1
|
Figueroa AL, Ali K, Berman G, Xu W, Deng W, Girard B, Yeakey A, Slobod K, Miller J, Das R, Priddy F. Safety and immunogenicity of an mRNA-1273 vaccine booster in adolescents. Hum Vaccin Immunother 2025; 21:2436714. [PMID: 39836458 DOI: 10.1080/21645515.2024.2436714] [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: 05/07/2024] [Revised: 10/11/2024] [Accepted: 11/28/2024] [Indexed: 01/22/2025] Open
Abstract
Safety, immunogenicity, and effectiveness of an mRNA-1273 50-μg booster were evaluated in adolescents (12-17 years), with and without pre-booster SARS-CoV-2 infection. Participants who had received the 2-dose mRNA-1273 100-µg primary series in the TeenCOVE trial (NCT04649151) were offered the mRNA-1273 50-μg booster. Primary objectives included safety and inference of effectiveness by establishing noninferiority of neutralizing antibody (nAb) responses after the booster compared with the nAb post-primary series of mRNA-1273 among young adults in COVE (NCT04470427). Binding antibody (bAb) responses against SARS-CoV-2 variants of interest and COVID-19 incidence after vaccination were also evaluated. Median boosting interval was 315 days. The mRNA-1273 booster was well-tolerated, with an acceptable safety profile. Relative to pre-booster, nAb geometric mean levels increased after the booster by 17.8-fold and 4.7-fold among pre-booster SARS-CoV-2-negative and -positive participants, respectively. Effectiveness was successfully inferred based on noninferiority of nAb levels from mRNA-1273 booster dose (Day 29) compared with nAb levels after mRNA-1273 primary series (Day 57) among young adults in COVE. Further, the booster increased bAb levels relative to pre-booster baseline against SARS-CoV-2 variants (alpha [B.1.1.7], beta [B.1.351], gamma [P.1], and delta [B.1.617.2]), regardless of pre-booster SARS-CoV-2 status. COVID-19 incidence (cases per 1000 person-months) was lower among boosted (0 cases) than non-boosted (95.766 cases) participants in January 2022, a peak period during the early omicron transmission. In summary, the mRNA-1273 50-μg booster induced robust nAb responses in previously vaccinated adolescents, regardless of SARS-CoV-2 serostatus. Effectiveness was successfully inferred and the booster was well-tolerated, with no new safety concerns identified.
Collapse
Affiliation(s)
- Amparo L Figueroa
- Clinical Development, Infectious Diseases, Moderna, Inc., Cambridge, MA, USA
| | - Kashif Ali
- Kool Kids Pediatrics, DM Clinical Research, Houston, TX, USA
| | - Gary Berman
- Clinical Research Institute, Allergy and Immunology, Minneapolis, MN, USA
| | - Wenqin Xu
- Biostatistics, Moderna, Inc., Cambridge, MA, USA
| | - Weiping Deng
- Biostatistics, Moderna, Inc., Cambridge, MA, USA
| | | | | | - Karen Slobod
- Cambridge ID & Immunology Consulting, LLC, Somerville, MA, USA
| | - Jacqueline Miller
- Research and Development, Infectious Disease, Moderna, Inc., Cambridge, MA, USA
| | - Rituparna Das
- Research and Development, Infectious Disease, Moderna, Inc., Cambridge, MA, USA
| | - Frances Priddy
- Research and Development, Infectious Disease, Moderna, Inc., Cambridge, MA, USA
| |
Collapse
|
2
|
Hurley LP, Kurlandsky K, Breslin K, Stein A, Hambidge SJ, Shoup JA, Reifler LM, Daley MF, Lewin B, Goddard K, Henninger ML, Nelson JC, Vazquez-Benitez G, Hanson KE, Fuller CC, Williams JT. Attitudes and beliefs regarding COVID-19 and COVID-19 Omicron booster vaccine among adults in the vaccine safety datalink, 2022-2023. Hum Vaccin Immunother 2025; 21:2467548. [PMID: 40179339 PMCID: PMC11980469 DOI: 10.1080/21645515.2025.2467548] [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] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 04/05/2025] Open
Abstract
COVID-19 vaccination rates are decreasing despite vaccination being the most effective tool against severe disease from COVID-19. From October 1, 2022, to February 1, 2023, we conducted a cross-sectional study among adults in the Vaccine Safety Datalink about attitudes and beliefs regarding bivalent COVID-19 Omicron booster vaccine (hereafter referred to as COVID-19 bivalent vaccine) stratifying by vaccination status and race and ethnicity. Analysis was weighted for response and selection bias. The response rate was 27% (385/1430); 33% [95% CI: 21%-44%] of respondents were 'fully vaccinated' (had received COVID-19 bivalent vaccine), 54% [42%-67%] were partially vaccinated, and 13% [7%-19%] were unvaccinated. Fully vaccinated adults were more likely to consider COVID-19 bivalent vaccine 'very effective' (64%, [43%-86%]) at preventing hospitalization due to COVID-19 than partially (31%, [12%-50%]) or unvaccinated (2%, [0%-6%]) adults. Fully vaccinated adults were more likely to report COVID-19 bivalent vaccine was 'very safe' (83%, [69%-98%]) than partially (43%, [23%-63%]) or unvaccinated adults (2%, 0%-6%). Non-Hispanic White adults were more likely to report COVID-19 bivalent vaccine was 'very safe' (71%, [54%-87%]) than Non-Hispanic Black (36%, [21%-50%]) and Hispanic (26%, [7%-45%]) adults. A dose-response effect between vaccination status and perceptions of COVID-19 bivalent vaccine safety and effectiveness was observed, with fully vaccinated respondents having the most favorable attitudes. Racial and ethnic differences in perceived vaccine safety were also found. Improved communication about vaccine effectiveness and safety is key to improving low vaccination rates.
Collapse
Affiliation(s)
- Laura P. Hurley
- Ambulatory Care Services, Denver Health and Hospitals, Denver, CO, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Kate Kurlandsky
- Ambulatory Care Services, Denver Health and Hospitals, Denver, CO, USA
| | - Kristin Breslin
- Ambulatory Care Services, Denver Health and Hospitals, Denver, CO, USA
| | - Amy Stein
- Ambulatory Care Services, Denver Health and Hospitals, Denver, CO, USA
| | - Simon J. Hambidge
- Ambulatory Care Services, Denver Health and Hospitals, Denver, CO, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Jo Ann Shoup
- Institute for Health Research, Kaiser Permanente Colorado, Aurora, CO, USA
| | - Liza M. Reifler
- Institute for Health Research, Kaiser Permanente Colorado, Aurora, CO, USA
| | - Matthew F. Daley
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
- Institute for Health Research, Kaiser Permanente Colorado, Aurora, CO, USA
| | - Bruno Lewin
- Department of Family Medicine, Kaiser Permanente Southern California, Pasadena, CA, USA
| | | | | | - Jennifer C. Nelson
- Biostatistics Unit, Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
| | | | | | - Candace C. Fuller
- Department of Population Medicine, Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Joshua T.B. Williams
- Ambulatory Care Services, Denver Health and Hospitals, Denver, CO, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
3
|
Smith DS, Postma M, Fisman D, Mould-Quevedo J. Cost-effectiveness models assessing COVID-19 booster vaccines across eight countries: A review of methods and data inputs. Vaccine 2025; 51:126879. [PMID: 39956089 DOI: 10.1016/j.vaccine.2025.126879] [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: 08/02/2024] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 02/18/2025]
Abstract
Coronavirus disease 2019 (COVID-19) continues to cause serious health consequences globally. Policy makers now assess cost effectiveness (CE) when evaluating COVID-19 vaccines. A targeted literature review was performed to examine recent CE evidence for COVID-19 vaccines, as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) transitions from pandemic to endemic, to identify best practices. Data were from large EU countries (UK, Spain, Germany, France, and Italy), US, Canada, and Australia. Nine CE studies met the inclusion criteria. Studies evaluated booster vaccination, and mainly considered mRNA vaccines. CE studies reported that COVID-19 vaccines provided health benefits and were cost-effective or showed cost-savings. Benefits were more pronounced in older and high-risk populations based on higher rates of COVID-19 hospitalization and death. CE findings were most sensitive to estimates of incidence of COVID-19, SARS-CoV-2 transmissibility, vaccine effectiveness, waning/duration of vaccine protection, and hospitalization costs. Most data inputs were sourced from real-world evidence (RWE). Lack of inclusion of some parameters, such as transmission modeling, productivity losses, and the impact of long COVID may undervalue COVID-19 vaccines. As SARS-CoV-2 evolves and COVID-19 vaccines are updated, continuous generation of RWE is needed to demonstrate the CE of COVID-19 vaccines in an ongoing manner.
Collapse
Affiliation(s)
- Darvin S Smith
- Department of Infectious Disease, San Mateo Medical Center, San Mateo, CA, USA
| | - Maarten Postma
- University Medical Center Groningen, University of Groningen, Groningen, the Netherlands; Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia; Faculty of Economics & Business, University of Groningen, Groningen, the Netherlands; Department of Pharmacology and Therapy, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - David Fisman
- Division of Epidemiology, Dalla Lana School of Public Health, Toronto, Canada
| | | |
Collapse
|
4
|
Jang G, Kim J, Thompson RN, Lee H. Modeling vaccination prioritization strategies for post-pandemic COVID-19 in the Republic of Korea accounting for under-reporting and age-structure. J Infect Public Health 2025; 18:102688. [PMID: 39913986 DOI: 10.1016/j.jiph.2025.102688] [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: 11/29/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Vaccination has played a key role in limiting the impacts of COVID-19. Even though the acute phase of the COVID-19 pandemic is now over, the potential for substantial numbers of cases and deaths due to novel SARS-CoV-2 variants remains. In the Republic of Korea, a strategy of vaccinating individuals in high-risk groups annually began in October 2023. METHODS We used mathematical modeling to assess the effectiveness of alternative vaccination strategies under different assumptions about the number of available vaccine doses. An age-structured transmission model was developed using vaccination and seropositivity data. Various vaccination scenarios were considered, taking into account the effect of undetected or unreported cases (with different levels of reporting by age group): S1: prioritizing vaccination towards the oldest individuals; S2: prioritizing vaccination towards the youngest individuals; and S3: spreading vaccines among all age groups. RESULTS Our analysis reveals three key findings. First, administering vaccines to older age groups reduces the number of deaths, while instead targeting younger individuals reduces the number of infections. Second, with approximately 6,000,000 doses available annually, it is recommended that older age groups are prioritized for vaccination, achieving a substantial reduction in the number of deaths compared to a scenario without vaccination. Finally, since case detection (and subsequent isolation) affects transmission, the number of cumulative cases was found to be affected substantially by changes in the reporting rate. CONCLUSIONS In conclusion, vaccination and case detection (facilitated by contact tracing) both play important roles in limiting the impacts of COVID-19. The mathematical modeling approach presented here provides a framework for assessing the effectiveness of different vaccination strategies in scenarios with limited vaccine supply.
