1
|
Jung M, Kim H, Choi E, Shin MK, Shin SJ. Enhancing vaccine effectiveness in the elderly to counter antibiotic resistance: The potential of adjuvants via pattern recognition receptors. Hum Vaccin Immunother 2024; 20:2317439. [PMID: 39693178 DOI: 10.1080/21645515.2024.2317439] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/24/2024] [Accepted: 02/07/2024] [Indexed: 12/20/2024] Open
Abstract
Vaccines are an effective way to prevent the emergence and spread of antibiotic resistance by preventing diseases and establishing herd immunity. However, the reduced effectiveness of vaccines in the elderly due to immunosenescence is one of the significant contributors to the increasing antibiotic resistance. To counteract this decline and enhance vaccine effectiveness in the elderly, adjuvants play a pivotal role. Adjuvants are designed to augment the effectiveness of vaccines by activating the innate immune system, particularly through pattern recognition receptors on antigen-presenting cells. To improve vaccine effectiveness in the elderly using adjuvants, it is imperative to select the appropriate adjuvants based on an understanding of immunosenescence and the mechanisms of adjuvant functions. This review demonstrates the phenomenon of immunosenescence and explores various types of adjuvants, including their mechanisms and their potential in improving vaccine effectiveness for the elderly, thereby contributing to developing more effective vaccines for this vulnerable demographic.
Collapse
Affiliation(s)
- Myunghwan Jung
- Department of Microbiology, Institute of Medical Science, Department of Convergence Medical Science, BK21 Center for Human Resource Development in the Bio-Health Industry, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - Hongmin Kim
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Eunsol Choi
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| | - Min-Kyoung Shin
- Department of Microbiology, Institute of Medical Science, Department of Convergence Medical Science, BK21 Center for Human Resource Development in the Bio-Health Industry, Gyeongsang National University College of Medicine, Jinju, South Korea
| | - Sung Jae Shin
- Department of Microbiology, Institute for Immunology and Immunological Diseases, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
2
|
Cnossen VM, van Leeuwen RP, Mazur NI, Vernhes C, ten Voorde W, Burggraaf J, de Visser SJ, Roestenberg M, Kamerling IMC. From setbacks to success: lessons from the journey of RSV vaccine development. Ther Adv Vaccines Immunother 2024; 12:25151355241308305. [PMID: 39711948 PMCID: PMC11660060 DOI: 10.1177/25151355241308305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 12/03/2024] [Indexed: 12/24/2024] Open
Abstract
Respiratory syncytial virus (RSV) causes high worldwide infant mortality, as well as a high disease burden in the elderly. Efforts in vaccine development over the past 60 years have recently delivered three approved vaccines and two monoclonal antibodies (mAbs). Looking back at the eventful history of RSV vaccine development, several factors can be identified that have hampered the developmental pathway, including the occurrence of enhanced RSV disease (ERD) in the first vaccine attempt and the difficulty in characterizing and stabilizing the pre-fusion F protein as a vaccine target. Moreover, the need for large trials to test vaccine efficacy, usually done late in development, and the lack of a correlate of protection (CoP) result in significant uncertainties in RSV vaccine development. The use of controlled human infection models (CHIMs) may provide a solution for some of these problems: through swift, cost-efficient and closely monitored assessment of vaccine safety and efficacy in early clinical phases, vaccines can either 'fail fast' or show results supporting further investments. Moreover, CHIMs facilitate the assessment of disease and could assist in the identification of a CoP supporting late-stage development. Although some factors may affect translatability to real-world vaccine efficacy, CHIMs can support the clinical development pathway in various ways. We advocate for, and demonstrate, a conceptual and rational design of RSV vaccine development. Assessing protective efficacy early on would result in the most cost-efficient pathway and identification of target populations should be done as early as possible. For RSV, elderly individuals and people in low- and middle-income countries are high-impact populations for RSV prevention. While RSV immunization is now available in certain regions, global access is not accomplished yet, and worldwide prevention does not seem within reach. Quick and cost-effective assessments of candidates currently in the pipeline could contribute to future successes in the battle against RSV.
Collapse
Affiliation(s)
- Victor M. Cnossen
- Centre for Human Drug Research, Zernikedreef 8, 2333 CL Leiden, The Netherlands
| | | | | | - Charlotte Vernhes
- Vaccines Europe, European Federation of Pharmaceutical Industries and Associations, Brussels, Belgium
| | | | | | - Saco J. de Visser
- Centre for Future Affordable & Sustainable Therapy Development (FAST), The Hague, The Netherlands
| | - Meta Roestenberg
- Centre for Human Drug Research, Leiden, The Netherlands
- Leiden University Centre for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, The Netherlands
| | - Ingrid M. C. Kamerling
- Centre for Human Drug Research, Leiden, The Netherlands
- Leiden University Centre for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
3
|
Jordan E, Jenkins V, Silbernagl G, Chávez MPV, Schmidt D, Schnorfeil F, Schultz S, Chen L, Salgado F, Jacquet JM, Welte T, De Moerlooze L. A multivalent RSV vaccine based on the modified vaccinia Ankara vector shows moderate protection against disease caused by RSV in older adults in a phase 3 clinical study. Vaccine 2024; 42:126427. [PMID: 39461302 DOI: 10.1016/j.vaccine.2024.126427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/01/2024] [Accepted: 10/06/2024] [Indexed: 10/29/2024]
Abstract
Respiratory syncytial virus (RSV) causes a significant disease burden in older adults. The live recombinant vaccine based on a nonreplicating modified vaccinia Ankara (MVA-BN) poxvirus, MVA-BN-RSV, encoding for multiple proteins of RSV subtypes A and B, was assessed for efficacy against respiratory disease caused by RSV. Adults aged ≥60 years, with or without underlying chronic conditions, were enrolled and randomized in a 1:1 ratio to receive a single dose of vaccine or placebo and were followed for disease caused by RSV infection during the 2022-2023 season. The 2 primary endpoints were RSV-associated lower respiratory tract disease (LRTD) with ≥3 and ≥ 2 symptoms; acute respiratory disease (ARD) was a key secondary endpoint. The humoral RSV-specific immune response was assessed at baseline and 14 days post-vaccination. Safety was evaluated by collection of solicited adverse events (AEs) and unsolicited AEs for 7 and 28 days post-vaccination respectively, and SAEs for the entire study period. In total, 18,348 participants were included in the final efficacy and safety analyses. Vaccine efficacy was 42.9 % (95 % CI: -16.1; 71.9) against RSV-associated LRTD with ≥3 symptoms, 59.0 % (95 % CI: 34.7; 74.3) against LRTD with ≥2 symptoms, and 48.8 % (95 % CI: 25.8; 64.7) against ARD. The primary objective was not met for LRTD with ≥3 symptoms since the lower bound of the 95 % CI was below 20 %, the prespecified success criterion. The vaccine-elicited immune response showed mean fold-increases of 1.7 for RSV A and B neutralizing antibodies and 2.9 and 4.3 for RSV-specific IgG and IgA, respectively. The vaccine displayed mild to moderate reactogenicity, and no safety concerns were identified. MVA-BN-RSV induced suboptimal protection against RSV-associated LRTD, likely due to suboptimal neutralizing antibody response. The vaccine had an acceptable safety profile and confirmed immunogenicity, overall showing promise for MVA-BN-vectored constructs targeting other diseases. Trial Registration:Clinicaltrials.gov Identifier NCT05238025 (Registered February 14, 2022).
Collapse
MESH Headings
- Humans
- Respiratory Syncytial Virus Vaccines/immunology
- Respiratory Syncytial Virus Vaccines/administration & dosage
- Respiratory Syncytial Virus Vaccines/adverse effects
- Respiratory Syncytial Virus Infections/prevention & control
- Respiratory Syncytial Virus Infections/immunology
- Male
- Female
- Aged
- Vaccinia virus/immunology
- Vaccinia virus/genetics
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Respiratory Syncytial Virus, Human/immunology
- Respiratory Syncytial Virus, Human/genetics
- Middle Aged
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/adverse effects
- Double-Blind Method
- Vaccine Efficacy
- Aged, 80 and over
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Vaccines, Attenuated/immunology
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/adverse effects
- Genetic Vectors/immunology
Collapse
Affiliation(s)
- Elke Jordan
- Bavarian Nordic, Bavarian Nordic GmbH, Martinsried, Germany.
| | - Victoria Jenkins
- Bavarian Nordic, Bavarian Nordic Switzerland AG, Zug, Switzerland
| | | | | | - Darja Schmidt
- Bavarian Nordic, Bavarian Nordic GmbH, Martinsried, Germany
| | | | | | - Liddy Chen
- Bavarian Nordic Inc., Durham, North Carolina, USA
| | - Fernanda Salgado
- Bavarian Nordic, Bavarian Nordic Switzerland AG, Zug, Switzerland
| | | | - Tobias Welte
- Medizinische Hochschule Hannover, Klinik für Pneumologie und Infektiologie Carl-Neuberg-Straße 1, 30625 Hannover, Germany
| | | |
Collapse
|
4
|
Gupta A, Rudra A, Reed K, Langer R, Anderson DG. Advanced technologies for the development of infectious disease vaccines. Nat Rev Drug Discov 2024; 23:914-938. [PMID: 39433939 DOI: 10.1038/s41573-024-01041-z] [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: 08/28/2024] [Indexed: 10/23/2024]
Abstract
Vaccines play a critical role in the prevention of life-threatening infectious disease. However, the development of effective vaccines against many immune-evading pathogens such as HIV has proven challenging, and existing vaccines against some diseases such as tuberculosis and malaria have limited efficacy. The historically slow rate of vaccine development and limited pan-variant immune responses also limit existing vaccine utility against rapidly emerging and mutating pathogens such as influenza and SARS-CoV-2. Additionally, reactogenic effects can contribute to vaccine hesitancy, further undermining the ability of vaccination campaigns to generate herd immunity. These limitations are fuelling the development of novel vaccine technologies to more effectively combat infectious diseases. Towards this end, advances in vaccine delivery systems, adjuvants, antigens and other technologies are paving the way for the next generation of vaccines. This Review focuses on recent advances in synthetic vaccine systems and their associated challenges, highlighting innovation in the field of nano- and nucleic acid-based vaccines.
Collapse
Affiliation(s)
- Akash Gupta
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Arnab Rudra
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Kaelan Reed
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
5
|
Lee YZ, Han J, Zhang YN, Ward G, Braz Gomes K, Auclair S, Stanfield RL, He L, Wilson IA, Zhu J. Rational design of uncleaved prefusion-closed trimer vaccines for human respiratory syncytial virus and metapneumovirus. Nat Commun 2024; 15:9939. [PMID: 39550381 PMCID: PMC11569192 DOI: 10.1038/s41467-024-54287-x] [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: 03/12/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) cause human respiratory diseases and are major targets for vaccine development. In this study, we design uncleaved prefusion-closed (UFC) trimers for the fusion protein (F) of both viruses by examining mutations critical to F metastability. For RSV, we assess four previous prefusion F designs, including the first and second generations of DS-Cav1, SC-TM, and 847A. We then identify key mutations that can maintain prefusion F in a native-like, closed trimeric form (up to 76%) without introducing any interprotomer disulfide bond. For hMPV, we develop a stable UFC trimer with a truncated F2-F1 linkage and an interprotomer disulfide bond. Dozens of UFC constructs are characterized by negative-stain electron microscopy (nsEM), x-ray crystallography (11 RSV-F structures and one hMPV-F structure), and antigenic profiling. Using an optimized RSV-F UFC trimer as bait, we identify three potent RSV neutralizing antibodies (NAbs) from a phage-displayed human antibody library, with a public NAb lineage targeting sites Ø and V and two cross-pneumovirus NAbs recognizing site III. In mouse immunization, rationally designed RSV-F and hMPV-F UFC trimers induce robust antibody responses with high neutralizing titers. Our study provides a foundation for future prefusion F-based RSV and hMPV vaccine development.
