. The risk from infection of SARS-CoV-2 has changed substantially throughout the COVID-19 pandemic with a decline of 86.03% (80.86, 89.35) and 89.67% (80.18, 93.93) in the IHR and IFR, respectively, since early 2021. From April 2022 until March 2023, the end of the ONS CIS study, we found fluctuating patterns in the severity of infection with the resumption of more normative mixing, resurgent epidemic waves, patterns of waning immunity, and emerging variants that have shown signs of convergent evolution.
Collapse
Affiliation(s)
- Thomas Ward
- UK Health Security Agency, Data, Analytics and Surveillance, Nobel House, London, SW1P 3JR, UK.
| | - Martyn Fyles
- UK Health Security Agency, Data, Analytics and Surveillance, Nobel House, London, SW1P 3JR, UK
| | - Alex Glaser
- UK Health Security Agency, Data, Analytics and Surveillance, Nobel House, London, SW1P 3JR, UK
| | - Robert S Paton
- UK Health Security Agency, Data, Analytics and Surveillance, Nobel House, London, SW1P 3JR, UK
| | - William Ferguson
- UK Health Security Agency, Data, Analytics and Surveillance, Nobel House, London, SW1P 3JR, UK
| | - Christopher E Overton
- UK Health Security Agency, Data, Analytics and Surveillance, Nobel House, London, SW1P 3JR, UK
- University of Liverpool, Department of Mathematical Sciences, Peach Street, Liverpool, UK
| |
Collapse
|
46
|
Wang W, Yellamsetty A, Edmonds RM, Barcavage SR, Bao S. COVID-19 vaccination-related tinnitus is associated with pre-vaccination metabolic disorders. Front Pharmacol 2024; 15:1374320. [PMID: 38841369 PMCID: PMC11150672 DOI: 10.3389/fphar.2024.1374320] [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: 01/22/2024] [Accepted: 04/29/2024] [Indexed: 06/07/2024] Open
Abstract
Cases of tinnitus have been reported following administration of COVID-19 vaccines. The aim of this study was to characterize COVID-19 vaccination-related tinnitus to assess whether there is a causal relationship, and to examine potential risk factors for COVID-19 vaccination-related tinnitus. We analyzed a survey on 398 cases of COVID-19 vaccination-related tinnitus, and 699,839 COVID-19 vaccine-related reports in the Vaccine Adverse Effect Reporting System (VAERS) database that was retrieved on 4 December 2021. We found that following COVID-19 vaccination, 1) tinnitus report frequencies for Pfizer, Moderna and Janssen vaccines in VAERS are 47, 51 and 70 cases per million full vaccination; 2) the symptom onset was often rapid; 3) more women than men reported tinnitus and the sex difference increased with age; 4) for 2-dose vaccines, the frequency of tinnitus was higher following the first dose than the second dose; 5) for 2-dose vaccines, the chance of worsening tinnitus symptoms after second dose was approximately 50%; 6) tinnitus was correlated with other neurological and psychiatric symptoms; 7) pre-existing metabolic syndromes were correlated with the severity of the reported tinnitus. These findings suggest that COVID-19 vaccination increases the risk of tinnitus, and metabolic disorders is a risk factor for COVID-19 vaccination-related tinnitus.