Collapse
Affiliation(s)
- Geunsoo Jang
- Nonlinear Dynamics and Mathematical Application Center, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Jihyeon Kim
- Department of Statistics, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Robin N Thompson
- Mathematical Institute, University of Oxford, Oxford OX2 6GG, United Kingdom
| | - Hyojung Lee
- Department of Statistics, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
5
|
Beck E, Georgieva M, Wang WJ, Gomez-Lievano A, Wang H, Gao Y, Kopel H, Bausch-Jurken M, Patterson-Lomba O, Mu F, Wu E, Van de Velde N. Indirect comparison of the relative vaccine effectiveness of mRNA-1283 vs. BNT162b2 vaccines against symptomatic COVID-19 among US adults. Curr Med Res Opin 2025; 41:721-732. [PMID: 39973309 DOI: 10.1080/03007995.2025.2466726] [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: 12/09/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND COVID-19 continues to pose a significant health burden, particularly among older adults. mRNA-1283 is a next-generation COVID-19 mRNA vaccine developed to enhance immune response. Findings from the Phase 3 NextCOVE trial comparing bivalent versions of mRNA-1273 and mRNA-1283 vaccines have recently become available. However, there are no head-to-head trials comparing mRNA-1283 and the BNT162b2 vaccine. OBJECTIVE To indirectly compare the effectiveness of mRNA-1283 and BNT162b2 against symptomatic COVID-19 among adults in the US. METHODS A targeted literature review was conducted to identify relevant studies comparing the mRNA-1273 and BNT162b2 bivalent vaccines. A real-world evidence (RWE) study by Kopel et al. (2023) assessing the relative vaccine effectiveness (rVE) of mRNA-1273 vs. BNT162b2 was selected for an indirect treatment comparison (ITC) against the NextCOVE trial using the Bucher method. Analyses were stratified by age group and sensitivity analyses were conducted using alternative outcome definitions. RESULTS Despite differences between NextCOVE and the Kopel study, comparability assessments supported a robust ITC. Among participants ≥18 years of age, the indirect rVE of mRNA-1283 vs. BNT162b2 against symptomatic COVID-19 was 15.3% (95% CI = 4.7-24.8%, p = 0.006). For adults ≥65 years of age, the rVE was 22.8% (95% CI = 3.7-38.1%, p = 0.022). Sensitivity analyses with alternative outcome definitions supported these estimates. CONCLUSION This analysis provides consistent and statistically significant evidence indicating the next-generation mRNA-1283 vaccine is more effective in preventing symptomatic COVID-19 than BNT162b2, with the largest effect in individuals aged ≥65. Consistent results across sensitivity analyses underscore the robustness of the findings, offering important evidence to inform vaccination decisions by policymakers, providers, and payers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Fan Mu
- Analysis Group, Inc, Boston, MA, USA
| | - Eric Wu
- Analysis Group, Inc, Boston, MA, USA
| | | |
Collapse
|
6
|
Case JB, Jain S, Suthar MS, Diamond MS. SARS-CoV-2: The Interplay Between Evolution and Host Immunity. Annu Rev Immunol 2025; 43:29-55. [PMID: 39705164 DOI: 10.1146/annurev-immunol-083122-043054] [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] [Indexed: 12/22/2024]
Abstract
The persistence of SARS-CoV-2 infections at a global level reflects the repeated emergence of variant strains encoding unique constellations of mutations. These variants have been generated principally because of a dynamic host immune landscape, the countermeasures deployed to combat disease, and selection for enhanced infection of the upper airway and respiratory transmission. The resulting viral diversity creates a challenge for vaccination efforts to maintain efficacy, especially regarding humoral aspects of protection. Here, we review our understanding of how SARS-CoV-2 has evolved during the pandemic, the immune mechanisms that confer protection, and the impact viral evolution has had on transmissibility and adaptive immunity elicited by natural infection and/or vaccination. Evidence suggests that SARS-CoV-2 evolution initially selected variants with increased transmissibility but currently is driven by immune escape. The virus likely will continue to drift to maintain fitness until countermeasures capable of disrupting transmission cycles become widely available.
Collapse
Affiliation(s)
- James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| | - Shilpi Jain
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael S Diamond
- Department of Pathology & Immunology; Department of Molecular Microbiology; and Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| |
Collapse
|
7
|
Magalhães BDAP, Medeiros Minasi J, Lobato RC, Lemos LC, de Britto LS, Barros RM, de Martínez AMB, da Hora VP. Globally approved vaccines for COVID-19: a systematic review. Braz J Microbiol 2025; 56:511-527. [PMID: 39786643 PMCID: PMC11885735 DOI: 10.1007/s42770-024-01600-x] [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: 07/09/2024] [Accepted: 12/17/2024] [Indexed: 01/12/2025] Open
Abstract
COVID-19 caused a public health emergency, which instituted a global effort to develop vaccines using different platforms, such as basic types and new-generation vaccines. Considering the importance of vaccination in preventing the severity of infectious diseases and the success in developing and approving vaccines against COVID-19 in record time, it is essential to learn about the characteristics of these vaccines. This study aimed to conduct a structured, systematic review following the PRISMA guideline, to analyze the general characteristics of vaccines approved globally for use against COVID-19. We used the list of approved vaccines available by the WHO as guidance to search for studies in the literature. We searched the terms "SARS-CoV-2 and vaccine and safety and efficacy" in the MEDLINE via PUBMED and Web of Science databases. We conducted the research on both bases, including complete articles published from January 2020 to June 2023. The selection of files occurred between May/2021 and June/2023. Therefore, the paper did not consider articles published after this period or vaccines approved after this moment. This study only included approved vaccines; phase three studies published in English. We found 11 published articles from phase three that met the established criteria. The vaccines included in this study were: Cominarty, mRNA-1273 or Spikevax, Vaxzevria or AZD1222 or Covishield, CoronaVac or PicoVacc, and Ad26.COV2.S, SputnikV or Gam-Covid-Vac, Covaxin, NVX-CoV2373 or Covovax or Nuvaxovid, WIV04 and HB02, CoVLP or Covifenz and Convidecia or Ad5-nCoV. We summarized the main findings of each vaccine, considering the vaccine composition, number of doses, efficacy analyses, and main adverse effects. In general, the vaccines had high efficacy rates and few adverse effects. Efficacy values are important for vaccine approval, but they will not necessarily reflect the real-world impact of vaccination. It was seen that the effectiveness of COV2.S, CoronaVac/PicoVacc, Cominarty, and Covaxin vaccines was lower than the efficacy, whereas, for AZD1222/Vaxzevria/Covishield, the two parameters remained at similar rates. All vaccines evaluated have different compositions, dosages, populations, and study designs. All are effective in at least preventing symptomatic COVID-19, causing mild or moderate adverse effects when present.
Collapse
Affiliation(s)
- Brenda de Almeida Perret Magalhães
- Interdisciplinary Group of Virology and Immunology, Faculty of Medicine, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil.
- Post-Graduate Program in Health Sciences, Faculty of Medicine, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil.
- Post-Graduate Program in Health Sciences, Faculty of Medicine, Federal University of Rio Grande (FURG), Visconde de Paranaguá Street, 102, Centro, Rio Grande, RS, 96203-900, Brazil.
| | - Jéssica Medeiros Minasi
- Post-Graduate Program in Health Sciences, Faculty of Medicine, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
| | - Rubens Caurio Lobato
- Interdisciplinary Group of Virology and Immunology, Faculty of Medicine, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
| | - Luiza Curi Lemos
- Interdisciplinary Group of Virology and Immunology, Faculty of Medicine, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
- Post-Graduate Program in Health Sciences, Faculty of Medicine, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
| | - Laryssa Saez de Britto
- Interdisciplinary Group of Virology and Immunology, Faculty of Medicine, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
| | - Rhaysa Madruga Barros
- Interdisciplinary Group of Virology and Immunology, Faculty of Medicine, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
| | - Ana Maria Barral de Martínez
- Interdisciplinary Group of Virology and Immunology, Faculty of Medicine, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
- Post-Graduate Program in Health Sciences, Faculty of Medicine, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
| | - Vanusa Pousada da Hora
- Interdisciplinary Group of Virology and Immunology, Faculty of Medicine, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
- Post-Graduate Program in Health Sciences, Faculty of Medicine, Federal University of Rio Grande (FURG), Rio Grande, RS, Brazil
| |
Collapse
|
8
|
Yao Y, Yang Y, Wu Q, Liu M, Bao W, Wang Q, Cheng M, Chen Y, Yu Y, Cai Y, Zhang M, Yao J, He H, Jin C, Zheng C, Jin T, Tong D. Neutralizing antibody test supports booster strategy for young individuals after SARS-CoV-2 Omicron breakthrough. Eur J Med Res 2025; 30:7. [PMID: 39757187 DOI: 10.1186/s40001-024-02240-5] [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: 11/08/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND The SARS-CoV-2 Omicron variant, since its initial detection, has rapidly spread across the globe, becoming the dominant strain. It is important to study the immune response of SARS-CoV-2 Omicron variant due to its remarkable ability to escape the majority of existing SARS-CoV-2 neutralizing antibodies. The surge in SARS-CoV-2 Omicron infections among most Chinese residents by the end of 2022 provides a unique opportunity to understand immune system's response to Omicron in populations with limited exposure to prior SARS-CoV-2 variants. METHODS We tested the levels of IgG, IgA, and IgM specific to the prototype SARS-CoV-2 RBD (receptor-binding domain) in blood samples from 636 individuals by chemical luminescence assay, ELISA and pseudovirus-based neutralization assay. RESULTS Inoculation with inactivated prototype SARS-CoV-2 vaccines or recombinant protein vaccines showed higher IgG levels after infection than the unvaccinated individuals. Moreover, the age resulted in different IgG levels after the Omicron infection as IgG level of the patients aged > 60 years was lower than that of patients aged < 60 years. This indicates that the IgG induced by SARS-CoV-2 Omicron breakthrough infection was different between old and young individuals. We found that a booster dose of the prototype SARS-CoV-2 vaccine led to a significant increase in the neutralizing immune response against the prototype SARS-CoV-2 and helped induce neutralizing antibodies against BA.5 and BF.7 variants after an Omicron breakthrough infection in young individuals, which is different from a previous report on older people. CONCLUSIONS These data suggest that the prototype SARS-CoV-2 booster vaccination helps induce high levels of neutralizing antibodies against Omicron BA.5 and BF.7 variants after Omicron breakthrough infection in young individuals. TRIAL REGISTRATION This study is a purely observational study.