Collapse
Affiliation(s)
- Yi-Zong Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jerome Han
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Yi-Nan Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Garrett Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | | | - Sarah Auclair
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Robyn L Stanfield
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
- Uvax Bio, LLC, Newark, DE, 19702, USA.
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
6
|
da Silva GS, Borges SG, Pozzebon BB, de Souza APD. Immune Responses to Respiratory Syncytial Virus Vaccines: Advances and Challenges. Microorganisms 2024; 12:2305. [PMID: 39597694 PMCID: PMC11596275 DOI: 10.3390/microorganisms12112305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Respiratory Syncytial Virus (RSV) is a leading cause of acute respiratory infections, particularly in children and the elderly. This virus primarily infects ciliated epithelial cells and activates alveolar macrophages and dendritic cells, triggering an innate antiviral response that releases pro-inflammatory cytokines. However, immunity generated by infection is limited, often leading to reinfection throughout life. This review focuses on the immune response elicited by newly developed and approved vaccines against RSV. A comprehensive search of clinical studies on RSV vaccine candidates conducted between 2013 and 2024 was performed. There are three primary target groups for RSV vaccines: pediatric populations, infants through maternal immunization, and the elderly. Different vaccine approaches address these groups, including subunit, live attenuated or chimeric, vector-based, and mRNA vaccines. To date, subunit RSV vaccines and the mRNA vaccine have been approved using the pre-fusion conformation of the F protein, which has been shown to induce strong immune responses. Nevertheless, several other vaccine candidates face challenges, such as modest increases in antibody production, highlighting the need for further research. Despite the success of the approved vaccines for adults older than 60 years and pregnant women, there remains a critical need for vaccines that can protect children older than six months, who are still highly vulnerable to RSV infections.
Collapse
Affiliation(s)
| | | | | | - Ana Paula Duarte de Souza
- Laboratory of Clinical and Experimental Immunology, Infant Center, School of Health Science, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre 90619-900, Brazil
| |
Collapse
|
7
|
Moreira AC, Ribeiro AB, Oliveira I, Sá M, Lameirão C, Marques P. Efficacy of anti-RSV vaccination in preventing respiratory syncytial virus disease and severe illness in older adults: a systematic review of randomized controlled trials. Eur Geriatr Med 2024; 15:1215-1229. [PMID: 39325332 DOI: 10.1007/s41999-024-01066-y] [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: 08/05/2024] [Accepted: 09/11/2024] [Indexed: 09/27/2024]
Abstract
PURPOSE Several vaccines against respiratory syncytial virus (RSV) are being developed. The objective of this systematic review is to determine the efficacy of these vaccines in preventing RSV disease in adults over 60 years of age, especially regarding severe illness. METHODS We researched MEDLINE, CENTRAL, Scopus, medRxiv, bioRxiv and ClinicalTrials.gov until April/2024. Randomized controlled trials studying an anti-RSV vaccine in adults above 60 years and reporting RSV-related acute respiratory infection/disease (ARI/ARD), lower respiratory tract infection/disease (LRTI/LRTD) and severe LRTI/LRTD were included. Vaccine efficacy was calculated as ([1 - risk ratio] × 100). Risk of bias and certainty of evidence were assessed by RoB2 tool and GRADE approach. The meta-analytical relative risk of ARI/ARD and LRTI/LRTD was estimated, with a subgroup analysis performed according to the vaccine type. The random effects model was used. RESULTS We included seven studies, five addressing prefusion vaccines and two regarding non-prefusion ones. The number of RSV-related LRTI/LRTD cases was reduced by vaccination, with a relative risk of 0.39 [CI 95% 0.22 to 0.69] and a vaccine efficacy of 61%. Considering the subgroup of prefusion vaccines alone, there is a relative risk of 0.27 [CI 95% 0.19 to 0.36] for LRTI/LRTD and a vaccine efficacy of 73%. ARI/ARD incidence presented a similar pattern. Only one trial reports results for severe LRTI/LRTD, with a vaccine efficacy of 94.1% [95% CI 62.4 to 99.9]. CONCLUSIONS Prefusion anti-RSV vaccines are efficacious in the prevention of LTRI/LTRD and ARI/ARD cases. More studies regarding severe LTRI/LTRD are necessary in the future.
Collapse
Affiliation(s)
- Ana Catarina Moreira
- USF Nova Salus, Unidade Local de Saúde Gaia-Espinho, Vila Nova de Gaia, Porto, Portugal.
| | - Ana Beatriz Ribeiro
- USF Santo André de Canidelo, Unidade Local de Saúde Gaia-Espinho, Vila Nova de Gaia, Porto, Portugal
| | - Inês Oliveira
- USF Nova Salus, Unidade Local de Saúde Gaia-Espinho, Vila Nova de Gaia, Porto, Portugal
| | - Mariana Sá
- USF Nova Salus, Unidade Local de Saúde Gaia-Espinho, Vila Nova de Gaia, Porto, Portugal
| | - Catarina Lameirão
- USF Nova Salus, Unidade Local de Saúde Gaia-Espinho, Vila Nova de Gaia, Porto, Portugal
| | - Patrícia Marques
- USF Nova Salus, Unidade Local de Saúde Gaia-Espinho, Vila Nova de Gaia, Porto, Portugal
| |
Collapse
|
8
|
Peng R, Chen C, Chen Q, Zhang Y, Huang R, Zhang Y, Li J. Global progress in clinical research on human respiratory syncytial virus vaccines. Front Microbiol 2024; 15:1457703. [PMID: 39286350 PMCID: PMC11402711 DOI: 10.3389/fmicb.2024.1457703] [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: 07/01/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Human respiratory syncytial virus (hRSV) not only affects newborns but also older adults, contributing to a substantial worldwide burden of disease. However, only three approved hRSV vaccines remain commercially available to date. The development of a safe, practical and broad-spectrum vaccine suitable for all age groups remains extremely challenging. Using five different approaches-live-attenuated, recombinant-vector, subunit, particle-based, and mRNA-nearly 30 hRSV vaccine candidates are currently conducting clinical trials worldwide; moreover, > 30 vaccines are under preclinical evaluation. This review presents a comprehensive overview of these hRSV vaccines along with prospects for the development of infectious disease vaccines in the post-COVID-19 pandemic era.
Collapse
Affiliation(s)
- Ruofan Peng
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenghao Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qian Chen
- School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Yuwen Zhang
- School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Renjin Huang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Yanjun Zhang
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Jianhua Li
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| |
Collapse
|
9
|
Zeng B, Liu X, Yang Q, Wang J, Ren Q, Sun F. Efficacy and safety of vaccines to prevent respiratory syncytial virus infection in infants and older adults: A systematic review and meta-analysis. Int J Infect Dis 2024; 146:107118. [PMID: 38878994 DOI: 10.1016/j.ijid.2024.107118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/22/2024] [Accepted: 05/23/2024] [Indexed: 07/08/2024] Open
Abstract
OBJECTIVES To determine the efficacy and safety of respiratory syncytial virus (RSV) vaccines in infants and older adults. METHODS We performed a systematic review and meta-analysis of randomized control trials that evaluated the efficacy of maternal RSV immunization against infections in infants, as well as the efficacy of RSV vaccines in older adults. The primary outcome was the vaccine efficacy against RSV-related lower respiratory tract disease (LRTD). Grading of Recommendations Assessment, Development and Evaluation criteria was used to evaluate the level of evidence. RESULTS Ten trials were included in the review. For maternal vaccination, the RSV vaccine showed favourable efficacy against RSV-related LRTD (vaccine efficacy 57.3%, 95% confidence interval [CI] 31.3-73.5; low certainty) and RSV-related severe LRTD (vaccine efficacy 81.9%, 95% CI 56.8-92.4; moderate certainty) in infants within 90 days after birth. For older adults, Meta-analysis showed that RSV vaccines could also reduce the risk of RSV-related LRTD (vaccine efficacy 78.3%, 95% CI 65.6-86.3; moderate certainty) and RSV-related severe LRTD (vaccine efficacy 86.5%, 95% CI 68.3-94.3; moderate certainty). There was no significant difference in serious adverse events between RSV vaccines and placebo. CONCLUSION RSV vaccines have the potential to offer protection against RSV disease in both infants and older adults, without apparent safety concerns.
Collapse
Affiliation(s)
- Baoqi Zeng
- Central Laboratory, Tianjin Fifth Central Hospital (Peking University Binhai Hospital), Tianjin, China; Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China; Tianjin Key Laboratory of Epigenetic for Organ Development of Preterm Infants, Tianjin Fifth Central Hospital, Tianjin, China; Emergency Department of Tianjin Fifth Central Hospital, Tianjin, China.
| | - Xiaozhi Liu
- Central Laboratory, Tianjin Fifth Central Hospital (Peking University Binhai Hospital), Tianjin, China; Tianjin Key Laboratory of Epigenetic for Organ Development of Preterm Infants, Tianjin Fifth Central Hospital, Tianjin, China; High Altitude Characteristic Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People's Hospital, Qinghai, China
| | - Qingqing Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Junjian Wang
- Emergency Department of Tianjin Fifth Central Hospital, Tianjin, China
| | - Qingcuo Ren
- High Altitude Characteristic Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People's Hospital, Qinghai, China
| | - Feng Sun
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| |
Collapse
|
10
|
Gonzalez KJ, Yim KC, Blanco JCG, Boukhvalova MS, Strauch EM. Systematic computer-aided disulfide design as a general strategy to stabilize prefusion class I fusion proteins. Front Immunol 2024; 15:1406929. [PMID: 39114655 PMCID: PMC11303214 DOI: 10.3389/fimmu.2024.1406929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/03/2024] [Indexed: 08/10/2024] Open
Abstract
Numerous enveloped viruses, such as coronaviruses, influenza, and respiratory syncytial virus (RSV), utilize class I fusion proteins for cell entry. During this process, the proteins transition from a prefusion to a postfusion state, undergoing substantial and irreversible conformational changes. The prefusion conformation has repeatedly shown significant potential in vaccine development. However, the instability of this state poses challenges for its practical application in vaccines. While non-native disulfides have been effective in maintaining the prefusion structure, identifying stabilizing disulfide bonds remains an intricate task. Here, we present a general computational approach to systematically identify prefusion-stabilizing disulfides. Our method assesses the geometric constraints of disulfide bonds and introduces a ranking system to estimate their potential in stabilizing the prefusion conformation. We hypothesized that disulfides restricting the initial stages of the conformational switch could offer higher stability to the prefusion state than those preventing unfolding at a later stage. The implementation of our algorithm on the RSV F protein led to the discovery of prefusion-stabilizing disulfides that supported our hypothesis. Furthermore, the evaluation of our top design as a vaccine candidate in a cotton rat model demonstrated robust protection against RSV infection, highlighting the potential of our approach for vaccine development.
Collapse
Affiliation(s)
- Karen J. Gonzalez
- Institute of Bioinformatics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States
- Department of Medicine, School of Medicine, Washington University, St. Louis, MO, United States
| | - Kevin C. Yim
- Sigmovir Biosystems, Inc., Rockville, MD, United States
| | | | | | - Eva-Maria Strauch
- Institute of Bioinformatics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, United States
- Department of Medicine, School of Medicine, Washington University, St. Louis, MO, United States
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, United States
| |
Collapse
|
11
|
Yamaue R, Torikai M, Terashima M, Mori H. KD-409, a Respiratory Syncytial Virus FG Chimeric Protein without the CX3C Chemokine Motif, Is an Efficient Respiratory Syncytial Virus Vaccine Preparation for Passive and Active Immunization in Mice. Vaccines (Basel) 2024; 12:753. [PMID: 39066391 PMCID: PMC11281633 DOI: 10.3390/vaccines12070753] [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: 06/05/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Although respiratory syncytial virus (RSV) vaccine development initiatives have existed for half a century, no candidate has been approved for application at all ages from neonates to children. Developing an effective and safe RSV vaccine for pediatric use is challenging owing to RSV-associated disease and vaccine-enhanced disease (VED). We aimed to design an RSV vaccine, KD-409, by structurally incorporating the F ectodomain and G protein central conserved domain without the CX3C chemokine motif and test its efficacy and safety. KD-409 formed rosette particles or trimmers. KD-409 immunization of mice mainly induced anti-RSV F protein IgG. The induced anti-F antibodies had a higher IgG2a/IgG1 ratio than pre-fusion F, suggesting that they induced Th1-dominant immunity. Active and passive immunities were assessed by analyzing the viral titers in BALB/c mice intranasally challenged with RSV after intramuscular KD-409 immunization and pups derived from mothers who were intramuscularly vaccinated with KD-409 twice, respectively. KD-409 was more effective than post-fusion F and had a lower minimum effective dose than pre-fusion F. Thus, KD-409 demonstrated great potential as a novel RSV vaccine candidate, outperforming existing RSV F-based candidates. Our findings provide a promising strategy to overcome RSV-associated acute lower respiratory infections without the risk of VED associated with traditional approaches.