Collapse
Affiliation(s)
- Weihua Wang
- Department of Physiology and Department of Otolaryngology—Head and Neck Surgery, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Anusha Yellamsetty
- Department of Audiology, College of Health and Human Sciences, San José State University, San José, CA, United States
| | | | | | - Shaowen Bao
- Department of Physiology and Department of Otolaryngology—Head and Neck Surgery, University of Arizona College of Medicine, Tucson, AZ, United States
| |
Collapse
|
47
|
Jin G, Wang R, Jin Y, Song Y, Wang T. From intramuscular to nasal: unleashing the potential of nasal spray vaccines against coronavirus disease 2019. Clin Transl Immunology 2024; 13:e1514. [PMID: 38770238 PMCID: PMC11103645 DOI: 10.1002/cti2.1514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 05/08/2024] [Accepted: 05/09/2024] [Indexed: 05/22/2024] Open
Abstract
Coronavirus disease 2019, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has affected 700 million people worldwide since its outbreak in 2019. The current pandemic strains, including Omicron and its large subvariant series, exhibit strong transmission and stealth. After entering the human body, the virus first infects nasal epithelial cells and invades host cells through the angiotensin-converting enzyme 2 receptor and transmembrane serine protease 2 on the host cell surface. The nasal cavity is an important body part that protects against the virus. Immunisation of the nasal mucosa produces immunoglobulin A antibodies that effectively neutralise viruses. Saline nasal irrigation, a type of physical therapy, can reduce the viral load in the nasal cavity and prevent viral infections to some extent. As a commonly used means to fight SARS-CoV-2, the intramuscular (IM) vaccine can induce the human body to produce a systemic immune response and immunoglobulin G antibody; however, the antibody is difficult to distribute to the nasal mucosa in time and cannot achieve a good preventive effect. Intranasal (IN) vaccines compensate for the shortcomings of IM vaccines, induce mucosal immune responses, and have a better effect in preventing infection. In this review, we discuss the nasal defence barrier, the harm caused by SARS-CoV-2, the mechanism of its invasion into host cells, nasal cleaning, IM vaccines and IN vaccines, and suggest increasing the development of IN vaccines, and use of IN vaccines as a supplement to IM vaccines.
Collapse
Affiliation(s)
- Ge Jin
- Faculty of MedicineDalian University of TechnologyDalianLiaoningChina
- Department of RadiotherapyCancer Hospital of China Medical University, Liaoning Cancer Hospital and InstituteShenyangLiaoningChina
| | - Runze Wang
- Department of RadiotherapyCancer Hospital of China Medical University, Liaoning Cancer Hospital and InstituteShenyangLiaoningChina
| | - Yi Jin
- Department of Breast SurgeryLiaoning Cancer Hospital and InstituteShenyangLiaoningChina
| | - Yingqiu Song
- Department of RadiotherapyCancer Hospital of China Medical University, Liaoning Cancer Hospital and InstituteShenyangLiaoningChina
| | - Tianlu Wang
- Faculty of MedicineDalian University of TechnologyDalianLiaoningChina
- Department of RadiotherapyCancer Hospital of China Medical University, Liaoning Cancer Hospital and InstituteShenyangLiaoningChina
- Department of RadiotherapyCancer Hospital of Dalian University of TechnologyDalianLiaoningChina
| |
Collapse
|
48
|
Sun Y, Huang W, Xiang H, Nie J. SARS-CoV-2 Neutralization Assays Used in Clinical Trials: A Narrative Review. Vaccines (Basel) 2024; 12:554. [PMID: 38793805 PMCID: PMC11125816 DOI: 10.3390/vaccines12050554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
Since the emergence of COVID-19, extensive research efforts have been undertaken to accelerate the development of multiple types of vaccines to combat the pandemic. These include inactivated, recombinant subunit, viral vector, and nucleic acid vaccines. In the development of these diverse vaccines, appropriate methods to assess vaccine immunogenicity are essential in both preclinical and clinical studies. Among the biomarkers used in vaccine evaluation, the neutralizing antibody level serves as a pivotal indicator for assessing vaccine efficacy. Neutralizing antibody detection methods can mainly be classified into three types: the conventional virus neutralization test, pseudovirus neutralization test, and surrogate virus neutralization test. Importantly, standardization of these assays is critical for their application to yield results that are comparable across different laboratories. The development and use of international or regional standards would facilitate assay standardization and facilitate comparisons of the immune responses induced by different vaccines. In this comprehensive review, we discuss the principles, advantages, limitations, and application of different SARS-CoV-2 neutralization assays in vaccine clinical trials. This will provide guidance for the development and evaluation of COVID-19 vaccines.