Collapse
Affiliation(s)
- Yichuan Yao
- Department of Ophthalmology, The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Yunru Yang
- Department of Ophthalmology, The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Qiqin Wu
- Department of Ophthalmology, The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Mengyao Liu
- Department of Ophthalmology, The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Wei Bao
- Institute of Public Health Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Qiutong Wang
- The Hospital of USTC, University of Science and Technology of China, Hefei, 230026, China
| | - Meijun Cheng
- Hefei National Research Center for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, CAS Key Laboratory of Innate Immunity and Chronic Disease, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230026, China
| | - Yunuo Chen
- Department of Ophthalmology, The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Yiting Yu
- Institute of Advanced Technology, University of Science and Technology of China, Hefei, 230031, China
| | - Yuan Cai
- Department of Ophthalmology, The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Mei Zhang
- Department of Ophthalmology, The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- Hefei National Research Center for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, CAS Key Laboratory of Brain Function and Disease, CAS Key Laboratory of Innate Immunity and Chronic Disease, Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, 230026, China
| | - Jingxue Yao
- Department of Ophthalmology, The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Hongliang He
- Department of Ophthalmology, The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Changjiang Jin
- The Hospital of USTC, University of Science and Technology of China, Hefei, 230026, China
| | - Changcheng Zheng
- Department of Ophthalmology, The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
- School of Life Science, West Campus University of Science and Technology of China, Room 718, No.443 Huangshan Road, Hefei, 230022, Anhui, China.
| | - Tengchuan Jin
- Department of Ophthalmology, The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
- School of Life Science, West Campus University of Science and Technology of China, Room 718, No.443 Huangshan Road, Hefei, 230022, Anhui, China.
| | - Dali Tong
- Department of Ophthalmology, The First Affiliated Hospital of USTC, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
- School of Life Science, West Campus University of Science and Technology of China, Room 718, No.443 Huangshan Road, Hefei, 230022, Anhui, China.
| |
Collapse
|
9
|
Debbag R, Rudin D, Ceddia F, Watkins J. The Impact of Vaccination on COVID-19, Influenza, and Respiratory Syncytial Virus-Related Outcomes: A Narrative Review. Infect Dis Ther 2025; 14:63-97. [PMID: 39739199 PMCID: PMC11724835 DOI: 10.1007/s40121-024-01079-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/06/2024] [Indexed: 01/02/2025] Open
Abstract
Vaccination represents a core preventive strategy for public health, with interrelated and multifaceted effects across health and socioeconomic domains. Beyond immediate disease prevention, immunization positively influences downstream health outcomes by mitigating complications of preexisting comorbidities and promoting healthy aging. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), influenza virus, and respiratory syncytial virus (RSV) are common respiratory viruses responsible for broad societal cost and substantial morbidity and mortality, particularly among at-risk individuals, including older adults and people with frailty or certain comorbid conditions. In this narrative review, we summarize the overall impact of vaccination for these 3 viruses, focusing on mRNA vaccines, each of which exhibits unique patterns of infection, risk, and transmission dynamics, but collectively represent a target for preventive strategies. Vaccines for COVID-19 (caused by SARS-CoV-2) and influenza are effective against the most severe outcomes, such as hospitalization and death; these vaccines represent the most potent and cost-effective interventions for the protection of population and individual health against COVID-19 and influenza, particularly for older adults and those with comorbid conditions. Based on promising results of efficacy for the prevention of RSV-associated lower respiratory tract disease, the first RSV vaccines were approved in 2023. Immunization strategies should account for various factors leading to poor uptake, including vaccine hesitancy, socioeconomic barriers to access, cultural beliefs, and lack of knowledge of vaccines and disease states. Coadministration of vaccines and combination vaccines, such as multicomponent mRNA vaccines, offer potential advantages in logistics and delivery, thus improving uptake and reducing barriers to adoption of new vaccines. The success of the mRNA vaccine platform was powerfully demonstrated during the COVID-19 pandemic; these and other new approaches show promise as a means to overcome existing challenges in vaccine development and to sustain protection against viral changes over time.A graphical abstract and video abstract is available with this article.
Collapse
Affiliation(s)
- Roberto Debbag
- Latin American Vaccinology Society, Buenos Aires, Argentina
| | | | | | - John Watkins
- Department of Population Medicine, Cardiff University, Cardiff, UK.
| |
Collapse
|
10
|
Lee A, Davido B, Beck E, Demont C, Joshi K, Kohli M, Maschio M, Uhart M, El Mouaddin N. Substantial reduction in the clinical and economic burden of disease following variant-adapted mRNA COVID-19 vaccines in immunocompromised patients in France. Hum Vaccin Immunother 2024; 20:2423474. [PMID: 39540209 PMCID: PMC11572258 DOI: 10.1080/21645515.2024.2423474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/07/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
An evaluation was conducted to predict the economic and clinical burden of vaccinating all immunocompromised (IC) individuals aged ≥30 years with mRNA-1273 variant-adapted COVID-19 vaccines versus BNT162b2 variant-adapted vaccines in Fall 2023 and Spring 2024 in France. The number of symptomatic SARS-CoV-2 infections, hospitalizations or deaths due to COVID-19, and long COVID cases, costs and quality-adjusted life years (QALYs) were estimated using a static decision-analytic model. Predicted vaccine effectiveness (VE) were based on real-world data from the original and BA.4/5 variant-adapted vaccines, suggesting higher protection against infection and hospitalization with mRNA-1273 vaccines. VE estimates were combined with COVID-19 incidence and probability of COVID-19 severe outcomes. Uncertainty surrounding VE, vaccine coverage, infection incidence, hospitalization and mortality rates, costs and QALYs were evaluated in sensitivity analyses. In an ideal situation where 100% coverage is achieved, the mRNA-1273 variant-adapted vaccine is predicted to prevent an additional 3,882 infections, 357 hospitalizations, 81 deaths, and 326 long COVID cases when compared to BNT162b2 variant-adapted vaccines in 230,000 IC individuals. This translates to €10.1 million cost-savings from a societal perspective and 645 QALYs gained. Results were consistent across all analyses and most sensitive to variations surrounding VE and coverage. These findings highlight the importance of increasing vaccine coverage, and ability to induce higher levels of protection with mRNA-1273 formulations in this vulnerable population.
Collapse
Affiliation(s)
- Amy Lee
- Quadrant Health Economics Inc, Cambridge, Ontario, Canada
| | - Benjamin Davido
- Maladies Infectieuses, Hôpital Universitaire Raymond-Poincaré, AP-HP Université Paris Saclay, Garches, France
| | - Ekkehard Beck
- Health Economics and Outcomes Research, Moderna Inc, Cambridge, MA, USA
| | - Clarisse Demont
- Health Economics and Outcomes Research, Moderna France, Paris, France
| | - Keya Joshi
- Health Economics and Outcomes Research, Moderna Inc, Cambridge, MA, USA
| | - Michele Kohli
- Quadrant Health Economics Inc, Cambridge, Ontario, Canada
| | | | - Mathieu Uhart
- Health Economics and Outcomes Research, Moderna France, Paris, France
| | - Nadia El Mouaddin
- Health Economics and Outcomes Research, Moderna France, Paris, France
| |
Collapse
|
11
|
Rajput V, Pramanik R, Nannaware K, Malik V, Matra S, Kumar S, Joshi S, Kadam P, Bhalerao U, Tupekar M, Deshpande D, Shah P, Sangewar P, Gogate N, Boargaonkar R, Patil D, Kale S, Bhalerao A, Jain N, Shashidhara LS, Kamble S, Dastager S, Karmodiya K, Dharne M. Wastewater surveillance in post-omicron silent phase uncovers silent waves and cryptic transmission of SARS-CoV-2 variants; a yearlong study in Western India. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 955:176833. [PMID: 39396788 DOI: 10.1016/j.scitotenv.2024.176833] [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: 05/22/2024] [Revised: 09/22/2024] [Accepted: 10/07/2024] [Indexed: 10/15/2024]
Abstract
Due to reduced clinical testing and evolving monitoring challenges, tracking the emergence and evolution of SARS-CoV-2 variants has become increasingly complex. To address this gap, we investigated the utility of wastewater-based epidemiology (WBE) as a complementary tool for SARS-CoV-2 variant surveillance in sewage treatment plants (STPs) across Pune, India. We analyzed 1128 wastewater samples collected between May 2022 and May 2023, using Illumina and nanopore sequencing techniques for robust detection and variant characterization. The study revealed critical findings, including "silent waves" with elevated viral load despite minimal clinical cases, suggesting potential cryptic transmission. These silent waves aligned with the dominance of Omicron BA.2 in June-July 2022 and emergence of the recombinant XBB clade in December 2022. Importantly, sequencing detected XBB lineages 130-253 days before their initial clinical identification, demonstrating its significant advantage in early variant detection. Furthermore, wastewater analysis revealed a higher degree of lineage diversity compared to clinical data, indicating its ability to capture a broader spectrum of circulating variants. The BA.2.86.X was identified 103 days prior to its clinical detection in Pune, highlighting WBE's remarkable lead time. Surprisingly, BF.7.X and BQ.X fragments were also detected in wastewater but not yet reported clinically. These findings demonstrate the remarkable value of WBE as an early warning tool for SARS-CoV-2 variants ahead of time. By revealing silent waves, enabling early variant detection, and capturing a broader viral spectrum, WBE effort could empower public health officials to make informed decisions and implement effective strategies to mitigate future waves, especially in contexts with declining clinical testing.