Collapse
Affiliation(s)
| | - Masaharu Torikai
- KM Biologics Co., Ltd., Kikuchi Research Center, 1314-1 Kyokushi Kawabe, Kikuchi-shi 869-1298, Japan; (R.Y.); (M.T.)
| | | | | |
Collapse
|
12
|
Pang Y, Lu H, Cao D, Zhu X, Long Q, Tian F, Long X, Li Y. Efficacy, immunogenicity and safety of respiratory syncytial virus prefusion F vaccine: systematic review and meta-analysis. BMC Public Health 2024; 24:1244. [PMID: 38711074 PMCID: PMC11075318 DOI: 10.1186/s12889-024-18748-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 04/30/2024] [Indexed: 05/08/2024] Open
Abstract
OBJECTIVE A notable research gap exists in the systematic review and meta-analysis concerning the efficacy, immunogenicity, and safety of the respiratory syncytial virus (RSV) prefusion F vaccine. METHODS We conducted a comprehensive search across PubMed, Embase, the Cochrane Central Register of Controlled Trials, and ClinicalTrials.gov to retrieve articles related to the efficacy, immunogenicity, and safety of RSV prefusion F vaccines, published through September 8, 2023. We adhered to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. RESULTS A total of 22 randomized controlled trials involving 78,990 participants were included in this systematic review and meta-analysis. The RSV prefusion F vaccine exhibited a vaccine effectiveness of 68% (95% CI: 59-75%) against RSV-associated acute respiratory illness, 70% (95% CI: 60-77%) against medically attended RSV-associated lower respiratory tract illness, and 87% (95% CI: 71-94%) against medically attended severe RSV-associated lower respiratory tract illness. Common reported local adverse reactions following RSV prefusion F vaccination include pain, redness, and swelling at the injection site, and systemic reactions such as fatigue, headache, myalgia, arthralgia, nausea, and chills. CONCLUSIONS Our meta-analysis suggests that vaccines using the RSV prefusion F protein as antigen exhibit appears broadly acceptable efficacy, immunogenicity, and safety in the population. In particular, it provides high protective efficiency against severe RSV-associated lower respiratory tract disease.
Collapse
Affiliation(s)
- Yi Pang
- Youjiang Medical University for Nationalities, Baise, China
| | - Haishan Lu
- Clinicopathological Diagnosis & Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise, China
| | - Demin Cao
- Clinicopathological Diagnosis & Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise, China
| | - Xiaoying Zhu
- Clinicopathological Diagnosis & Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise, China
| | - Qinqin Long
- Clinicopathological Diagnosis & Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise, China
| | - Fengqin Tian
- Clinicopathological Diagnosis & Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China
- Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise, China
| | - Xidai Long
- Clinicopathological Diagnosis & Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China.
- Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise, China.
| | - Yulei Li
- Clinicopathological Diagnosis & Research Center, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, China.
- Key Laboratory of Tumor Molecular Pathology of Guangxi Higher Education Institutes, Baise, China.
| |
Collapse
|
13
|
Riccò M, Cascio A, Corrado S, Bottazzoli M, Marchesi F, Gili R, Giuri PG, Gori D, Manzoni P. Efficacy of Respiratory Syncytial Virus Vaccination to Prevent Lower Respiratory Tract Illness in Older Adults: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Vaccines (Basel) 2024; 12:500. [PMID: 38793751 PMCID: PMC11126042 DOI: 10.3390/vaccines12050500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/26/2024] Open
Abstract
A systematic review and meta-analysis was designed in order to ascertain the effectiveness of respiratory syncytial virus (RSV) vaccination in preventing lower respiratory tract diseases (LRTD) in older adults (age ≥ 60 years). Studies reporting on randomized controlled trials (RCTs) were searched for in three databases (PubMed, Embase, and Scopus) and the preprint repository medRxiv until 31 March 2024. A total of nine studies were eventually included, two of which were conference proceedings. Our analysis included five RCTs on five RSV vaccines (RSVpreF, RSVPreF3, Ad26.RSV.preF, MEDI7510, and mRNA-1345). The meta-analysis documented a pooled vaccine efficacy of 81.38% (95% confidence interval (95% CI) 70.94 to 88.06) for prevention of LRTD with three or more signs/symptoms during the first RSV season after the delivery of the vaccine. Follow-up data were available for RSVPreF3 (2 RSV seasons), RSVpreF (mid-term estimates of second RSV season), and mRNA-1345 (12 months after the delivery of the primer), with a pooled VE of 61.15% (95% CI 45.29 to 72.40). After the first season, the overall risk for developing RSV-related LRTD was therefore substantially increased (risk ratio (RR) 4.326, 95% CI 2.415; 7.748). However, all estimates were affected by substantial heterogeneity, as suggested by the 95% CI of I2 statistics, which could be explained by inconsistencies in the design of the parent studies, particularly when dealing with case definition. In conclusion, adult RSV vaccination was quite effective in preventing LRTD in older adults, but the overall efficacy rapidly decreased in the second season after the delivery of the vaccine. Because of the heterogenous design of the parent studies, further analyses are required before tailoring specific public health interventions.
Collapse
Affiliation(s)
- Matteo Riccò
- AUSL–IRCCS di Reggio Emilia, Servizio di Prevenzione e Sicurezza Negli Ambienti di Lavoro (SPSAL), Local Health Unit of Reggio Emilia, 42122 Reggio Emilia, Italy
| | - Antonio Cascio
- Infectious and Tropical Diseases Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, “G D’Alessandro”, University of Palermo, AOUP P. Giaccone, 90127 Palermo, Italy;
| | - Silvia Corrado
- ASST Rhodense, Dipartimento della donna e Area Materno-Infantile, UOC Pediatria, 20024 Milano, Italy
| | - Marco Bottazzoli
- Department of Otorhinolaryngology, APSS Trento, 38122 Trento, Italy
| | - Federico Marchesi
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Renata Gili
- Department of Prevention, Turin Local Health Authority, 10125 Torino, Italy
| | | | - Davide Gori
- Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy;
| | - Paolo Manzoni
- Department of Public Health and Pediatric Sciences, University of Torino School of Medicine, 10125 Turin, Italy;
| |
Collapse
|
14
|
Lee YZ, Han J, Zhang YN, Ward G, Gomes KB, Auclair S, Stanfield RL, He L, Wilson IA, Zhu J. A tale of two fusion proteins: understanding the metastability of human respiratory syncytial virus and metapneumovirus and implications for rational design of uncleaved prefusion-closed trimers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.07.583986. [PMID: 38496645 PMCID: PMC10942449 DOI: 10.1101/2024.03.07.583986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) cause human respiratory diseases and are major targets for vaccine development. In this study, we designed uncleaved prefusion-closed (UFC) trimers for the fusion (F) proteins of both viruses by examining mutations critical to F metastability. For RSV, we assessed four previous prefusion F designs, including the first and second generations of DS-Cav1, SC-TM, and 847A. We then identified key mutations that can maintain prefusion F in a native-like, closed trimeric form (up to 76%) without introducing any interprotomer disulfide bond. For hMPV, we developed a stable UFC trimer with a truncated F2-F1 linkage and an interprotomer disulfide bond. Tens of UFC constructs were characterized by negative-stain electron microscopy (nsEM), x-ray crystallography (11 RSV-F and one hMPV-F structures), and antigenic profiling. Using an optimized RSV-F UFC trimer as bait, we identified three potent RSV neutralizing antibodies (NAbs) from a phage-displayed human antibody library, with a public NAb lineage targeting sites Ø and V and two cross-pneumovirus NAbs recognizing site III. In mouse immunization, rationally designed RSV-F and hMPV-F UFC trimers induced robust antibody responses with high neutralizing titers. Our study provides a foundation for future prefusion F-based RSV and hMPV vaccine development.
Collapse
Affiliation(s)
- Yi-Zong Lee
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Jerome Han
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Yi-Nan Zhang
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Garrett Ward
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Keegan Braz Gomes
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Sarah Auclair
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Robyn L Stanfield
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Linling He
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Ian A Wilson
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
- Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, California 92037, USA
| | - Jiang Zhu
- Department of Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, California 92037, USA
- Department of Immunology and Microbiology, The Scripps Research Institute, La Jolla, California 92037, USA
| |
Collapse
|
15
|
Gonzalez KJ, Huang J, Criado MF, Banerjee A, Tompkins SM, Mousa JJ, Strauch EM. A general computational design strategy for stabilizing viral class I fusion proteins. Nat Commun 2024; 15:1335. [PMID: 38351001 PMCID: PMC10864359 DOI: 10.1038/s41467-024-45480-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
Many pathogenic viruses rely on class I fusion proteins to fuse their viral membrane with the host cell membrane. To drive the fusion process, class I fusion proteins undergo an irreversible conformational change from a metastable prefusion state to an energetically more stable postfusion state. Mounting evidence underscores that antibodies targeting the prefusion conformation are the most potent, making it a compelling vaccine candidate. Here, we establish a computational design protocol that stabilizes the prefusion state while destabilizing the postfusion conformation. With this protocol, we stabilize the fusion proteins of the RSV, hMPV, and SARS-CoV-2 viruses, testing fewer than a handful of designs. The solved structures of these designed proteins from all three viruses evidence the atomic accuracy of our approach. Furthermore, the humoral response of the redesigned RSV F protein compares to that of the recently approved vaccine in a mouse model. While the parallel design of two conformations allows the identification of energetically sub-optimal positions for one conformation, our protocol also reveals diverse molecular strategies for stabilization. Given the clinical significance of viruses using class I fusion proteins, our algorithm can substantially contribute to vaccine development by reducing the time and resources needed to optimize these immunogens.
Collapse
Affiliation(s)
- Karen J Gonzalez
- Institute of Bioinformatics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, 30602, USA
| | - Jiachen Huang
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Miria F Criado
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, USA
| | - Avik Banerjee
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Stephen M Tompkins
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
| | - Jarrod J Mousa
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, 30602, USA
- Department of Biochemistry and Molecular Biology, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, 30602, USA
| | - Eva-Maria Strauch
- Institute of Bioinformatics, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, 30602, USA.
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA, 30602, USA.