Collapse
Affiliation(s)
- Yeqing Sun
- School of Life Sciences, Jilin University, Changchun 130012, China;
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102629, China;
| | - Weijin Huang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102629, China;
| | - Hongyu Xiang
- School of Life Sciences, Jilin University, Changchun 130012, China;
| | - Jianhui Nie
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, State Key Laboratory of Drug Regulatory Science, NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, Beijing 102629, China;
| |
Collapse
|
49
|
Berry MT, Khan SR, Schlub TE, Notaras A, Kunasekaran M, Grulich AE, MacIntyre CR, Davenport MP, Khoury DS. Predicting vaccine effectiveness for mpox. Nat Commun 2024; 15:3856. [PMID: 38719852 PMCID: PMC11078999 DOI: 10.1038/s41467-024-48180-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
The Modified Vaccinia Ankara vaccine developed by Bavarian Nordic (MVA-BN) was widely deployed to prevent mpox during the 2022 global outbreak. This vaccine was initially approved for mpox based on its reported immunogenicity (from phase I/II trials) and effectiveness in animal models, rather than evidence of clinical efficacy. However, no validated correlate of protection after vaccination has been identified. Here we performed a systematic search and meta-analysis of the available data to test whether vaccinia-binding ELISA endpoint titer is predictive of vaccine effectiveness against mpox. We observe a significant correlation between vaccine effectiveness and vaccinia-binding antibody titers, consistent with the existing assumption that antibody levels may be a correlate of protection. Combining this data with analysis of antibody kinetics after vaccination, we predict the durability of protection after vaccination and the impact of dose spacing. We find that delaying the second dose of MVA-BN vaccination will provide more durable protection and may be optimal in an outbreak with limited vaccine stock. Although further work is required to validate this correlate, this study provides a quantitative evidence-based approach for using antibody measurements to predict the effectiveness of mpox vaccination.
Collapse
Affiliation(s)
- Matthew T Berry
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Shanchita R Khan
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - Timothy E Schlub
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
- Sydney School of Public Health, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Adriana Notaras
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | | | - Andrew E Grulich
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
| | - C Raina MacIntyre
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia
- College of Public Service and Community Solutions, and College of Health Solutions, Arizona State University, Tempe, AZ, USA
| | - Miles P Davenport
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia.
| | - David S Khoury
- Kirby Institute, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
50
|
Ma B, Tao M, Li Z, Zheng Q, Wu H, Chen P. Mucosal vaccines for viral diseases: Status and prospects. Virology 2024; 593:110026. [PMID: 38373360 DOI: 10.1016/j.virol.2024.110026] [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: 09/19/2023] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Virus-associated infectious diseases are highly detrimental to human health and animal husbandry. Among all countermeasures against infectious diseases, prophylactic vaccines, which developed through traditional or novel approaches, offer potential benefits. More recently, mucosal vaccines attract attention for their extraordinary characteristics compared to conventional parenteral vaccines, particularly for mucosal-related pathogens. Representatively, coronavirus disease 2019 (COVID-19), a respiratory disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), further accelerated the research and development efforts for mucosal vaccines by thoroughly investigating existing strategies or involving novel techniques. While several vaccine candidates achieved positive progresses, thus far, part of the current COVID-19 mucosal vaccines have shown poor performance, which underline the need for next-generation mucosal vaccines and corresponding platforms. In this review, we summarized the typical mucosal vaccines approved for humans or animals and sought to elucidate the underlying mechanisms of these successful cases. In addition, mucosal vaccines against COVID-19 that are in human clinical trials were reviewed in detail since this public health event mobilized all advanced technologies for possible solutions. Finally, the gaps in developing mucosal vaccines, potential solutions and prospects were discussed. Overall, rational application of mucosal vaccines would facilitate the establishing of mucosal immunity and block the transmission of viral diseases.
Collapse
Affiliation(s)
- Bingjie Ma
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Mengxiao Tao
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Zhili Li
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Quanfang Zheng
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Haigang Wu
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China
| | - Peirong Chen
- College of Animal Science and Technology, Xinyang Agriculture and Forestry University, Xinyang, China.
| |
Collapse
|