Collapse
Affiliation(s)
- Vinay Rajput
- National Collection of Industrial Microorganisms (NCIM), Biochemical Sciences Division, CSIR-National Chemical Laboratory (NCL), Pune 411008, Maharashtra, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Rinka Pramanik
- National Collection of Industrial Microorganisms (NCIM), Biochemical Sciences Division, CSIR-National Chemical Laboratory (NCL), Pune 411008, Maharashtra, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Kiran Nannaware
- National Collection of Industrial Microorganisms (NCIM), Biochemical Sciences Division, CSIR-National Chemical Laboratory (NCL), Pune 411008, Maharashtra, India
| | - Vinita Malik
- National Collection of Industrial Microorganisms (NCIM), Biochemical Sciences Division, CSIR-National Chemical Laboratory (NCL), Pune 411008, Maharashtra, India
| | - Sejal Matra
- National Collection of Industrial Microorganisms (NCIM), Biochemical Sciences Division, CSIR-National Chemical Laboratory (NCL), Pune 411008, Maharashtra, India
| | - Shubham Kumar
- National Collection of Industrial Microorganisms (NCIM), Biochemical Sciences Division, CSIR-National Chemical Laboratory (NCL), Pune 411008, Maharashtra, India
| | - Sai Joshi
- National Collection of Industrial Microorganisms (NCIM), Biochemical Sciences Division, CSIR-National Chemical Laboratory (NCL), Pune 411008, Maharashtra, India
| | - Pradnya Kadam
- Department of Biology, Indian Institute of Science Education and Research (IISER), Pune 41108, Maharashtra, India
| | - Unnati Bhalerao
- Department of Biology, Indian Institute of Science Education and Research (IISER), Pune 41108, Maharashtra, India
| | - Manisha Tupekar
- Department of Biology, Indian Institute of Science Education and Research (IISER), Pune 41108, Maharashtra, India
| | - Dipti Deshpande
- Department of Biology, Indian Institute of Science Education and Research (IISER), Pune 41108, Maharashtra, India
| | - Priyanki Shah
- The Pune Knowledge Cluster (PKC), Savitribai Phule Pune University (SPPU), Pune, Maharashtra, India
| | - Poornima Sangewar
- The Pune Knowledge Cluster (PKC), Savitribai Phule Pune University (SPPU), Pune, Maharashtra, India
| | - Niharika Gogate
- The Pune Knowledge Cluster (PKC), Savitribai Phule Pune University (SPPU), Pune, Maharashtra, India
| | | | - Dhawal Patil
- Ecosan Services Foundation (ESF), Pune 411030, Maharashtra, India
| | - Saurabh Kale
- Ecosan Services Foundation (ESF), Pune 411030, Maharashtra, India
| | - Asim Bhalerao
- Fluid Robotics Private Limited (FRPL), Pune 411052, Maharashtra, India
| | - Nidhi Jain
- Fluid Robotics Private Limited (FRPL), Pune 411052, Maharashtra, India
| | - L S Shashidhara
- Department of Biology, Indian Institute of Science Education and Research (IISER), Pune 41108, Maharashtra, India; The Pune Knowledge Cluster (PKC), Savitribai Phule Pune University (SPPU), Pune, Maharashtra, India; National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research Bellary Road, Bangalore 560065, Karnataka, India
| | - Sanjay Kamble
- Chemical Engineering and Process Development (CEPD) Division, CSIR-National Chemical Laboratory, Pune 411008, Maharashtra, India
| | - Syed Dastager
- National Collection of Industrial Microorganisms (NCIM), Biochemical Sciences Division, CSIR-National Chemical Laboratory (NCL), Pune 411008, Maharashtra, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Krishanpal Karmodiya
- Department of Biology, Indian Institute of Science Education and Research (IISER), Pune 41108, Maharashtra, India
| | - Mahesh Dharne
- National Collection of Industrial Microorganisms (NCIM), Biochemical Sciences Division, CSIR-National Chemical Laboratory (NCL), Pune 411008, Maharashtra, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India.
| |
Collapse
|
12
|
Laghlali G, Wiest MJ, Karadag D, Warang P, O'Konek JJ, Chang LA, Park SC, Yan V, Farazuddin M, Janczak KW, García-Sastre A, Baker JR, Wong PT, Schotsaert M. Enhanced mucosal SARS-CoV-2 immunity after heterologous intramuscular mRNA prime/intranasal protein boost vaccination with a combination adjuvant. Mol Ther 2024; 32:4448-4466. [PMID: 39489918 PMCID: PMC11638833 DOI: 10.1016/j.ymthe.2024.10.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 09/11/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
Current COVID-19 mRNA vaccines delivered intramuscularly (IM) induce effective systemic immunity, but with suboptimal immunity at mucosal sites, limiting their ability to impart sterilizing immunity. There is strong interest in rerouting immune responses induced in the periphery by parenteral vaccination to the portal entry site of respiratory viruses, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), by mucosal vaccination. We previously demonstrated the combination adjuvant, NE/IVT, consisting of a nanoemulsion (NE) and an RNA-based RIG-I agonist (IVT) induces potent systemic and mucosal immune responses in protein-based SARS-CoV-2 vaccines administered intranasally (IN). Herein, we demonstrate priming IM with mRNA followed by heterologous IN boosting with NE/IVT adjuvanted recombinant antigen induces strong mucosal and systemic antibody responses and enhances antigen-specific T cell responses in mucosa-draining lymph nodes compared to IM/IM and IN/IN prime/boost regimens. While all regimens induced cross-neutralizing antibodies against divergent variants and sterilizing immunity in the lungs of challenged mice, mucosal vaccination, either as homologous prime/boost or heterologous IN boost after IM mRNA prime, was required to impart sterilizing immunity in the upper respiratory tract. Our data demonstrate the benefit of hybrid regimens whereby strong immune responses primed via IM vaccination are rerouted by IN vaccination to mucosal sites to provide optimal protection against SARS-CoV-2.
Collapse
MESH Headings
- Animals
- SARS-CoV-2/immunology
- Mice
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- COVID-19/prevention & control
- COVID-19/immunology
- Administration, Intranasal
- Immunity, Mucosal
- Antibodies, Viral/immunology
- Injections, Intramuscular
- Female
- Immunization, Secondary
- Humans
- Antibodies, Neutralizing/immunology
- Adjuvants, Immunologic/administration & dosage
- mRNA Vaccines/immunology
- Vaccination/methods
- Adjuvants, Vaccine/administration & dosage
- Spike Glycoprotein, Coronavirus/immunology
- Spike Glycoprotein, Coronavirus/genetics
Collapse
Affiliation(s)
- Gabriel Laghlali
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pharmaceutics, Ghent University, Ghent, Belgium
| | - Matthew J Wiest
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Dilara Karadag
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Prajakta Warang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica J O'Konek
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA; Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lauren A Chang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seok-Chan Park
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vivian Yan
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mohammad Farazuddin
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA; Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Katarzyna W Janczak
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA; Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James R Baker
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA; Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Pamela T Wong
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan Medical School, Ann Arbor, MI, USA; Mary H. Weiser Food Allergy Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Michael Schotsaert
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
13
|
Zhang J, Xu Y, Chen M, Wang S, Lin G, Huang Y, Yang C, Yang Y, Song Y. Spatial Engineering of Heterotypic Antigens on a DNA Framework for the Preparation of Mosaic Nanoparticle Vaccines with Enhanced Immune Activation against SARS-CoV-2 Variants. Angew Chem Int Ed Engl 2024; 63:e202412294. [PMID: 39030890 DOI: 10.1002/anie.202412294] [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: 07/01/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/22/2024]
Abstract
Mosaic nanoparticle vaccines with heterotypic antigens exhibit broad-spectrum antiviral capabilities, but the impact of antigen proportions and distribution patterns on vaccine-induced immunity remains largely unexplored. Here, we present a DNA nanotechnology-based strategy for spatially assembling heterotypic antigens to guide the rational design of mosaic nanoparticle vaccines. By utilizing two aptamers with orthogonal selectivity for the original SARS-CoV-2 spike trimer and Omicron receptor-binding domain (RBD), along with a DNA soccer-ball framework, we precisely manipulate the spacing, stoichiometry, and overall distribution of heterotypic antigens to create mosaic nanoparticles with average, bipolar, and unipolar antigen distributions. Systematic in vitro and in vivo immunological investigations demonstrate that 30 heterotypic antigens in equivalent proportions, with an average distribution, lead to higher production of broad-spectrum neutralizing antibodies compared to the bipolar and unipolar distributions. Furthermore, the precise assembly utilizing our developed methodology reveals that a mere increment of five Omicron RBD antigens on a nanoparticle (from 15 to 20) not only diminishes neutralization against the Omicron variant but also triggers excessive inflammation. This work provides a unique perspective on the rational design of mosaic vaccines by highlighting the significance of the spatial placement and proportion of heterotypic antigens in their structure-activity mechanisms.
Collapse
Affiliation(s)
- Jialu Zhang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yunyun Xu
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Mingying Chen
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| | - Shengwen Wang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
- College of Chemistry and Materials Science, Shanghai Normal University, Shanghai, 200234, China
| | - Guihong Lin
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| | - Yihao Huang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yang Yang
- Institute of Molecular Medicine and Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, State Key Laboratory of Oncogenes and Related Genes, Department of Laboratory Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yanling Song
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, the Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Xiamen University, Xiamen, Fujian, 361005, China
| |
Collapse
|
14
|
Pardi N, Krammer F. mRNA vaccines for infectious diseases - advances, challenges and opportunities. Nat Rev Drug Discov 2024; 23:838-861. [PMID: 39367276 DOI: 10.1038/s41573-024-01042-y] [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] [Accepted: 08/27/2024] [Indexed: 10/06/2024]
Abstract
The concept of mRNA-based vaccines emerged more than three decades ago. Groundbreaking discoveries and technological advancements over the past 20 years have resolved the major roadblocks that initially delayed application of this new vaccine modality. The rapid development of nucleoside-modified COVID-19 mRNA vaccines demonstrated that this immunization platform is easy to develop, has an acceptable safety profile and can be produced at a large scale. The flexibility and ease of antigen design have enabled mRNA vaccines to enter development for a wide range of viruses as well as for various bacteria and parasites. However, gaps in our knowledge limit the development of next-generation mRNA vaccines with increased potency and safety. A deeper understanding of the mechanisms of action of mRNA vaccines, application of novel technologies enabling rational antigen design, and innovative vaccine delivery strategies and vaccination regimens will likely yield potent novel vaccines against a wide range of pathogens.
Collapse
Affiliation(s)
- Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
15
|
Cobey S. Vaccination against rapidly evolving pathogens and the entanglements of memory. Nat Immunol 2024; 25:2015-2023. [PMID: 39384979 DOI: 10.1038/s41590-024-01970-2] [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: 06/12/2024] [Accepted: 08/26/2024] [Indexed: 10/11/2024]
Abstract
Immune memory determines infection risk and responses to future infections and vaccinations over potentially decades of life. Despite its centrality, the dynamics of memory to antigenically variable pathogens remains poorly understood. This Review examines how past exposures shape B cell responses to vaccinations with influenza and SARS-CoV-2. An overriding feature of vaccinations with these pathogens is the recall of primary responses, often termed 'imprinting' or 'original antigenic sin'. These recalled responses can inhibit the generation of new responses unless some incompletely defined conditions are met. Depending on the context, immune memory can increase or decrease the total neutralizing antibody response to variant antigens, with apparent consequences for protection. These effects are easier to measure experimentally than epidemiologically, but there is evidence that both early and recent exposures influence vaccine effectiveness. A few immunological interactions between adaptive immune responses and antigens might explain the seemingly discrepant effects of memory. Overall, the complex observations point to a need for more quantitative approaches to integrate high-dimensional immune data from populations with diverse exposure histories. Such approaches could help identify optimal vaccination strategies against antigenically diverse pathogens.