- Department of Medicine, School of Medicine, Washington University, St. Louis, MO, 63110, USA.
| |
Collapse
|
16
|
Jordan E, Kabir G, Schultz S, Silbernagl G, Schmidt D, Jenkins VA, Weidenthaler H, Stroukova D, Martin BK, De Moerlooze L. Reduced Respiratory Syncytial Virus Load, Symptoms, and Infections: A Human Challenge Trial of MVA-BN-RSV Vaccine. J Infect Dis 2023; 228:999-1011. [PMID: 37079393 PMCID: PMC10582911 DOI: 10.1093/infdis/jiad108] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/20/2023] [Accepted: 04/19/2023] [Indexed: 04/21/2023] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) causes significant disease burden in older adults. MVA-BN-RSV is a novel poxvirus-vectored vaccine encoding internal and external RSV proteins. METHODS In a phase 2a randomized double-blind, placebo-controlled trial, healthy participants aged 18 to 50 years received MVA-BN-RSV or placebo, then were challenged 4 weeks later with RSV-A Memphis 37b. Viral load was assessed from nasal washes. RSV symptoms were collected. Antibody titers and cellular markers were assessed before and after vaccination and challenge. RESULTS After receiving MVA-BN-RSV or placebo, 31 and 32 participants, respectively, were challenged. Viral load areas under the curve from nasal washes were lower (P = .017) for MVA-BN-RSV (median = 0.00) than placebo (median = 49.05). Total symptom scores also were lower (median = 2.50 and 27.00, respectively; P = .004). Vaccine efficacy against symptomatic, laboratory-confirmed or culture-confirmed infection was 79.3% to 88.5% (P = .022 and .013). Serum immunoglobulin A and G titers increased approximately 4-fold after MVA-BN-RSV vaccination. Interferon-γ-producing cells increased 4- to 6-fold after MVA-BN-RSV in response to stimulation with the encoded RSV internal antigens. Injection site pain occurred more frequently with MVA-BN-RSV. No serious adverse events were attributed to vaccination. CONCLUSIONS MVA-BN-RSV vaccination resulted in lower viral load and symptom scores, fewer confirmed infections, and induced humoral and cellular responses. CLINICAL TRIALS REGISTRATION NCT04752644.
Collapse
|
17
|
Ruckwardt TJ. The road to approved vaccines for respiratory syncytial virus. NPJ Vaccines 2023; 8:138. [PMID: 37749081 PMCID: PMC10519952 DOI: 10.1038/s41541-023-00734-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/13/2023] [Indexed: 09/27/2023] Open
Abstract
After decades of work, several interventions to prevent severe respiratory syncytial virus (RSV) disease in high-risk infant and older adult populations have finally been approved. There were many setbacks along the road to victory. In this review, I will discuss the impact of RSV on human health and how structure-based vaccine design set the stage for numerous RSV countermeasures to advance through late phase clinical evaluation. While there are still many RSV countermeasures in preclinical and early-stage clinical trials, this review will focus on products yielding long-awaited efficacy results. Finally, I will discuss some challenges and next steps needed to declare a global victory against RSV.
Collapse
Affiliation(s)
- Tracy J Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, 20892, USA.
| |
Collapse
|
18
|
Ou L, Chen SJ, Teng IT, Yang L, Zhang B, Zhou T, Biju A, Cheng C, Kong WP, Morano NC, Stancofski ESD, Todd JP, Tsybovsky Y, Wang S, Zheng CY, Mascola JR, Shapiro L, Woodward RA, Buchholz UJ, Kwong PD. Structure-based design of a single-chain triple-disulfide-stabilized fusion-glycoprotein trimer that elicits high-titer neutralizing responses against human metapneumovirus. PLoS Pathog 2023; 19:e1011584. [PMID: 37738240 PMCID: PMC10516418 DOI: 10.1371/journal.ppat.1011584] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 07/29/2023] [Indexed: 09/24/2023] Open
Abstract
The Pneumoviridae family of viruses includes human metapneumovirus (HMPV) and respiratory syncytial virus (RSV). The closely related Paramyxoviridae family includes parainfluenza viruses (PIVs). These three viral pathogens cause acute respiratory tract infections with substantial disease burden in the young, the elderly, and the immune-compromised. While promising subunit vaccines are being developed with prefusion-stabilized forms of the fusion glycoproteins (Fs) of RSV and PIVs, for which neutralizing titers elicited by the prefusion (pre-F) conformation of F are much higher than for the postfusion (post-F) conformation, with HMPV, pre-F and post-F immunogens described thus far elicit similar neutralizing responses, and it has been unclear which conformation, pre-F or post-F, would be the most effective HMPV F-vaccine immunogen. Here, we investigate the impact of further stabilizing HMPV F in the pre-F state. We replaced the furin-cleavage site with a flexible linker, creating a single chain F that yielded increased amounts of pre-F stabilized trimers, enabling the generation and assessment of F trimers stabilized by multiple disulfide bonds. Introduced prolines could increase both expression yields and antigenic recognition by the pre-F specific antibody, MPE8. The cryo-EM structure of a triple disulfide-stabilized pre-F trimer with the variable region of antibody MPE8 at 3.25-Å resolution confirmed the formation of designed disulfides and provided structural details on the MPE8 interface. Immunogenicity assessments in naïve mice showed the triple disulfide-stabilized pre-F trimer could elicit high titer neutralization, >10-fold higher than elicited by post-F. Immunogenicity assessments in pre-exposed rhesus macaques showed the triple disulfide-stabilized pre-F could recall high neutralizing titers after a single immunization, with little discrimination in the recall response between pre-F and post-F immunogens. However, the triple disulfide-stabilized pre-F adsorbed HMPV-directed responses from commercially available pooled human immunoglobulin more fully than post-F. Collectively, these results suggest single-chain triple disulfide-stabilized pre-F trimers to be promising HMPV-vaccine antigens.
Collapse
Affiliation(s)
- Li Ou
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Steven J. Chen
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - I-Ting Teng
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lijuan Yang
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Baoshan Zhang
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Tongqing Zhou
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Andrea Biju
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Cheng Cheng
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Wing-Pui Kong
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Nicholas C. Morano
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
- Department of Biochemistry and Molecular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, United States of America
| | | | - John-Paul Todd
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yaroslav Tsybovsky
- Electron Microscopy Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Shuishu Wang
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Cheng-Yan Zheng
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John R. Mascola
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lawrence Shapiro
- Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York, United States of America
- Department of Biochemistry and Molecular Biophysics, Columbia University Vagelos College of Physicians and Surgeons, New York, New York, United States of America
| | - Ruth A. Woodward
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ursula J. Buchholz
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Maryland, United States of America
| | - Peter D. Kwong
- Vaccine Research Center, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
19
|
Umemoto S, Nakahashi-Ouchida R, Yuki Y, Kurokawa S, Machita T, Uchida Y, Mori H, Yamanoue T, Shibata T, Sawada SI, Ishige K, Hirano T, Fujihashi K, Akiyoshi K, Kurashima Y, Tokuhara D, Ernst PB, Suzuki M, Kiyono H. Cationic-nanogel nasal vaccine containing the ectodomain of RSV-small hydrophobic protein induces protective immunity in rodents. NPJ Vaccines 2023; 8:106. [PMID: 37488116 PMCID: PMC10366164 DOI: 10.1038/s41541-023-00700-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 06/22/2023] [Indexed: 07/26/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of upper and lower respiratory tract infection, especially in children and the elderly. Various vaccines containing the major transmembrane surface proteins of RSV (proteins F and G) have been tested; however, they have either afforded inadequate protection or are associated with the risk of vaccine-enhanced disease (VED). Recently, F protein-based maternal immunization and vaccines for elderly patients have shown promising results in phase III clinical trials, however, these vaccines have been administered by injection. Here, we examined the potential of using the ectodomain of small hydrophobic protein (SHe), also an RSV transmembrane surface protein, as a nasal vaccine antigen. A vaccine was formulated using our previously developed cationic cholesteryl-group-bearing pullulan nanogel as the delivery system, and SHe was linked in triplicate to pneumococcal surface protein A as a carrier protein. Nasal immunization of mice and cotton rats induced both SHe-specific serum IgG and mucosal IgA antibodies, preventing viral invasion in both the upper and lower respiratory tracts without inducing VED. Moreover, nasal immunization induced greater protective immunity against RSV in the upper respiratory tract than did systemic immunization, suggesting a critical role for mucosal RSV-specific IgA responses in viral elimination at the airway epithelium. Thus, our nasal vaccine induced effective protection against RSV infection in the airway mucosa and is therefore a promising vaccine candidate for further development.
Collapse
Affiliation(s)
- Shingo Umemoto
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Otorhinolaryngology & Head and Neck Surgery, Faculty of Medicine, Oita University, Oita, Japan
- Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), Department of Medicine, School of Medicine, San Diego, CA, USA
| | - Rika Nakahashi-Ouchida
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
| | - Yoshikazu Yuki
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- HanaVax Inc, Tokyo, Japan
| | - Shiho Kurokawa
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
| | - Tomonori Machita
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
| | - Yohei Uchida
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
| | - Hiromi Mori
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
| | - Tomoyuki Yamanoue
- Division of Mucosal Vaccines, International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
| | - Takehiko Shibata
- Department of Microbiology, Tokyo Medical University, Tokyo, Japan
- Department of Immunology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Shin-Ichi Sawada
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kazuya Ishige
- Biochemicals Division, Yamasa Corporation, Chiba, Japan
| | - Takashi Hirano
- Department of Otorhinolaryngology & Head and Neck Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Kohtaro Fujihashi
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
- Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Department of Pediatric Dentistry, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Yosuke Kurashima
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), Department of Medicine, School of Medicine, San Diego, CA, USA
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan
- Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
- Institute for Advanced Academic Research, Chiba University, Chiba, Japan
- Department of Innovative Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Daisuke Tokuhara
- Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), Department of Medicine, School of Medicine, San Diego, CA, USA
- Department of Pediatrics, Wakayama Medical University, Wakayama, Japan
| | - Peter B Ernst
- Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), Department of Medicine, School of Medicine, San Diego, CA, USA
- Division of Comparative Pathology and Medicine, Department of Pathology, University of California, San Diego, CA, USA
- Center for Veterinary Sciences and Comparative Medicine, University of California, San Diego, CA, USA
- Future Medicine Education and Research Organization, Chiba University, Chiba, Japan
| | - Masashi Suzuki
- Department of Otorhinolaryngology & Head and Neck Surgery, Faculty of Medicine, Oita University, Oita, Japan
| | - Hiroshi Kiyono
- Division of Mucosal Immunology, IMSUT Distinguished Professor Unit, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
- Chiba University-University of California San Diego Center for Mucosal Immunology, Allergy and Vaccine (CU-UCSD cMAV), Department of Medicine, School of Medicine, San Diego, CA, USA.
- Department of Human Mucosal Vaccinology, Chiba University Hospital, Chiba, Japan.
- Chiba University Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba, Japan.
- HanaVax Inc, Tokyo, Japan.
- Future Medicine Education and Research Organization, Chiba University, Chiba, Japan.
- Mucosal Immunology and Allergy Therapeutics, Institute for Global Prominent Research, Chiba University, Chiba, Japan.
| |
Collapse
|
20
|
Walsh EE, Pérez Marc G, Zareba AM, Falsey AR, Jiang Q, Patton M, Polack FP, Llapur C, Doreski PA, Ilangovan K, Rämet M, Fukushima Y, Hussen N, Bont LJ, Cardona J, DeHaan E, Castillo Villa G, Ingilizova M, Eiras D, Mikati T, Shah RN, Schneider K, Cooper D, Koury K, Lino MM, Anderson AS, Jansen KU, Swanson KA, Gurtman A, Gruber WC, Schmoele-Thoma B. Efficacy and Safety of a Bivalent RSV Prefusion F Vaccine in Older Adults. N Engl J Med 2023; 388:1465-1477. [PMID: 37018468 DOI: 10.1056/nejmoa2213836] [Citation(s) in RCA: 219] [Impact Index Per Article: 109.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection causes considerable illness in older adults. The efficacy and safety of an investigational bivalent RSV prefusion F protein-based (RSVpreF) vaccine in this population are unknown. METHODS In this ongoing, phase 3 trial, we randomly assigned, in a 1:1 ratio, adults (≥60 years of age) to receive a single intramuscular injection of RSVpreF vaccine at a dose of 120 μg (RSV subgroups A and B, 60 μg each) or placebo. The two primary end points were vaccine efficacy against seasonal RSV-associated lower respiratory tract illness with at least two or at least three signs or symptoms. The secondary end point was vaccine efficacy against RSV-associated acute respiratory illness. RESULTS At the interim analysis (data-cutoff date, July 14, 2022), 34,284 participants had received RSVpreF vaccine (17,215 participants) or placebo (17,069 participants). RSV-associated lower respiratory tract illness with at least two signs or symptoms occurred in 11 participants in the vaccine group (1.19 cases per 1000 person-years of observation) and 33 participants in the placebo group (3.58 cases per 1000 person-years of observation) (vaccine efficacy, 66.7%; 96.66% confidence interval [CI], 28.8 to 85.8); 2 cases (0.22 cases per 1000 person-years of observation) and 14 cases (1.52 cases per 1000 person-years of observation), respectively, occurred with at least three signs or symptoms (vaccine efficacy, 85.7%; 96.66% CI, 32.0 to 98.7). RSV-associated acute respiratory illness occurred in 22 participants in the vaccine group (2.38 cases per 1000 person-years of observation) and 58 participants in the placebo group (6.30 cases per 1000 person-years of observation) (vaccine efficacy, 62.1%; 95% CI, 37.1 to 77.9). The incidence of local reactions was higher with vaccine (12%) than with placebo (7%); the incidences of systemic events were similar (27% and 26%, respectively). Similar rates of adverse events through 1 month after injection were reported (vaccine, 9.0%; placebo, 8.5%), with 1.4% and 1.0%, respectively, considered by the investigators to be injection-related. Severe or life-threatening adverse events were reported in 0.5% of vaccine recipients and 0.4% of placebo recipients. Serious adverse events were reported in 2.3% of participants in each group through the data-cutoff date. CONCLUSIONS RSVpreF vaccine prevented RSV-associated lower respiratory tract illness and RSV-associated acute respiratory illness in adults (≥60 years of age), without evident safety concerns. (Funded by Pfizer; RENOIR ClinicalTrials.gov number, NCT05035212; EudraCT number, 2021-003693-31.).