Collapse
Affiliation(s)
- Sarah Cobey
- Department of Ecology and Evolution, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
16
|
Gram MA, Thiesson EM, Pihlström N, Perälä J, Poukka E, Leino T, Ljung R, Andersson NW, Hviid A. Comparative effectiveness of bivalent BA.4-5 or BA.1 mRNA booster vaccines among immunocompromised individuals across three Nordic countries: A nationwide cohort study. J Infect 2024; 89:106261. [PMID: 39218308 DOI: 10.1016/j.jinf.2024.106261] [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: 06/20/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVES To estimate the effectiveness and waning of the bivalent BA.4-5 or BA.1 mRNA booster vaccine against Covid-19-related hospitalization and death in immunocompromised individuals. METHODS Nationwide analyses across Nordic countries from 1 September 2022 to 31 October 2023 using a matched cohort design. Individuals boosted with a BA.4-5 or BA.1 vaccine were matched 1:1 with unboosted individuals. The outcomes of interest were country-combined vaccine effectiveness (VE) estimates against Covid-19-related hospitalization and death at day 270 of follow-up. Waning was assessed in 45-day intervals. RESULTS A total of 352,762 BA.4-5 and 191,070 BA.1 booster vaccine doses were included. At day 270, the comparative VE against Covid-19-related hospitalization was 34.2% (95% CI, 7.1% to 61.3%) for the bivalent BA.4-5 vaccine and 42.6% (95% CI, 31.3% to 53.9%) for the BA.1 vaccine compared with matched unboosted. The comparative VE against Covid-19-related death was 53.9% (95% CI, 38.6% to 69.3%) for the bivalent BA.4-5 vaccine and 57.9% (95% CI, 48.5% to 67.4%) for the BA.1 vaccine. CONCLUSIONS In immunocompromised individuals, vaccination with bivalent BA.4-5 or BA.1 booster lowered the risk of Covid-19-related hospitalization and death over a follow-up period of 9 months. The effectiveness was highest during the first months since vaccination with subsequent gradual waning.
Collapse
Affiliation(s)
- Mie Agermose Gram
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark.
| | | | - Nicklas Pihlström
- Division of Licensing, Swedish Medical Products Agency, Uppsala, Sweden
| | - Jori Perälä
- Infectious Disease Control and Vaccinations Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Eero Poukka
- Infectious Disease Control and Vaccinations Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland; Department of Public Health, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tuija Leino
- Infectious Disease Control and Vaccinations Unit, Department of Health Security, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Rickard Ljung
- Division of Use and Information, Swedish Medical Products Agency, Uppsala, Sweden; Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Anders Hviid
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark; Pharmacovigilance Research Center, Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
17
|
Choi YS, Ryu S, Kim RK, Chiara A, Baek S, Nam H, Park E, Kim EK, Choe YJ, Kwon D, Choi WS. Effectiveness of the Bivalent mRNA COVID-19 Vaccine for Preventing Critical Infection From the SARS-CoV-2 Omicron Variant in the Republic of Korea. J Korean Med Sci 2024; 39:e258. [PMID: 39355951 PMCID: PMC11444818 DOI: 10.3346/jkms.2024.39.e258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/18/2024] [Indexed: 10/03/2024] Open
Abstract
BACKGROUND This retrospective observational matched cohort study assessed the differences in critical infections caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) during the omicron-predominant period of the coronavirus disease 2019 (COVID-19) pandemic. We evaluated the vaccine effectiveness of bivalent mRNA vaccine compared to unvaccinated individuals. METHODS We collected COVID-19 case data from the Korean COVID-19 vaccine effectiveness cohort. We calculated the probability of critical COVID-19 cases by comparing the vaccinated and unvaccinated groups. RESULTS The risk of being critically infected due to SAR-CoV-2 infection was 5.96 times higher (95% confidence interval, 5.63-6.38) among older individuals who were unvaccinated compared to those who received the bivalent COVID-19 vaccine. CONCLUSION Our findings indicate that the bivalent vaccine reduces the disease burden of the SARS-CoV-2 omicron variant, particularly among the older population. Further studies are warranted to determine the effectiveness of booster doses of vaccines for SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Young-Sook Choi
- Director for Epidemiological Investigation Analysis, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Sukhyun Ryu
- Department of Preventive Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | - Achangwa Chiara
- Department of Preventive Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Soojin Baek
- Director for Epidemiological Investigation Analysis, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Hojin Nam
- Director for Epidemiological Investigation Analysis, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Eunkyung Park
- Director for Epidemiological Investigation Analysis, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Eun Kyoung Kim
- Director for Epidemiological Investigation Analysis, Korea Disease Control and Prevention Agency, Cheongju, Korea
| | - Young June Choe
- Department of Pediatrics, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea
| | - Donghyok Kwon
- Director for Epidemiological Investigation Analysis, Korea Disease Control and Prevention Agency, Cheongju, Korea.
| | - Won Suk Choi
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Ansan Hospital, Korea University College of Medicine, Seoul, Korea.
| |
Collapse
|
18
|
Figueroa AL, Azzi JR, Eghtesad B, Priddy F, Stolman D, Siangphoe U, Leony Lasso I, de Windt E, Girard B, Zhou H, Miller JM, Das R. Safety and Immunogenicity of the mRNA-1273 Coronavirus Disease 2019 Vaccine in Solid Organ Transplant Recipients. J Infect Dis 2024; 230:e591-e600. [PMID: 38513368 PMCID: PMC11420796 DOI: 10.1093/infdis/jiae140] [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: 09/12/2023] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/23/2024] Open
Abstract
BACKGROUND Solid organ transplant recipients (SOTRs) are at high risk for severe COVID-19. METHODS This open-label, phase 3b trial evaluated mRNA-1273 in 137 kidney and 77 liver SOTRs and 20 immunocompetent participants. In part A, SOTRs received three 100-µg doses of mRNA-1273; immunocompetent participants received 2 doses. In part B, an additional 100-µg dose was offered ≥4 months after the primary series. Here, we report interim trial results. RESULTS mRNA-1273 was well-tolerated in SOTRs. Four serious adverse events were considered vaccine related by the investigator in 3 SOTRs with preexisting comorbidities. No vaccine-related biopsy-proven organ rejection events or deaths were reported. mRNA-1273 elicited modest neutralizing antibody responses after dose 2 and improved responses after dose 3 in SOTRs. Post-dose 3 responses among liver SOTRs were comparable to post-dose 2 responses in immunocompetent participants. Post-additional dose responses were increased in SOTRs, regardless of primary series vaccination. In liver SOTRs, post-additional dose responses were ∼3-fold higher versus post-dose 2 but lower than immunocompetent participant responses. Most kidney SOTRs received multiple immunosuppressants and had reduced antibody responses versus liver SOTRs. CONCLUSIONS mRNA-1273 was well-tolerated, and dose 3 and the additional dose improved antibody responses among SOTRs. CLINICAL TRIALS REGISTRATION NCT04860297.
Collapse
Affiliation(s)
| | - Jamil R Azzi
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Androsavich JR. Frameworks for transformational breakthroughs in RNA-based medicines. Nat Rev Drug Discov 2024; 23:421-444. [PMID: 38740953 DOI: 10.1038/s41573-024-00943-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2024] [Indexed: 05/16/2024]
Abstract
RNA has sparked a revolution in modern medicine, with the potential to transform the way we treat diseases. Recent regulatory approvals, hundreds of new clinical trials, the emergence of CRISPR gene editing, and the effectiveness of mRNA vaccines in dramatic response to the COVID-19 pandemic have converged to create tremendous momentum and expectation. However, challenges with this relatively new class of drugs persist and require specialized knowledge and expertise to overcome. This Review explores shared strategies for developing RNA drug platforms, including layering technologies, addressing common biases and identifying gaps in understanding. It discusses the potential of RNA-based therapeutics to transform medicine, as well as the challenges associated with improving applicability, efficacy and safety profiles. Insights gained from RNA modalities such as antisense oligonucleotides (ASOs) and small interfering RNAs are used to identify important next steps for mRNA and gene editing technologies.
Collapse
Affiliation(s)
- John R Androsavich
- RNA Accelerator, Pfizer Inc, Cambridge, MA, USA.
- Ginkgo Bioworks, Boston, MA, USA.
| |
Collapse
|
20
|
Song S, Madewell ZJ, Liu M, Miao Y, Xiang S, Huo Y, Sarkar S, Chowdhury A, Longini IM, Yang Y. A systematic review and meta-analysis on the effectiveness of bivalent mRNA booster vaccines against Omicron variants. Vaccine 2024; 42:3389-3396. [PMID: 38653679 DOI: 10.1016/j.vaccine.2024.04.049] [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: 01/22/2024] [Revised: 04/02/2024] [Accepted: 04/15/2024] [Indexed: 04/25/2024]
Abstract
BACKGROUND A global shift to bivalent mRNA vaccines is ongoing to counterbalance the diminishing effectiveness of the original monovalent vaccines due to the evolution of SARS-CoV-2 variants, yet substantial variation in the bivalent vaccine effectiveness (VE) exists across studies and a complete picture is lacking. METHODS We searched papers evaluating absolute or relative effectiveness of SARS-CoV-2 BA.1 type or BA.4/5 type bivalent mRNA vaccines on eight publication databases published from September 1st, 2022, to November 8th, 2023. Pooled VE against Omicron-associated infection and severe events (hospitalization and/or death) was estimated in reference to unvaccinated, ≥2 original monovalent doses, and ≥ 3 original monovalent doses. RESULTS From 630 citations identified, 28 studies were included, involving 55,393,303 individuals. Bivalent boosters demonstrated higher effectiveness against symptomatic or any infection for all ages combined, with an absolute VE of 53.5 % (95 % CI: -22.2-82.3 %) when compared to unvaccinated and relative VE of 30.8 % (95 % CI: 22.5-38.2 %) and 28.4 % (95 % CI: 10.2-42.9 %) when compared to ≥ 2 and ≥ 3 original monovalent doses, respectively. The corresponding VE estimates for adults ≥ 60 years old were 22.5 % (95 % CI: 16.8-39.8 %), 31.4 % (95 % CI: 27.7-35.0 %), and 30.6 % (95 % CI: -13.2-57.5 %). Pooled bivalent VE estimates against severe events were higher, 72.9 % (95 % CI: 60.5-82.4 %), 57.6 % (95 % CI: 42.4-68.8 %), and 62.1 % (95 % CI: 54.6-68.3 %) for all ages, and 72.0 % (95 % CI: 51.4-83.9 %), 63.4 % (95 % CI: 41.0-77.3 %), and 60.7 % (95 % CI: 52.4-67.6 %) for adults ≥ 60 years old, compared to unvaccinated, ≥2 original monovalent doses, and ≥ 3 original monovalent doses, respectively. CONCLUSIONS The bivalent boosters demonstrated superior protection against severe outcomes than the original monovalent boosters across age groups, highlighting the critical need for improving vaccine coverage, especially among the vulnerable older subpopulation.
Collapse
Affiliation(s)
- Shangchen Song
- Department of Biostatistics, College of Public Health and health Professions, University of Florida, Gainesville, FL, USA
| | - Zachary J Madewell
- Department of Biostatistics, College of Public Health and health Professions, University of Florida, Gainesville, FL, USA
| | - Mingjin Liu
- Department of Statistics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Yu Miao
- Department of Statistics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Shaolin Xiang
- Department of Statistics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Yanan Huo
- Gilead Sciences, Inc, Foster City, CA, USA
| | - Shoumi Sarkar
- Department of Biostatistics, College of Public Health and health Professions, University of Florida, Gainesville, FL, USA
| | - Amily Chowdhury
- Department of Computer Science, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Ira M Longini
- Department of Biostatistics, College of Public Health and health Professions, University of Florida, Gainesville, FL, USA
| | - Yang Yang
- Department of Statistics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA.
| |
Collapse
|
21
|
Burri DJ, Renz L, Mueller M, Pagallies F, Klinkhardt U, Amann R, Derouazi M. Novel Multi-Antigen Orf-Virus-Derived Vaccine Elicits Protective Anti-SARS-CoV-2 Response in Monovalent and Bivalent Formats. Vaccines (Basel) 2024; 12:490. [PMID: 38793740 PMCID: PMC11126134 DOI: 10.3390/vaccines12050490] [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: 03/27/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/26/2024] Open
Abstract
Prime-2-CoV_Beta is a novel Orf virus (ORFV)-based COVID-19 vaccine candidate expressing both the nucleocapsid and spike proteins of SARS-CoV-2 with the receptor-binding domain (RBD) of the Beta strain. This candidate was shown to be safe and immunogenic in a first-in-human Phase I clinical trial. With the shift in the immune landscape toward the Omicron variant and the widespread vaccine- and/or infection-derived immunity, further pre-clinical research was needed to characterize Prime-2-CoV. Here, we quantified the humoral and cellular response to Prime-2-CoV_Beta in pre-immunized mice and compared the protective efficacy of mono- and bivalent variant-based Prime-2-CoV vaccine candidates in hamsters. Prime-2-CoV_Beta induced robust humoral and cellular immune responses in naïve animals but did not further boost antibody titers in the tested setting when given as repeat booster at short interval. We furthermore showed that Prime-2-CoV_Beta-based mono- and bivalent immunization strategies produced comparable immunogenicity and protection from infection. Our results highlight the potential of the Orf virus as a vaccine platform against SARS-CoV-2 and potentially other infectious viruses.
Collapse
Affiliation(s)
- Dominique Julien Burri
- Speransa Therapeutics, Frankfurt am Main, 60327 Frankfurt, Germany; (D.J.B.); (L.R.); (U.K.)
| | - Louis Renz
- Speransa Therapeutics, Frankfurt am Main, 60327 Frankfurt, Germany; (D.J.B.); (L.R.); (U.K.)
| | - Melanie Mueller
- Institute of Immunology, University Hospital of Tübingen, 72016 Tübingen, Germany; (M.M.)
| | - Felix Pagallies
- Institute of Immunology, University Hospital of Tübingen, 72016 Tübingen, Germany; (M.M.)
| | - Ute Klinkhardt
- Speransa Therapeutics, Frankfurt am Main, 60327 Frankfurt, Germany; (D.J.B.); (L.R.); (U.K.)
| | - Ralf Amann
- Institute of Immunology, University Hospital of Tübingen, 72016 Tübingen, Germany; (M.M.)
| | - Madiha Derouazi
- Speransa Therapeutics, Frankfurt am Main, 60327 Frankfurt, Germany; (D.J.B.); (L.R.); (U.K.)
| |
Collapse
|
22
|
Plumb ID, Briggs Hagen M, Wiegand R, Dumyati G, Myers C, Harland KK, Krishnadasan A, James Gist J, Abedi G, Fleming-Dutra KE, Chea N, Lee JE, Kellogg M, Edmundson A, Britton A, Wilson LE, Lovett SA, Ocampo V, Markus TM, Smithline HA, Hou PC, Lee LC, Mower W, Rwamwejo F, Steele MT, Lim SC, Schrading WA, Chinnock B, Beiser DG, Faine B, Haran JP, Nandi U, Chipman AK, LoVecchio F, Eucker S, Femling J, Fuller M, Rothman RE, Curlin ME, Talan DA, Mohr NM. Effectiveness of a bivalent mRNA vaccine dose against symptomatic SARS-CoV-2 infection among U.S. Healthcare personnel, September 2022-May 2023. Vaccine 2024; 42:2543-2552. [PMID: 37973512 PMCID: PMC10994739 DOI: 10.1016/j.vaccine.2023.10.072] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/19/2023]
Abstract
BACKGROUND Bivalent mRNA vaccines were recommended since September 2022. However, coverage with a recent vaccine dose has been limited, and there are few robust estimates of bivalent VE against symptomatic SARS-CoV-2 infection (COVID-19). We estimated VE of a bivalent mRNA vaccine dose against COVID-19 among eligible U.S. healthcare personnel who had previously received monovalent mRNA vaccine doses. METHODS We conducted a case-control study in 22 U.S. states, and enrolled healthcare personnel with COVID-19 (case-participants) or without COVID-19 (control-participants) during September 2022-May 2023. Participants were considered eligible for a bivalent mRNA dose if they had received 2-4 monovalent (ancestral-strain) mRNA vaccine doses, and were ≥67 days after the most recent vaccine dose. We estimated VE of a bivalent mRNA dose using conditional logistic regression, accounting for matching by region and four-week calendar period. We adjusted estimates for age group, sex, race and ethnicity, educational level, underlying health conditions, community COVID-19 exposure, prior SARS-CoV-2 infection, and days since the last monovalent mRNA dose. RESULTS Among 3,647 healthcare personnel, 1,528 were included as case-participants and 2,119 as control-participants. Participants received their last monovalent mRNA dose a median of 404 days previously; 1,234 (33.8%) also received a bivalent mRNA dose a median of 93 days previously. Overall, VE of a bivalent dose was 34.1% (95% CI, 22.6%-43.9%) against COVID-19 and was similar by product, days since last monovalent dose, number of prior doses, age group, and presence of underlying health conditions. However, VE declined from 54.8% (95% CI, 40.7%-65.6%) after 7-59 days to 21.6% (95% CI 5.6%-34.9%) after ≥60 days. CONCLUSIONS Bivalent mRNA COVID-19 vaccines initially conferred approximately 55% protection against COVID-19 among U.S. healthcare personnel. However, protection waned after two months. These findings indicate moderate initial protection against symptomatic SARS-CoV-2 infection by remaining up-to-date with COVID-19 vaccines.
Collapse
Affiliation(s)
- Ian D Plumb
- National Center for Immunizations and Respiratory Diseases, Centers for Disease Control & Prevention, Atlanta, GA, USA.
| | - Melissa Briggs Hagen
- National Center for Immunizations and Respiratory Diseases, Centers for Disease Control & Prevention, Atlanta, GA, USA
| | - Ryan Wiegand
- National Center for Immunizations and Respiratory Diseases, Centers for Disease Control & Prevention, Atlanta, GA, USA
| | - Ghinwa Dumyati
- University of Rochester Medical Center, Rochester, NY, USA
| | | | | | | | - Jade James Gist
- National Center for Immunizations and Respiratory Diseases, Centers for Disease Control & Prevention, Atlanta, GA, USA
| | - Glen Abedi
- National Center for Immunizations and Respiratory Diseases, Centers for Disease Control & Prevention, Atlanta, GA, USA
| | - Katherine E Fleming-Dutra
- National Center for Immunizations and Respiratory Diseases, Centers for Disease Control & Prevention, Atlanta, GA, USA
| | - Nora Chea
- National Center for Emerging and Zoonotic Diseases, Centers for Disease Control & Prevention, USA
| | - Jane E Lee
- California Emerging Infections Program, Oakland, CA, USA
| | | | - Alexandra Edmundson
- Connecticut Emerging Infections Program, Yale School of Public Health, CT, USA
| | - Amber Britton
- Georgia Emerging Infections Program and Emory University School of Medicine, Atlanta, GA, USA
| | - Lucy E Wilson
- Maryland Emerging Infections Program, Maryland Department of Health and University of Maryland, Baltimore, MD, USA
| | | | - Valerie Ocampo
- Public Health Division, Oregon Health Authority, OR, USA
| | | | | | - Peter C Hou
- Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | - Mark T Steele
- University of Missouri-Kansas City, Kansas City, MO, USA
| | - Stephen C Lim
- University Medical Center New Orleans, LSU Health Sciences Center, New Orleans, LA, USA
| | | | | | | | | | - John P Haran
- University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Utsav Nandi
- University of Mississippi Medical Center, Jackson, MS, USA
| | | | | | | | - Jon Femling
- University of New Mexico Health Science Center, USA
| | | | - Richard E Rothman
- Department of Emergency Medicine, Johns Hopkins University, Baltimore, MD, USA
| | | | | | | |
Collapse
|
23
|
Matusali G, Vergori A, Cimini E, Mariotti D, Mazzotta V, Lepri AC, Colavita F, Gagliardini R, Notari S, Meschi S, Fusto M, Tartaglia E, Girardi E, Maggi F, Antinori A. Poor durability of the neutralizing response against XBB sublineages after a bivalent mRNA COVID-19 booster dose in persons with HIV. J Med Virol 2024; 96:e29598. [PMID: 38624044 DOI: 10.1002/jmv.29598] [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: 01/04/2024] [Revised: 03/18/2024] [Accepted: 04/01/2024] [Indexed: 04/17/2024]
Abstract
We estimated the dynamics of the neutralizing response against XBB sublineages and T cell response in persons with HIV (PWH) with previous AIDS and/or CD4 < 200/mm3 receiving the bivalent original strain/BA.4-5 booster dose in fall 2022. Samples were collected before the shot (Day 0), 15 days, 3, and 6 months after. PWH were stratified by immunization status: hybrid immunity (HI; vaccination plus COVID-19) versus nonhybrid immunity (nHI; vaccination only). Fifteen days after the booster, 16% and 30% of PWH were nonresponders in terms of anti-XBB.1.16 or anti-EG.5.1 nAbs, respectively. Three months after, a significant waning of anti-XBB.1.16, EG.5.1 and -XBB.1 nAbs was observed both in HI and nHI but nAbs in HI were higher than in nHI. Six months after both HI and nHI individuals displayed low mean levels of anti-XBB.1.16 and EG.5.1 nAbs. Regarding T cell response, IFN-γ values were stable over time and similar in HI and nHI. Our data showed that in PWH, during the prevalent circulation of the XBB.1.16, EG.5.1, and other XBB sublineages, a mRNA bivalent vaccine might not confer broad protection against them. With a view to the 2023/2024 vaccination campaign, the use of the monovalent XBB.1.5 mRNA vaccine should be urgently warranted in PWH to provide adequate protection.