Collapse
Affiliation(s)
- Edward E Walsh
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Gonzalo Pérez Marc
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Agnieszka M Zareba
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Ann R Falsey
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Qin Jiang
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Michael Patton
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Fernando P Polack
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Conrado Llapur
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Pablo A Doreski
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Kumar Ilangovan
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Mika Rämet
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Yasushi Fukushima
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Nazreen Hussen
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Louis J Bont
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Jose Cardona
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Elliot DeHaan
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Giselle Castillo Villa
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Marinela Ingilizova
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Daniel Eiras
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Tarek Mikati
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Rupal N Shah
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Katherine Schneider
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - David Cooper
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Kenneth Koury
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Maria-Maddalena Lino
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Annaliesa S Anderson
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Kathrin U Jansen
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Kena A Swanson
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Alejandra Gurtman
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - William C Gruber
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| | - Beate Schmoele-Thoma
- From the University of Rochester Medical Center, Rochester (E.E.W., A.R.F.), and Vaccine Research and Development (E.D., G.C.V., M.I., D.E., T.M., R.N.S., K.S., D.C., K.K., K.U.J., K.A.S., A.G., W.C.G.) and Worldwide Research, Development, and Medical (A.S.A.), Pfizer, Pearl River - both in New York; iTrials-Hospital Militar Central (G.P.M.), Fundación INFANT (F.P.P.), and Fundación Respirar Clinical Research Unit (P.A.D.), Buenos Aires, and Clinica Mayo de Urgencias Médicas Cruz Blanca, San Miguel de Tucumán (C.L.) - all in Argentina; Vaccine Research and Development, Pfizer, Collegeville, PA (A.M.Z., Q.J.); Vaccine Research and Development, Pfizer, Hurley, United Kingdom (M.P.); Vaccine Research and Development, Pfizer, Raleigh, NC (K.I.); Faculty of Medicine and Health Technology, Tampere University, and Finnish Vaccine Research - both in Tampere, Finland (M.R.); Fukuwa Clinic, Tokyo (Y.F.); Netcare Lakeview Hospital, Benoni, South Africa (N.H.); the Departments of Pediatrics and Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, and Respiratory Syncytial Virus Network Foundation, Zeist - both in the Netherlands (L.J.B.); Indago Research and Health Center, Hialeah, FL (J.C.); Worldwide Safety, Pfizer, Milan (M.-M.L.); and Vaccine Research and Development, Pfizer Pharma, Berlin (B.S.-T.)
| |
Collapse
|
21
|
Saeland E, van der Fits L, Bolder R, Heemskerk-van der Meer M, Drijver J, van Polanen Y, Vaneman C, Tettero L, Cox F, Serroyen J, Jorgensen MJ, Langedijk JPM, Schuitemaker H, Callendret B, Zahn RC. Combination Ad26.RSV.preF/preF protein vaccine induces superior protective immunity compared with individual vaccine components in preclinical models. NPJ Vaccines 2023; 8:45. [PMID: 36949051 PMCID: PMC10033289 DOI: 10.1038/s41541-023-00637-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/27/2023] [Indexed: 03/24/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of severe respiratory disease for which no licensed vaccine is available. We have previously shown that a prefusion (preF) conformation-stabilized RSV F protein antigen and an adenoviral vector encoding RSV preF protein (Ad26.RSV.preF) are immunogenic and protective in animals when administered as single components. Here, we evaluated a combination of the 2 components, administered as a single injection. Strong induction of both humoral and cellular responses was shown in RSV-naïve and pre-exposed mice and pre-exposed African green monkeys (AGMs). Both components of the combination vaccine contributed to humoral immune responses, while the Ad26.RSV.preF component was the main contributor to cellular immune responses in both mice and AGMs. Immunization with the combination elicited superior protection against RSV A2 challenge in cotton rats. These results demonstrate the advantage of a combination vaccine and support further clinical development.
Collapse
Affiliation(s)
- Eirikur Saeland
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands.
| | | | - Renske Bolder
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | | | - Joke Drijver
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | | | | | | | - Freek Cox
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | - Jan Serroyen
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | - Matthew J Jorgensen
- Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | | | | | | | - Roland C Zahn
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| |
Collapse
|
22
|
Gonzalez KJ, Huang J, Criado MF, Banerjee A, Tompkins S, Mousa JJ, Strauch EM. A general computational design strategy for stabilizing viral class I fusion proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.16.532924. [PMID: 36993551 PMCID: PMC10055117 DOI: 10.1101/2023.03.16.532924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Many pathogenic viruses, including influenza virus, Ebola virus, coronaviruses, and Pneumoviruses, rely on class I fusion proteins to fuse viral and cellular membranes. To drive the fusion process, class I fusion proteins undergo an irreversible conformational change from a metastable prefusion state to an energetically more favorable and stable postfusion state. An increasing amount of evidence exists highlighting that antibodies targeting the prefusion conformation are the most potent. However, many mutations have to be evaluated before identifying prefusion-stabilizing substitutions. We therefore established a computational design protocol that stabilizes the prefusion state while destabilizing the postfusion conformation. As a proof of concept, we applied this principle to the fusion protein of the RSV, hMPV, and SARS-CoV-2 viruses. For each protein, we tested less than a handful of designs to identify stable versions. Solved structures of designed proteins from the three different viruses evidenced the atomic accuracy of our approach. Furthermore, the immunological response of the RSV F design compared to a current clinical candidate in a mouse model. While the parallel design of two conformations allows identifying and selectively modifying energetically less optimized positions for one conformation, our protocol also reveals diverse molecular strategies for stabilization. We recaptured many approaches previously introduced manually for the stabilization of viral surface proteins, such as cavity-filling, optimization of polar interactions, as well as postfusion-disruptive strategies. Using our approach, it is possible to focus on the most impacting mutations and potentially preserve the immunogen as closely as possible to its native version. The latter is important as sequence re-design can cause perturbations to B and T cell epitopes. Given the clinical significance of viruses using class I fusion proteins, our algorithm can substantially contribute to vaccine development by reducing the time and resources needed to optimize these immunogens.
Collapse
Affiliation(s)
- Karen J Gonzalez
- Institute of Bioinformatics, Franklin College of Arts and Sciences, University of Georgia; Athens, GA 30602, USA
| | - Jiachen Huang
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
| | - Miria F Criado
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Department of Pathobiology, Auburn University; Auburn, AL 36849, USA
| | - Avik Banerjee
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
| | - Stephen Tompkins
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
| | - Jarrod J Mousa
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Department of Biochemistry and Molecular Biology, Franklin College of Arts and Sciences, University of Georgia; Athens, GA 30602, USA
| | - Eva-Maria Strauch
- Institute of Bioinformatics, Franklin College of Arts and Sciences, University of Georgia; Athens, GA 30602, USA
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia; Athens, GA 30602, USA
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia; Athens, GA 30602, USA
| |
Collapse
|
23
|
Cox F, Saeland E, Thoma A, van den Hoogen W, Tettero L, Drijver J, Vaneman C, van Polanen Y, Ritschel T, Bastian AR, Callendret B, Zahn R, van der Fits L. RSV A2-Based Prefusion F Vaccine Candidates Induce RSV A and RSV B Cross Binding and Neutralizing Antibodies and Provide Protection against RSV A and RSV B Challenge in Preclinical Models. Vaccines (Basel) 2023; 11:vaccines11030672. [PMID: 36992257 DOI: 10.3390/vaccines11030672] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/10/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023] Open
Abstract
RSV is divided into two antigenic subtypes, RSV A and RSV B, which is largely based on the variation in the G protein, while the fusion protein F is more conserved and a target for antibody-mediated neutralization. Here we evaluate the breadth of the protective immune responses across RSV A and RSV B subtypes, induced by vaccines based on the RSV A-based fusion protein, stabilized in the prefusion conformation (preF) in preclinical models. Immunization of naïve cotton rats with preF subunit or preF encoded by a replication incompetent Adenoviral 26, induced antibodies capable of neutralizing recent RSV A and RSV B clinical isolates, as well as protective efficacy against a challenge with RSV A and RSV B strains. Similarly, induction of cross-neutralizing antibodies was observed after immunization with Ad26-encoded preF, preF protein or a mix of both (Ad26/preF protein) in RSV pre-exposed mice and African Green Monkeys. Transfer of serum of human subjects immunized with Ad26/preF protein into cotton rats provide protection against challenges with both RSV A and RSV B, with complete protection against both strains observed in the lower respiratory tract. In contrast, almost no protection against RSV A and B infection was observed after the transfer of a human serum pool isolated pre-vaccination. These results collectively show that the RSV A-based monovalent Ad26/preF protein vaccine induced neutralizing antibodies, as well as protection against both RSV A and RSV B subtypes in animals, including by passive transfer of human antibodies alone, suggesting that clinical efficacy against both subtypes can be achieved.