Collapse
Affiliation(s)
- Giulia Matusali
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Alessandra Vergori
- Viral Immunodeficiency Unit, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Eleonora Cimini
- Immunology Unit, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Davide Mariotti
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Valentina Mazzotta
- Viral Immunodeficiency Unit, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Alessandro Cozzi Lepri
- Institute for Global Health, University College of London, Centre for Clinical Research, Epidemiology, Modeling and Evaluation (CREME), London, UK
| | - Francesca Colavita
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Roberta Gagliardini
- Viral Immunodeficiency Unit, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Stefania Notari
- Immunology Unit, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Silvia Meschi
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Marisa Fusto
- Viral Immunodeficiency Unit, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Eleonora Tartaglia
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Enrico Girardi
- Scientific Direction, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Andrea Antinori
- Viral Immunodeficiency Unit, National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| |
Collapse
|
24
|
Laghlali G, Wiest MJ, Karadag D, Warang P, O'Konek JJ, Chang LA, Park S, Farazuddin M, Landers JJ, Janczak KW, García-Sastre A, Baker JR, Wong PT, Schotsaert M. Enhanced mucosal B- and T-cell responses against SARS-CoV-2 after heterologous intramuscular mRNA prime/intranasal protein boost vaccination with a combination adjuvant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.28.587260. [PMID: 38586014 PMCID: PMC10996704 DOI: 10.1101/2024.03.28.587260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Current COVID-19 mRNA vaccines delivered intramuscularly (IM) induce effective systemic immunity, but with suboptimal immunity at mucosal sites, limiting their ability to impart sterilizing immunity. There is strong interest in rerouting immune responses induced in the periphery by parenteral vaccination to the portal entry site of respiratory viruses, such as SARS-CoV-2, by mucosal vaccination. We previously demonstrated the combination adjuvant, NE/IVT, consisting of a nanoemulsion (NE) and an RNA-based RIG-I agonist (IVT) induces potent systemic and mucosal immune responses in protein-based SARS-CoV-2 vaccines administered intranasally (IN). Herein, we demonstrate priming IM with mRNA followed by heterologous IN boosting with NE/IVT adjuvanted recombinant antigen induces strong mucosal and systemic antibody responses and enhances antigen-specific T cell responses in mucosa-draining lymph nodes compared to IM/IM and IN/IN prime/boost regimens. While all regimens induced cross-neutralizing antibodies against divergent variants and sterilizing immunity in the lungs of challenged mice, mucosal vaccination, either as homologous prime/boost or heterologous IN boost after IM mRNA prime was required to impart sterilizing immunity in the upper respiratory tract. Our data demonstrate the benefit of hybrid regimens whereby strong immune responses primed via IM vaccination are rerouted by IN vaccination to mucosal sites to provide optimal protection to SARS-CoV-2.
Collapse
|
25
|
Costa Rocha VP, Souza Machado BA, Barreto BC, Quadros HC, Santana Fernandes AM, Lima EDS, Bandeira ME, Meira CS, Moraes dos Santos Fonseca L, Erasmus J, Khandhar A, Berglund P, Reed S, José da Silva Badaró R, Pereira Soares MB. A polyvalent RNA vaccine reduces the immune imprinting phenotype in mice and induces neutralizing antibodies against omicron SARS-CoV-2. Heliyon 2024; 10:e25539. [PMID: 38370238 PMCID: PMC10869778 DOI: 10.1016/j.heliyon.2024.e25539] [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: 12/02/2023] [Revised: 01/29/2024] [Accepted: 01/29/2024] [Indexed: 02/20/2024] Open
Abstract
Immune imprinting is now evident in COVID-19 vaccinated people. This phenomenon may impair the development of effective neutralizing antibodies against variants of concern (VoCs), mainly Omicron and its subvariants. Consequently, the boost doses with bivalent vaccines have not shown a significant gain of function regarding the neutralization of Omicron. The approach to design COVID-19 vaccines must be revised to improve the effectiveness against VoCs. Here, we took advantage of the self-amplifying characteristic of RepRNA and developed a polyvalent formulation composed of mRNA from five VoCs. LION/RepRNA Polyvalent induced neutralizing antibodies in mice previously immunized with LION/RepRNA D614G and reduced the imprinted phenotype associated with low neutralization capacity of Omicron B.1.1.529 pseudoviruses. The polyvalent vaccine can be a strategy to handle the low neutralization of Omicron VoC, despite booster doses with either monovalent or bivalent vaccines.
Collapse
Affiliation(s)
- Vinicius Pinto Costa Rocha
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC, Salvador, Bahia, Brazil
- Gonçalo Moniz Institute, FIOCRUZ, Salvador, Bahia, Brazil
| | - Bruna Aparecida Souza Machado
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC, Salvador, Bahia, Brazil
- University Center SENAI CIMATEC, Salvador, Bahia, Brazil
| | | | - Helenita Costa Quadros
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC, Salvador, Bahia, Brazil
- Gonçalo Moniz Institute, FIOCRUZ, Salvador, Bahia, Brazil
| | | | - Eduarda dos Santos Lima
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC, Salvador, Bahia, Brazil
| | - Mariana Evangelista Bandeira
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC, Salvador, Bahia, Brazil
| | - Cássio Santana Meira
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC, Salvador, Bahia, Brazil
- Gonçalo Moniz Institute, FIOCRUZ, Salvador, Bahia, Brazil
| | | | | | | | | | | | - Roberto José da Silva Badaró
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC, Salvador, Bahia, Brazil
| | - Milena Botelho Pereira Soares
- SENAI Institute of Innovation (ISI) in Health Advanced Systems (CIMATEC ISI SAS), SENAI CIMATEC, Salvador, Bahia, Brazil
- Gonçalo Moniz Institute, FIOCRUZ, Salvador, Bahia, Brazil
| |
Collapse
|
26
|
Cheng MQ, Li R, Weng ZY, Song G. Relative effectiveness of bivalent COVID-19 vaccine: a systematic review and meta-analysis. Front Med (Lausanne) 2024; 10:1322396. [PMID: 38384317 PMCID: PMC10879625 DOI: 10.3389/fmed.2023.1322396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/14/2023] [Indexed: 02/23/2024] Open
Abstract
Objective The rapid development of COVID-19 bivalent vaccines (BVs) has encompassed both the original virus strains and the variant strain. However, the effectiveness of BVs is largely unknown. Therefore, we conducted a systematic review and meta-analysis of the effectiveness of BVs. Methods Literature research was conducted through PubMed, Cochrane Library, Embase, and Web of Science up until November 4, 2023. Both randomized control trials and observational studies were considered for inclusion. Pooled estimates were calculated using a random effects model. The Newcastle-Ottawa Scale (NOS) was used to assess the risk of bias in cohort and case-control studies. Results A total of 1,174 articles were reviewed and 22 eligible studies were included. All included studies were observational (15 cohort studies, 7 case-control studies). The total number of participants was 39,673,160, and the number of people vaccinated with BVs as an intervention group was 11,585,182. Two mRNA BVs were mainly involved, including the ancestral strain and the BA.1 or BA.4-5 variants. Meta-analysis results showed, compared with the monovalent vaccines (MVs), the relative effectiveness (rVE) of the BVs in COVID-19-associated infections/symptomatic infections, illnesses, hospitalizations, and deaths was 30.90% [95% confidence interval (CI), 8.43-53.37], 39.83% (95% CI, 27.34-52.32), 59.70% (95% CI, 44.08-75.32), and 72.23% (95% CI, 62.08-82.38), respectively. For those aged 50 years and older, BVs provided an additional 49.69% (95% CI, 41.44-57.94) effective protection compared with MVs. During the dominance period of the omicron XBB variant strain, BVs provided an additional 47.63% (95% CI, 27.45-67.82) effective protection compared with MVs. Conclusion Our findings show that the rVE of BVs in preventing COVID-19-associated infections, symptomatic infections, illnesses, hospitalizations, and deaths is higher compared to MVs. Particularly for people over 50 years of age and during the Omicron variant XBB dominance phase, BVs provided superior protection. Therefore, BVs may have a broader application in the prevention and control of coronaviruses variant.
Collapse
Affiliation(s)
- Meng-qun Cheng
- Department of Reproductive Medicine, The Puer People's Hospital, Pu’er, China
| | - Rong Li
- Department of Pharmacy, The Puer People's Hospital, Pu’er, China
| | - Zhi-ying Weng
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, China
| | - Gao Song
- Department of Pharmacy, The Puer People's Hospital, Pu’er, China
| |
Collapse
|
27
|
Zech F, Jung C, Jacob T, Kirchhoff F. Causes and Consequences of Coronavirus Spike Protein Variability. Viruses 2024; 16:177. [PMID: 38399953 PMCID: PMC10892391 DOI: 10.3390/v16020177] [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: 12/28/2023] [Revised: 01/20/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Coronaviruses are a large family of enveloped RNA viruses found in numerous animal species. They are well known for their ability to cross species barriers and have been transmitted from bats or intermediate hosts to humans on several occasions. Four of the seven human coronaviruses (hCoVs) are responsible for approximately 20% of common colds (hCoV-229E, -NL63, -OC43, -HKU1). Two others (SARS-CoV-1 and MERS-CoV) cause severe and frequently lethal respiratory syndromes but have only spread to very limited extents in the human population. In contrast the most recent human hCoV, SARS-CoV-2, while exhibiting intermediate pathogenicity, has a profound impact on public health due to its enormous spread. In this review, we discuss which initial features of the SARS-CoV-2 Spike protein and subsequent adaptations to the new human host may have helped this pathogen to cause the COVID-19 pandemic. Our focus is on host forces driving changes in the Spike protein and their consequences for virus infectivity, pathogenicity, immune evasion and resistance to preventive or therapeutic agents. In addition, we briefly address the significance and perspectives of broad-spectrum therapeutics and vaccines.
Collapse
Affiliation(s)
- Fabian Zech
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| | - Christoph Jung
- Institute of Electrochemistry, Ulm University, 89081 Ulm, Germany; (C.J.); (T.J.)
- Helmholtz-Institute Ulm (HIU) Electrochemical Energy Storage, 89081 Ulm, Germany
- Karlsruhe Institute of Technology (KIT), 76021 Karlsruhe, Germany
| | - Timo Jacob
- Institute of Electrochemistry, Ulm University, 89081 Ulm, Germany; (C.J.); (T.J.)
- Helmholtz-Institute Ulm (HIU) Electrochemical Energy Storage, 89081 Ulm, Germany
- Karlsruhe Institute of Technology (KIT), 76021 Karlsruhe, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany
| |
Collapse
|
28
|
Joshi K, Scholz S, Maschio M, Kohli M, Lee A, Fust K, Ultsch B, Van de Velde N, Beck E. Clinical impact and cost-effectiveness of the updated COVID-19 mRNA Autumn 2023 vaccines in Germany. J Med Econ 2024; 27:39-50. [PMID: 38050685 DOI: 10.1080/13696998.2023.2290388] [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: 10/18/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023]
Abstract
OBJECTIVES To assess the potential clinical impact and cost-effectiveness of coronavirus disease 2019 (COVID-19) mRNA vaccines updated for Autumn 2023 in adults aged ≥60 years and high-risk persons aged 30-59 years in Germany over a 1-year analytic time horizon (September 2023-August 2024). METHODS A compartmental Susceptible-Exposed-Infected-Recovered model was updated and adapted to the German market. Numbers of symptomatic infections, a number of COVID-19 related hospitalizations and deaths, costs, and quality-adjusted life-years (QALYs) gained were calculated using a decision tree model. The incremental cost-effectiveness ratio of an Autumn 2023 Moderna updated COVID-19 (mRNA-1273.815) vaccine was compared to no additional vaccination. Potential differences between the mRNA-1273.815 and the Autumn Pfizer-BioNTech updated COVID-19 (XBB.1.5 BNT162b2) vaccines, as well as societal return on investment for the mRNA-1273.815 vaccine relative to no vaccination, were also examined. RESULTS Compared to no autumn vaccination, the mRNA-1273.815 campaign is predicted to prevent approximately 1,697,900 symptomatic infections, 85,400 hospitalizations, and 4,100 deaths. Compared to an XBB.1.5 BNT162b2 campaign, the mRNA-1273.815 campaign is also predicted to prevent approximately 90,100 symptomatic infections, 3,500 hospitalizations, and 160 deaths. Across both analyses we found the mRNA-1273.815 campaign to be dominant. CONCLUSIONS The mRNA-1273.815 vaccine can be considered cost-effective relative to the XBB.1.5 BNT162b2 vaccine and highly likely to provide more benefits and save costs compared to no vaccine in Germany, and to offer high societal return on investment.
Collapse
Affiliation(s)
| | | | | | - Michele Kohli
- Quadrant Health Economics Inc, Cambridge, ON, Canada
| | - Amy Lee
- Quadrant Health Economics Inc, Cambridge, ON, Canada
| | - Kelly Fust
- Quadrant Health Economics Inc, Cambridge, ON, Canada
| | | | | | | |
Collapse
|
29
|
Bausch-Jurken M, Dawson RS, Ceddia F, Urdaneta V, Marks MA, Doi Y. A descriptive review on the real-world impact of Moderna, Inc. COVID-19 vaccines. Expert Rev Vaccines 2024; 23:914-943. [PMID: 39269429 DOI: 10.1080/14760584.2024.2402955] [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: 06/13/2024] [Revised: 08/26/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
INTRODUCTION Since the original COVID-19 vaccines were developed, abundant clinical trial and real-world evidence evaluating the efficacy, effectiveness, and safety of COVID-19 vaccines has been collected. Knowledge of the relative benefits and risks of COVID-19 vaccines is essential for building trust within target populations, ensuring they remain effectively and safely protected against an enduring infectious threat. AREAS COVERED This descriptive review discusses the benefits and risks associated with marketed Moderna, Inc. mRNA-based COVID-19 vaccines, focusing on their real-world effectiveness and safety profiles in various age groups. Adverse events of interest and potential benefits of vaccination are reviewed, including reduced risk for severe COVID-19 and long-term health outcomes, reduced economic and societal costs, and reduced risk for SARS-CoV-2 transmission. EXPERT OPINION Post-marketing safety and real-world data for Moderna, Inc. COVID-19 mRNA vaccines strongly support a positive benefit - risk profile favoring vaccination across all age groups. Although COVID-19 is no longer considered a global health pandemic, health risks associated with SARS-CoV-2 infection remain high. Concerted efforts are required to engage communities and maintain protection through vaccination. Continued surveillance of emerging variants and monitoring of vaccine safety and effectiveness are crucial for ensuring sustained protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Mary Bausch-Jurken
- Medical Affairs - Scientific Communication, Moderna, Inc, Cambridge, MA, USA
| | - Rachel S Dawson
- Medical Affairs - Scientific Communication, Moderna, Inc, Cambridge, MA, USA
| | - Francesca Ceddia
- Medical Affairs - Scientific Communication, Moderna, Inc, Cambridge, MA, USA
| | - Veronica Urdaneta
- Clinical Safety and Risk Management, Moderna, Inc, Cambridge, MA, USA
| | - Morgan A Marks
- Medical Affairs - Scientific Communication, Moderna, Inc, Cambridge, MA, USA
| | - Yohei Doi
- Departments of Microbiology and Infectious Diseases, Fujita Health University School of Medicine, Toyoake, Japan
- Division of Infectious Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
30
|
Kohli M, Maschio M, Lee A, Joshi K, Carroll S, Balogh O, Van de Velde N, Beck E. The potential clinical impact and cost-effectiveness of the updated COVID-19 mRNA Autumn 2024 vaccines in the United Kingdom. J Med Econ 2024; 27:1359-1372. [PMID: 39479770 DOI: 10.1080/13696998.2024.2413288] [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: 08/28/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 11/09/2024]
Abstract
AIMS To estimate the potential clinical impact and cost-effectiveness of a United Kingdom (UK) Autumn 2024 vaccination campaign with an updated Moderna COVID-19 vaccine in adults ≥65 years and eligible persons 6 months to 64 years of age over a 1-year time horizon (September 2024-August 2025). MATERIALS AND METHODS A compartmental Susceptible-Exposed-Infected-Recovered model was adapted to reflect COVID-19 cases in the UK. Numbers of symptomatic infections, COVID-19-related hospitalizations and deaths, costs, and quality-adjusted life-years (QALYs) were predicted using a decision tree. The incremental cost-effectiveness ratio (ICER) of an updated Moderna mRNA vaccine (Moderna Autumn 2024 Campaign) was compared to No Autumn 2024 vaccine and to an updated Pfizer-BioNTech mRNA Autumn 2024 vaccine, from a healthcare perspective. RESULTS The Moderna Autumn 2024 Vaccination Campaign is predicted to decrease the expected 8.3 million symptomatic infections with no vaccination by 19% to 6.7 million. Hospitalizations, long COVID cases, and deaths are expected to decline by 27,000 (-38%), 59,000 (-19%), and 6,000 (-43%), respectively. The Moderna Autumn 2024 Campaign will increase QALYs by 78,000 and costs by £665 million, yielding an ICER of £8,500/QALY gained. Sensitivity analyses suggest that vaccine effectiveness (VE) and waning, symptomatic infection incidence, hospitalization rates, and mortality rates drive cost-effectiveness. Vaccination remains cost-effective when lowering the target population to ≥50 years. Use of the Moderna vaccine is expected to prevent 8,000 more hospitalizations and 1,700 more deaths than the updated Pfizer-BioNTech vaccine. CONCLUSIONS Vaccination of the eligible population would contribute to significant reductions in hospitalizations, deaths, and long COVID in the UK in the 2024-2025 season. Expanding the target population continues to be cost-effective. Use of the Moderna Autumn 2024 Campaign is predicted to reduce SARS-CoV-2 infections and associated outcomes in a cost-effective manner and will contribute to a more resilient healthcare system in the UK.
Collapse
Affiliation(s)
| | | | - Amy Lee
- Quadrant Health Economics Inc, Cambridge, Canada
| | | | | | | | | | | |
Collapse
|
31
|
Bruxvoort KJ, Sy LS, Hong V, Lewin B, Qian L, Huang X, Holmquist KJ, Han B, Xu S. Factors associated with uptake of bivalent mRNA COVID-19 vaccines in a large US health care system. Vaccine 2023; 41:7460-7468. [PMID: 37953096 PMCID: PMC10954380 DOI: 10.1016/j.vaccine.2023.11.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/27/2023] [Accepted: 11/04/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Bivalent mRNA COVID-19 vaccines were developed to provide protection against the original SARS-CoV-2 strain and Omicron BA.4/BA.5 variants, but uptake in the United States has been low. Sociodemographic disparities in COVID-19 vaccine uptake have been documented, but it is unclear if similar disparities persist among individuals who previously completed a primary series of monovalent COVID-19 vaccine. METHODS We conducted a retrospective cohort study at Kaiser Permanente Southern California (KPSC) including youth aged 5-17 years and adults aged ≥18 years who were KPSC members and had completed a primary series of monovalent COVID-19 vaccine. Individuals were followed from index date (date of eligibility for bivalent vaccine) to 03/31/2023 to ascertain receipt of any dose of bivalent mRNA COVID-19 vaccine or until disenrollment from KPSC or death. Multivariable robust Poisson regression was conducted to assess the adjusted relative risk and 95 % confidence intervals of factors associated with receipt of bivalent vaccine. RESULTS The final cohorts included 305,339 youth and 2,534,619 adults, of whom 19.5 % and 30.7 %, respectively, had received bivalent COVID-19 vaccine. Factors associated with being more likely to receive bivalent COVID-19 vaccine included older age, Asian race, more prior year outpatient and virtual visits, Charlson score ≥1, and immunocompromised status. Factors associated with being less likely to receive a bivalent COVID-19 vaccine included age 12-17 vs 5-11 years, Hispanic and non-Hispanic Black race/ethnicity, ≥1 prior year inpatient or emergency department visits, prior history of SARS-CoV-2 infection (adults only), Medicaid insurance, and higher neighborhood deprivation index. CONCLUSION Even among youth and adults who had previously received a primary series of monovalent COVID-19 vaccine, sociodemographic and clinical disparities were observed in receipt of bivalent COVID-19 vaccine. These findings are critical to inform equitable strategies for the implementation of the updated monovalent COVID-19 vaccine targeting the Omicron XBB strain.
Collapse
Affiliation(s)
- Katia J Bruxvoort
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL 35233, United States; Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S. Los Robles Ave, 5th Floor, Pasadena, CA 91101, United States.
| | - Lina S Sy
- Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S. Los Robles Ave, 5th Floor, Pasadena, CA 91101, United States
| | - Vennis Hong
- Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S. Los Robles Ave, 5th Floor, Pasadena, CA 91101, United States
| | - Bruno Lewin
- Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S. Los Robles Ave, 5th Floor, Pasadena, CA 91101, United States
| | - Lei Qian
- Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S. Los Robles Ave, 5th Floor, Pasadena, CA 91101, United States
| | - Xuan Huang
- Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S. Los Robles Ave, 5th Floor, Pasadena, CA 91101, United States
| | - Kimberly J Holmquist
- Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S. Los Robles Ave, 5th Floor, Pasadena, CA 91101, United States
| | - Bing Han
- Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S. Los Robles Ave, 5th Floor, Pasadena, CA 91101, United States
| | - Stanley Xu
- Department of Research & Evaluation, Kaiser Permanente Southern California, 100 S. Los Robles Ave, 5th Floor, Pasadena, CA 91101, United States; Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA 91101, United States
| |
Collapse
|