Collapse
Affiliation(s)
- Freek Cox
- Janssen Vaccines & Prevention B.V. Archimedesweg 4-6, 2333 CN Leiden, The Netherlands
| | - Eirikur Saeland
- Janssen Vaccines & Prevention B.V. Archimedesweg 4-6, 2333 CN Leiden, The Netherlands
| | - Anne Thoma
- Janssen Vaccines & Prevention B.V. Archimedesweg 4-6, 2333 CN Leiden, The Netherlands
| | - Ward van den Hoogen
- Janssen Vaccines & Prevention B.V. Archimedesweg 4-6, 2333 CN Leiden, The Netherlands
| | - Lisanne Tettero
- Janssen Vaccines & Prevention B.V. Archimedesweg 4-6, 2333 CN Leiden, The Netherlands
| | - Joke Drijver
- Janssen Vaccines & Prevention B.V. Archimedesweg 4-6, 2333 CN Leiden, The Netherlands
| | - Cornelis Vaneman
- Janssen Vaccines & Prevention B.V. Archimedesweg 4-6, 2333 CN Leiden, The Netherlands
| | - Yolinda van Polanen
- Janssen Vaccines & Prevention B.V. Archimedesweg 4-6, 2333 CN Leiden, The Netherlands
| | - Tina Ritschel
- Janssen Vaccines & Prevention B.V. Archimedesweg 4-6, 2333 CN Leiden, The Netherlands
| | | | - Benoit Callendret
- Janssen Vaccines & Prevention B.V. Archimedesweg 4-6, 2333 CN Leiden, The Netherlands
| | - Roland Zahn
- Janssen Vaccines & Prevention B.V. Archimedesweg 4-6, 2333 CN Leiden, The Netherlands
| | - Leslie van der Fits
- Janssen Vaccines & Prevention B.V. Archimedesweg 4-6, 2333 CN Leiden, The Netherlands
| |
Collapse
|
24
|
Falsey AR, Williams K, Gymnopoulou E, Bart S, Ervin J, Bastian AR, Menten J, De Paepe E, Vandenberghe S, Chan EKH, Sadoff J, Douoguih M, Callendret B, Comeaux CA, Heijnen E. Efficacy and Safety of an Ad26.RSV.preF-RSV preF Protein Vaccine in Older Adults. N Engl J Med 2023; 388:609-620. [PMID: 36791161 DOI: 10.1056/nejmoa2207566] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) can cause serious lower respiratory tract disease in older adults, but no licensed RSV vaccine currently exists. An adenovirus serotype 26 RSV vector encoding a prefusion F (preF) protein (Ad26.RSV.preF) in combination with RSV preF protein was previously shown to elicit humoral and cellular immunogenicity. METHODS We conducted a randomized, double-blind, placebo-controlled, phase 2b, proof-of-concept trial to evaluate the efficacy, immunogenicity, and safety of an Ad26.RSV.preF-RSV preF protein vaccine. Adults who were 65 years of age or older were randomly assigned in a 1:1 ratio to receive vaccine or placebo. The primary end point was the first occurrence of RSV-mediated lower respiratory tract disease that met one of three case definitions: three or more symptoms of lower respiratory tract infection (definition 1), two or more symptoms of lower respiratory tract infection (definition 2), and either two or more symptoms of lower respiratory tract infection or one or more symptoms of lower respiratory tract infection plus at least one systemic symptom (definition 3). RESULTS Overall, 5782 participants were enrolled and received an injection. RSV-mediated lower respiratory tract disease meeting case definitions 1, 2, and 3 occurred in 6, 10, and 13 vaccine recipients and in 30, 40, and 43 placebo recipients, respectively. Vaccine efficacy was 80.0% (94.2% confidence interval [CI], 52.2 to 92.9), 75.0% (94.2% CI, 50.1 to 88.5), and 69.8% (94.2% CI, 43.7 to 84.7) for case definitions 1, 2, and 3, respectively. After vaccination, RSV A2 neutralizing antibody titers increased by a factor of 12.1 from baseline to day 15, a finding consistent with other immunogenicity measures. Percentages of participants with solicited local and systemic adverse events were higher in the vaccine group than in the placebo group (local, 37.9% vs. 8.4%; systemic, 41.4% vs. 16.4%); most adverse events were mild to moderate in severity. The frequency of serious adverse events was similar in the vaccine group and the placebo group (4.6% and 4.7%, respectively). CONCLUSIONS In adults 65 years of age or older, Ad26.RSV.preF-RSV preF protein vaccine was immunogenic and prevented RSV-mediated lower respiratory tract disease. (Funded by Janssen Vaccines and Prevention; CYPRESS ClinicalTrials.gov number, NCT03982199.).
Collapse
Affiliation(s)
- Ann R Falsey
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - Kristi Williams
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - Efi Gymnopoulou
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - Stephan Bart
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - John Ervin
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - Arangassery R Bastian
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - Joris Menten
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - Els De Paepe
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - Sjouke Vandenberghe
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - Eric K H Chan
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - Jerald Sadoff
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - Macaya Douoguih
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - Benoit Callendret
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - Christy A Comeaux
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| | - Esther Heijnen
- From the University of Rochester School of Medicine, Rochester, NY (A.R.F.); Janssen Vaccines and Prevention, Leiden, the Netherlands (K.W., A.R.B., J.S., M.D., B.C., C.A.C., E.H.); Janssen Infectious Diseases, Beerse, Belgium (E.G., J.M., E.D.P., S.V.); Trial Professionals Consultant Group, Woodstock, MD (S.B.); AMR Kansas City, Kansas City, MO (J.E.); and Janssen Global Services, Raritan, NJ (E.K.H.C.)
| |
Collapse
|
25
|
Jenkins VA, Hoet B, Hochrein H, De Moerlooze L. The Quest for a Respiratory Syncytial Virus Vaccine for Older Adults: Thinking beyond the F Protein. Vaccines (Basel) 2023; 11:382. [PMID: 36851260 PMCID: PMC9963583 DOI: 10.3390/vaccines11020382] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/19/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a common cause of paediatric respiratory tract infection and causes a significant health burden in older adults. Natural immunity to RSV is incomplete, permitting recurrent symptomatic infection over an individual's lifespan. When combined with immunosenescence, this increases older adults' susceptibility to more severe disease symptoms. As RSV prophylaxis is currently limited to infants, older adults represent an important target population for RSV vaccine development. The relationship between RSV and our immune systems is complex, and these interactions require deeper understanding to tailor an effective vaccine candidate towards older adults. To date, vaccine candidates targeting RSV antigens, including pre-F, F, G (A), G (B), M2-1, and N, have shown efficacy against RSV infection in older adults in clinical trial settings. Although vaccine candidates have demonstrated robust neutralising IgG and cellular responses, it is important that research continues to investigate the RSV immune response in order to further understand how the choice of antigenic target site may impact vaccine effectiveness. In this article, we discuss the Phase 3 vaccine candidates being tested in older adults and review the hurdles that must be overcome to achieve effective protection against RSV.
Collapse
|
26
|
Leroux-Roels I, Bruhwyler J, Stergiou L, Sumeray M, Joye J, Maes C, Lambert PH, Leroux-Roels G. Double-Blind, Placebo-Controlled, Dose-Escalating Study Evaluating the Safety and Immunogenicity of an Epitope-Specific Chemically Defined Nanoparticle RSV Vaccine. Vaccines (Basel) 2023; 11:vaccines11020367. [PMID: 36851245 PMCID: PMC9967611 DOI: 10.3390/vaccines11020367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND V-306 is a virus-like particle-based vaccine candidate displaying respiratory syncytial virus (RSV) F site II protein mimetics (FsIIm) as an antigenic epitope. METHODS This was a randomized, placebo-controlled, double-blind, dose-escalating, first-in-human study, conducted in 60 women aged 18-45 years. Twenty subjects per cohort (15 vaccine and five placebo) received two V-306 intramuscular administrations on Days 0 and 56 at 15 µg, 50 µg, or 150 µg. Safety and immunogenicity were assessed after each vaccination and for 1 year in total. RESULTS V-306 was safe and well tolerated at all dose levels, with no increase in reactogenicity and unsolicited adverse events between the first and second administrations. At 50 µg and 150 µg, V-306 induced an increase in FsIIm-specific immunoglobulin G (IgG) titers, which lasted at least 4 months. This did not translate into an increase in RSV-neutralizing antibody titers, which were already high at baseline. No increase in the anti-F protein-specific IgG titers was observed, which were also high in most subjects at baseline due to past natural infections. CONCLUSIONS V-306 was safe and well-tolerated. Future modifications of the vaccine and assay conditions will likely improve the results of vaccination.
Collapse
Affiliation(s)
- Isabel Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University Hospital, Corneel Heymanslaan 10, B-9000 Ghent, Belgium
| | - Jacques Bruhwyler
- Expert Clinical Services Organization (ECSOR) sa/nv, Rue de la Station 78, B-1630 Linkebeek, Belgium
| | - Lilli Stergiou
- Virometix AG, Wagistrasse 14, 8952 Schlieren, Switzerland
- Correspondence: ; Tel.: +41-4343-38660
| | - Mark Sumeray
- Virometix AG, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Jasper Joye
- Center for Vaccinology (CEVAC), Ghent University Hospital, Corneel Heymanslaan 10, B-9000 Ghent, Belgium
| | - Cathy Maes
- Center for Vaccinology (CEVAC), Ghent University Hospital, Corneel Heymanslaan 10, B-9000 Ghent, Belgium
| | - Paul-Henri Lambert
- Department of Paediatrics, Gynecology and Obstetrics, University of Geneva, Rue du Général Dufour 24, 1211 Geneva, Switzerland
| | - Geert Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University Hospital, Corneel Heymanslaan 10, B-9000 Ghent, Belgium
| |
Collapse
|
27
|
Chang LA, Phung E, Crank MC, Morabito KM, Villafana T, Dubovsky F, Falloon J, Esser MT, Lin BC, Chen GL, Graham BS, Ruckwardt TJ. A prefusion-stabilized RSV F subunit vaccine elicits B cell responses with greater breadth and potency than a postfusion F vaccine. Sci Transl Med 2022; 14:eade0424. [PMID: 36542692 PMCID: PMC11345946 DOI: 10.1126/scitranslmed.ade0424] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
There is currently no licensed vaccine for respiratory syncytial virus (RSV). Here, we assess the effect of RSV fusion protein (F) conformation on B cell responses in a post hoc comparison of samples from the DS-Cav1 [prefusion (pre-F)] and MEDI7510 [postfusion (post-F)] vaccine clinical trials. We compared the magnitude and quality of the serological and B cell responses across time points and vaccines. We measured RSV A and B neutralization, F-binding immunoglobulin G titers, and competition assays at week 0 (before vaccination) and week 4 (after vaccination) to evaluate antibody specificity and potency. To compare B cell specificity and activation, we used pre-F and post-F probes in tandem with a 17-color immunophenotyping flow cytometry panel at week 0 (before vaccination) and week 1 (after vaccination). Our data demonstrate that both DS-Cav1 and MEDI7510 vaccination robustly elicit F-specific antibodies and B cells, but DS-Cav1 elicited antibodies that more potently neutralized both RSV A and B. The superior potency was mediated by antibodies that bind antigenic sites on the apex of pre-F that are not present on post-F. In the memory (CD27+) B cell compartment, vaccination with DS-Cav1 or MEDI7510 elicited B cells with different epitope specificities. B cells preferentially binding the pre-F probe were activated in DS-Cav1-vaccinated participants but not in MEDI7510-vaccinated participants. Our findings emphasize the importance of using pre-F as an immunogen in humans because of its deterministic role in eliciting highly potent neutralizing antibodies and memory B cells.
Collapse
Affiliation(s)
- Lauren A. Chang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
- Present address: Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Present address: Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Authors contributed equally to this manuscript
| | - Emily Phung
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
- Present address: GlaxoSmithKline, Rockville, MD 20850, USA
- Authors contributed equally to this manuscript
| | - Michelle C. Crank
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
- Present address: Institute for Asthma and Allergy, Chevy Chase, MD 20815, USA
| | - Kaitlyn M. Morabito
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Tonya Villafana
- Vaccines & Immune Therapies, BioPharma R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Filip Dubovsky
- Vaccines & Immune Therapies, BioPharma R&D, AstraZeneca, Gaithersburg, MD 20878, USA
- Present address: Novavax, Gaithersburg, MD 20878, USA
| | - Judith Falloon
- Vaccines & Immune Therapies, BioPharma R&D, AstraZeneca, Gaithersburg, MD 20878, USA
- Present address: Horizon Therapeutics, Gaithersburg, MD 20878, USA
| | - Mark T. Esser
- Vaccines & Immune Therapies, BioPharma R&D, AstraZeneca, Gaithersburg, MD 20878, USA
| | - Bob C. Lin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
| | - Grace L. Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
- Present address: Moderna, Cambridge, MA 02139, USA
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
- Present address: Departments of Medicine and Microbiology, Biochemistry, and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Tracy J. Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD 20892, USA
- Senior author
- Lead contact
| |
Collapse
|
28
|
Montesinos-Guevara C, Buitrago-Garcia D, Felix ML, Guerra CV, Hidalgo R, Martinez-Zapata MJ, Simancas-Racines D. Vaccines for the common cold. Cochrane Database Syst Rev 2022; 12:CD002190. [PMID: 36515550 PMCID: PMC9749450 DOI: 10.1002/14651858.cd002190.pub6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The common cold is a spontaneously remitting infection of the upper respiratory tract, characterised by a runny nose, nasal congestion, sneezing, cough, malaise, sore throat, and fever (usually < 37.8 ºC). Whilst the common cold is generally not harmful, it is a cause of economic burden due to school and work absenteeism. In the United States, economic loss due to the common cold is estimated at more than USD 40 billion per year, including an estimate of 70 million workdays missed by employees, 189 million school days missed by children, and 126 million workdays missed by parents caring for children with a cold. Additionally, data from Europe show that the total cost per episode may be up to EUR 1102. There is also a large expenditure due to inappropriate antimicrobial prescription. Vaccine development for the common cold has been difficult due to antigenic variability of the common cold viruses; even bacteria can act as infective agents. Uncertainty remains regarding the efficacy and safety of interventions for preventing the common cold in healthy people, thus we performed an update of this Cochrane Review, which was first published in 2011 and updated in 2013 and 2017. OBJECTIVES To assess the clinical effectiveness and safety of vaccines for preventing the common cold in healthy people. SEARCH METHODS We searched the Cochrane Central Register of Controlled Trials (CENTRAL) (April 2022), MEDLINE (1948 to April 2022), Embase (1974 to April 2022), CINAHL (1981 to April 2022), and LILACS (1982 to April 2022). We also searched three trials registers for ongoing studies, and four websites for additional trials (April 2022). We did not impose any language or date restrictions. SELECTION CRITERIA Randomised controlled trials (RCTs) of any virus vaccine compared with placebo to prevent the common cold in healthy people. DATA COLLECTION AND ANALYSIS We used Cochrane's Screen4Me workflow to assess the initial search results. Four review authors independently performed title and abstract screening to identify potentially relevant studies. We retrieved the full-text articles for those studies deemed potentially relevant, and the review authors independently screened the full-text reports for inclusion in the review, recording reasons for exclusion of the excluded studies. Any disagreements were resolved by discussion or by consulting a third review author when needed. Two review authors independently collected data on a data extraction form, resolving any disagreements by consensus or by involving a third review author. We double-checked data transferred into Review Manager 5 software. Three review authors independently assessed risk of bias using RoB 1 tool as outlined in the Cochrane Handbook for Systematic Reviews of Interventions. We carried out statistical analysis using Review Manager 5. We did not conduct a meta-analysis, and we did not assess publication bias. We used GRADEpro GDT software to assess the certainty of the evidence and to create a summary of findings table. MAIN RESULTS: We did not identify any new RCTs for inclusion in this update. This review includes one RCT conducted in 1965 with an overall high risk of bias. The RCT included 2307 healthy young men in a military facility, all of whom were included in the analyses, and compared the effect of three adenovirus vaccines (live, inactivated type 4, and inactivated type 4 and 7) against a placebo (injection of physiological saline or gelatin capsule). There were 13 (1.14%) events in 1139 participants in the vaccine group, and 14 (1.19%) events in 1168 participants in the placebo group. Overall, we do not know if there is a difference between the adenovirus vaccine and placebo in reducing the incidence of the common cold (risk ratio 0.95, 95% confidence interval 0.45 to 2.02; very low-certainty evidence). Furthermore, no difference in adverse events when comparing live vaccine preparation with placebo was reported. We downgraded the certainty of the evidence to very low due to unclear risk of bias, indirectness because the population of this study was only young men, and imprecision because confidence intervals were wide and the number of events was low. The included study did not assess vaccine-related or all-cause mortality. AUTHORS' CONCLUSIONS: This Cochrane Review was based on one study with very low-certainty evidence, which showed that there may be no difference between the adenovirus vaccine and placebo in reducing the incidence of the common cold. We identified a need for well-designed, adequately powered RCTs to investigate vaccines for the common cold in healthy people. Future trials on interventions for preventing the common cold should assess a variety of virus vaccines for this condition, and should measure such outcomes as common cold incidence, vaccine safety, and mortality (all-cause and related to the vaccine).
Collapse
Affiliation(s)
- Camila Montesinos-Guevara
- Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Diana Buitrago-Garcia
- Institute of Social and Preventive Medicine (ISPM), Graduate School of Health Sciences, University of Bern, Bern, Switzerland
| | - Maria L Felix
- Departamento de Neonatología, Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Claudia V Guerra
- Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Ricardo Hidalgo
- Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| | - Maria José Martinez-Zapata
- Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
- Iberoamerican Cochrane Centre, Biomedical Research Institute Sant Pau (IIB Sant Pau), CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Daniel Simancas-Racines
- Cochrane Ecuador, Centro de Investigación en Salud Pública y Epidemiología Clínica (CISPEC), Facultad de Ciencias de la Salud Eugenio Espejo, Universidad UTE, Quito, Ecuador
| |
Collapse
|
29
|
Qiu X, Xu S, Lu Y, Luo Z, Yan Y, Wang C, Ji J. Development of mRNA vaccines against respiratory syncytial virus (RSV). Cytokine Growth Factor Rev 2022; 68:37-53. [PMID: 36280532 DOI: 10.1016/j.cytogfr.2022.10.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 02/06/2023]
Abstract
Respiratory syncytial virus (RSV) is a single-stranded negative-sense RNA virus that is the primary etiologic pathogen of bronchitis and pneumonia in infants and the elderly. Currently, no preventative vaccine has been approved for RSV infection. However, advances in the characterization, and structural resolution, of the RSV surface fusion glycoprotein have revolutionized RSV vaccine development by providing a new target for preventive interventions. In general, six different approaches have been adopted in the development of preventative RSV therapeutics, namely, particle-based vaccines, vector-based vaccines, live-attenuated or chimeric vaccines, subunit vaccines, mRNA vaccines, and monoclonal antibodies. Among these preventive interventions, MVA-BN-RSV, RSVpreF3, RSVpreF, Ad26. RSV.preF, nirsevimab, clesrovimab and mRNA-1345 is being tested in phase 3 clinical trials, and displays the most promising in infant or elderly populations. Accompanied by the huge success of mRNA vaccines in COVID-19, mRNA vaccines have been rapidly developed, with many having entered clinical studies, in which they have demonstrated encouraging results and acceptable safety profiles. In fact, Moderna has received FDA approval, granting fast-track designation for an investigational single-dose mRNA-1345 vaccine against RSV in adults over 60 years of age. Hence, mRNA vaccines may represent a new, more successful, chapter in the continued battle to develop effective preventative measures against RSV. This review discusses the structure, life cycle, and brief history of RSV, while also presenting the current advancements in RSV preventatives, with a focus on the latest progress in RSV mRNA vaccine development. Finally, future prospects for this field are presented.
Collapse
Affiliation(s)
- Xirui Qiu
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Siyan Xu
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Lu
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zichen Luo
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yangtian Yan
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chuyue Wang
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianjian Ji
- Jiangsu Key Laboratory of Pediatric Respiratory Disease, Institute of Pediatrics, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
30
|
Savic M, Penders Y, Shi T, Branche A, Pirçon J. Respiratory syncytial virus disease burden in adults aged 60 years and older in high-income countries: A systematic literature review and meta-analysis. Influenza Other Respir Viruses 2022; 17:e13031. [PMID: 36369772 PMCID: PMC9835463 DOI: 10.1111/irv.13031] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV)-associated acute respiratory infection (ARI) is an underrecognized cause of illness in older adults. We conducted a systematic literature review and meta-analysis to estimate the RSV disease burden in adults ≥60 years in high-income countries. METHODS Data on RSV-ARI and hospitalization attack rates and in-hospital case fatality rates (hCFR) in adults ≥60 years from the United States, Canada, European countries, Japan, and South Korea were collected based on a systematic literature search (January 1, 2000-November 3, 2021) or via other methods (citation search, unpublished studies cited by a previous meta-analysis, gray literature, and an RSV-specific abstract booklet). A random effects meta-analysis was performed on estimates from the included studies. RESULTS Twenty-one studies were included in the meta-analysis. The pooled estimates were 1.62% (95% confidence interval [CI]: 0.84-3.08) for RSV-ARI attack rate, 0.15% (95% CI: 0.09-0.22) for hospitalization attack rate, and 7.13% (95% CI: 5.40-9.36) for hCFR. In 2019, this would translate into approximately 5.2 million cases, 470,000 hospitalizations, and 33,000 in-hospital deaths in ≥60-year-old adults in high-income countries. CONCLUSIONS RSV disease burden in adults aged ≥60 years in high-income countries is higher than previously estimated, highlighting the need for RSV prophylaxis in this age group.
Collapse
Affiliation(s)
| | | | - Ting Shi
- Usher InstituteUniversity of EdinburghEdinburghUK
| | - Angela Branche
- Division of Infectious Diseases, Department of MedicineUniversity of RochesterRochesterNew YorkUSA
| | | |
Collapse
|
31
|
Soto JA, Galvez NMS, Rivera DB, Díaz FE, Riedel CA, Bueno SM, Kalergis AM. From animal studies into clinical trials: the relevance of animal models to develop vaccines and therapies to reduce disease severity and prevent hRSV infection. Expert Opin Drug Discov 2022; 17:1237-1259. [PMID: 36093605 DOI: 10.1080/17460441.2022.2123468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Human respiratory syncytial virus (hRSV) is an important cause of lower respiratory tract infections in the pediatric and the geriatric population worldwide. There is a substantial economic burden resulting from hRSV disease during winter. Although no vaccines have been approved for human use, prophylactic therapies are available for high-risk populations. Choosing the proper animal models to evaluate different vaccine prototypes or pharmacological treatments is essential for developing efficient therapies against hRSV. AREAS COVERED This article describes the relevance of using different animal models to evaluate the effect of antiviral drugs, pharmacological molecules, vaccine prototypes, and antibodies in the protection against hRSV. The animal models covered are rodents, mustelids, bovines, and nonhuman primates. Animals included were chosen based on the available literature and their role in the development of the drugs discussed in this manuscript. EXPERT OPINION Choosing the correct animal model is critical for exploring and testing treatments that could decrease the impact of hRSV in high-risk populations. Mice will continue to be the most used preclinical model to evaluate this. However, researchers must also explore the use of other models such as nonhuman primates, as they are more similar to humans, prior to escalating into clinical trials.
Collapse
Affiliation(s)
- J A Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - N M S Galvez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - D B Rivera
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - F E Díaz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - C A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - S M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
32
|
Stuart ASV, Virta M, Williams K, Seppa I, Hartvickson R, Greenland M, Omoruyi E, Bastian AR, Haazen W, Salisch N, Gymnopoulou E, Callendret B, Faust SN, Snape MD, Heijnen E. Phase 1/2a Safety and Immunogenicity of an Adenovirus 26 Vector Respiratory Syncytial Virus (RSV) Vaccine Encoding Prefusion F in Adults 18-50 Years and RSV-Seropositive Children 12-24 Months. J Infect Dis 2022; 227:71-82. [PMID: 36259542 PMCID: PMC9796164 DOI: 10.1093/infdis/jiac407] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/28/2022] [Accepted: 10/17/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) remains a leading cause of pediatric morbidity, with no approved vaccine. We assessed the safety and immunogenicity of the Ad26.RSV.preF vaccine candidate in adults and children. METHODS In this randomized, double-blind, phase 1/2a, placebo-controlled study, 12 adults (18-50 years) and 36 RSV-seropositive children (12-24 months) were randomized 2:1 to Ad26.RSV.preF (1 × 1011 viral particles [vp] for adults, 5 × 1010 vp for children) or placebo, at day 1 and 29, with 6-month immunogenicity and 1-year safety follow-up. Respiratory syncytial virus infection was an exploratory outcome in children. RESULTS In adults, solicited adverse events (AEs) were generally mild to moderate, with no serious AEs. In children, no vaccination-related serious AEs were reported; fever was reported in 14 (58.3%) Ad26.RSV.preF recipients. Baseline pediatric geometric mean titers for RSV A2 neutralization increased from 121 (95% confidence interval [CI], 76-191) to 1608 (95% CI, 730-3544) at day 29, and 2235 (95% CI, 1586-3150) at day 57, remaining elevated over 7 months. Respiratory syncytial virus infection was confirmed in fewer children receiving Ad26.RSV.preF (1, 4.2%) than placebo (5, 41.7%). CONCLUSIONS Ad26.RSV.preF demonstrated immunogenicity in healthy adults and toddlers, with no safety concerns raised. Evaluations in RSV-seronegative children are underway.
Collapse
Affiliation(s)
- Arabella S V Stuart
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | | | | | | | | | - Melanie Greenland
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | | | | | - Wouter Haazen
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | - Nadine Salisch
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | | | | | - Saul N Faust
- NIHR Southampton Clinical Research Facility and NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom,Faculty of Medicine and Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Matthew D Snape
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, United Kingdom,Oxford NIHR – Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Esther Heijnen
- Correspondence: Esther Heijnen, MD, Janssen Vaccines & Prevention BV, Leiden, 2333 CN, The Netherlands ()
| |
Collapse
|
33
|
Brakel KA, Ma Y, Binjawadagi R, Harder O, Watts M, Li J, Binjawadagi B, Niewiesk S. Codon-optimization of the respiratory syncytial virus (RSV) G protein expressed in a vesicular stomatitis virus (VSV) vector improves immune responses in a cotton rat model. Virology 2022; 575:101-110. [PMID: 36096069 DOI: 10.1016/j.virol.2022.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 08/17/2022] [Accepted: 08/31/2022] [Indexed: 10/14/2022]
Abstract
Respiratory syncytial virus is an important cause of pneumonia in children, the elderly, and immunocompromised individuals. The attachment (G) protein of RSV generates neutralizing antibodies in natural RSV infection which correlate with protection against disease. The immune response to RSV is typically short-lived, which may be related to the heavy glycosylation of RSV-G. In order to improve its immunogenicity, we expressed G protein mutants in a vesicular stomatitis virus (VSV) vector system and tested their ability to protect cotton rats from RSV challenge. We found that the most protective construct was codon-optimized RSV-G, followed by wild-type G and membrane-bound G. Constructs which expressed the G protein with reduced glycosylation or the secreted G protein provided either partial or no protection. Our results demonstrate that modifications to the G protein are not advantageous in a VSV vector system, and that an intact, codon-optimized G is a superior vaccine candidate.
Collapse
Affiliation(s)
- Kelsey A Brakel
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Yuanmei Ma
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Rashmi Binjawadagi
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Olivia Harder
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Mauria Watts
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Jianrong Li
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Basavaraj Binjawadagi
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States; Ceva Sante Animale, Lenexa, KS, United States
| | - Stefan Niewiesk
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States.
| |
Collapse
|
34
|
Simões EAF. Respiratory Syncytial Virus Disease in Young Children and Older Adults in Europe: A Burden and Economic Perspective. J Infect Dis 2022; 226:S1-S9. [PMID: 35822854 DOI: 10.1093/infdis/jiac252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 06/22/2022] [Indexed: 01/07/2023] Open
Affiliation(s)
- Eric A F Simões
- Department of Pediatrics, Children's Hospital Colorado, Aurora, Colorado, USA, Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, USA, and Department of Epidemiology, Colorado School of Public Health, Aurora, Colorado, USA
| |
Collapse
|
35
|
Phung E, Chang LA, Mukhamedova M, Yang L, Nair D, Rush SA, Morabito KM, McLellan JS, Buchholz UJ, Mascola JR, Crank MC, Chen G, Graham BS, Ruckwardt TJ. Elicitation of pneumovirus-specific B cell responses by a prefusion-stabilized respiratory syncytial virus F subunit vaccine. Sci Transl Med 2022; 14:eabo5032. [PMID: 35731888 PMCID: PMC11340646 DOI: 10.1126/scitranslmed.abo5032] [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] [Indexed: 11/02/2022]
Abstract
Respiratory syncytial virus (RSV) is a substantial cause of morbidity and mortality globally. A candidate RSV prefusion (pre-F)-stabilized subunit vaccine, DS-Cav1, has previously been shown to elicit potent and durable neutralizing activity in a phase 1 clinical trial in healthy adults. Here, we used fluorescently labeled probes and flow cytometry to evaluate the antigen specificity and phenotype of RSV F-specific B cells longitudinally after DS-Cav1 immunization. Peripheral blood mononuclear cells (PBMCs) collected at time points before the first immunization through the end of the trial at 44 weeks were assessed by flow cytometry. Our data demonstrate a rapid increase in the frequency of pre-F-specific IgG+ and IgA+ B cells after the first immunization and a modest increase after a second immunization at week 12. Nearly all F-specific B cells down-regulated CD21 and up-regulated the proliferation marker CD71 after the first immunization, with less pronounced activation after the second immunization. Memory B cells (CD27+CD21+) specific for pre-F remained elevated above baseline at 44 weeks after vaccination. DS-Cav1 vaccination also activated human metapneumovirus (HMPV) cross-reactive B cells capable of binding prefusion-stabilized HMPV F protein and increased HMPV F-binding antibodies and neutralizing activity for HMPV in some participants. In summary, vaccination with RSV pre-F resulted in the expansion and activation of RSV and HMPV F-specific B cells that were maintained above baseline for at least 10 months and could contribute to long-term pneumovirus immunity.
Collapse
Affiliation(s)
- Emily Phung
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Present address: GlaxoSmithKline, Rockville, MD 20850, USA
| | - Lauren A. Chang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Present address: Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Maryam Mukhamedova
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lijuan Yang
- RNA Viruses Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Deepika Nair
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Scott A. Rush
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Kaitlyn M. Morabito
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jason S. McLellan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Ursula J. Buchholz
- RNA Viruses Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - John R. Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michelle C. Crank
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Present address: Institute for Asthma and Allergy, Chevy Chase, MD 20815, USA
| | - Grace Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Present address: Moderna, Cambridge, MA 02139, USA
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tracy J. Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
36
|
Endt K, Wollmann Y, Haug J, Bernig C, Feigl M, Heiseke A, Kalla M, Hochrein H, Suter M, Chaplin P, Volkmann A. A Recombinant MVA-Based RSV Vaccine Induces T-Cell and Antibody Responses That Cooperate in the Protection Against RSV Infection. Front Immunol 2022; 13:841471. [PMID: 35774800 PMCID: PMC9238321 DOI: 10.3389/fimmu.2022.841471] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/13/2022] [Indexed: 11/25/2022] Open
Abstract
Respiratory syncytial virus (RSV) causes a respiratory disease with a potentially fatal outcome especially in infants and elderly individuals. Several vaccines failed in pivotal clinical trials, and to date, no vaccine against RSV has been licensed. We have developed an RSV vaccine based on the recombinant Modified Vaccinia Virus Ankara-BN® (MVA-RSV), containing five RSV-specific antigens that induced antibody and T-cell responses, which is currently tested in clinical trials. Here, the immunological mechanisms of protection were evaluated to determine viral loads in lungs upon vaccination of mice with MVA-RSV followed by intranasal RSV challenge. Depletion of CD4 or CD8 T cells, serum transfer, and the use of genetically engineered mice lacking the ability to generate either RSV-specific antibodies (T11µMT), the IgA isotype (IgA knockout), or CD8 T cells (β2M knockout) revealed that complete protection from RSV challenge is dependent on CD4 and CD8 T cells as well as antibodies, including IgA. Thus, MVA-RSV vaccination optimally protects against RSV infection by employing multiple arms of the adaptive immune system.
Collapse
Affiliation(s)
- Kathrin Endt
- Bavarian Nordic GmbH, Martinsried, Germany
- *Correspondence: Kathrin Endt,
| | | | - Jana Haug
- Bavarian Nordic GmbH, Martinsried, Germany
| | | | | | | | | | | | - Mark Suter
- University of Zürich, Dekanat Vetsuisse-Fakultät Immunology, Zurich, Switzerland
| | | | | |
Collapse
|
37
|
Baber J, Arya M, Moodley Y, Jaques A, Jiang Q, Swanson KA, Cooper D, Maddur MS, Loschko J, Gurtman A, Jansen KU, Gruber WC, Dormitzer PR, Schmoele-Thoma B. A Phase 1/2 Study of a Respiratory Syncytial Virus Prefusion F Vaccine With and Without Adjuvant in Healthy Older Adults. J Infect Dis 2022; 226:2054-2063. [PMID: 35543281 PMCID: PMC9749002 DOI: 10.1093/infdis/jiac189] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/29/2022] [Accepted: 05/09/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is an important cause of disease in older adults. We evaluated the safety and immunogenicity of a stabilized RSV prefusion F subunit (RSVpreF) vaccine candidate with/without adjuvant in adults aged 65-85 years. METHODS Primary cohort participants were equally randomized to 1 of 7 RSVpreF formulations: 60 µg with either Al(OH)3 or CpG/Al(OH)3, 120 µg with either Al(OH)3 or CpG/Al(OH)3, 240 µg with either Al(OH)3 or CpG/Al(OH)3, 240 µg unadjuvanted, or placebo, administered concomitantly with high-dose seasonal inactivated influenza vaccine (SIIV). Participants in the month 0,2 cohort were randomized to RSVpreF 240 µg with CpG/Al(OH)3 or placebo, administered at months 0 and 2. RESULTS All RSVpreF vaccine candidates elicited robust and persistent serum neutralizing responses when administered alone or with SIIV. There was no notable difference in neutralizing response between the formulations, including those containing CpG. In the month 0,2 cohort, there was no booster effect of dose 2. SIIV responses were similar or slightly lower with concomitant administration of RSVpreF. Most systemic and local reactions were mild and more frequent after RSVpreF than placebo. CONCLUSIONS RSVpreF formulations were well tolerated and elicited robust neutralizing responses in older adults; however, CpG/Al(OH)3 did not further enhance responses. Clinical Trials Registration. NCT03572062.
Collapse
Affiliation(s)
- James Baber
- Correspondence: James Baber, MBChB, MPH, Pfizer Vaccine Clinical Research and Development, Level 15-18, 151 Clarence Street, Sydney 2000, Australia ()
| | - Mark Arya
- Australian Clinical Research Network, Maroubra, Australia
| | - Yuben Moodley
- School of Medicine, University of Western Australia, Perth, WA, Australia
| | - Anna Jaques
- Vaccine Clinical Research, Pfizer Inc, Sydney, Australia
| | - Qin Jiang
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - Kena A Swanson
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - David Cooper
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - Mohan S Maddur
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - Jakob Loschko
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - Alejandra Gurtman
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - Kathrin U Jansen
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - William C Gruber
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - Philip R Dormitzer
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | | |
Collapse
|
38
|
Walsh EE, Falsey AR, Scott DA, Gurtman A, Zareba AM, Jansen KU, Gruber WC, Dormitzer P, Swanson KA, Radley D, Gomme E, Cooper D, Schmoele-Thoma B. A Randomized Phase 1/2 Study of a Respiratory Syncytial Virus Prefusion F Vaccine. J Infect Dis 2022; 225:1357-1366. [PMID: 34932102 PMCID: PMC9016447 DOI: 10.1093/infdis/jiab612] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/16/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Protection against human respiratory syncytial virus (RSV) remains an unmet need potentially addressable by maternal immunization. This phase 1/2 study evaluated a bivalent prefusion F vaccine (RSVpreF) with antigens from RSV subgroups A and B. METHODS Adults 18-49 years old (N = 618) were randomized to receive placebo or 60, 120, or 240 µg RSVpreF with or without Al(OH)3. Safety and immunogenicity were evaluated. RESULTS RSVpreF recipients more frequently reported local reactions and systemic events than placebo recipients; these were mostly mild or moderate. No vaccine-related serious adverse events occurred through 12 months postvaccination. All RSVpreF formulations induced 1-month postvaccination virus-neutralizing titers higher than those associated with protection of high-risk infants by palivizumab, the only prophylactic currently available for RSV. Geometric mean fold rises (GMFRs) across RSVpreF doses/formulations were 10.6-16.9 for RSV A and 10.3-19.8 for RSV B at 1 month postvaccination, greater than those historically elicited by postfusion F vaccines. GMFRs were 3.9-5.2 and 3.7-5.1, respectively, at 12 months postvaccination. CONCLUSIONS RSVpreF formulations were safe, well tolerated, and induced robust neutralizing responses in adults. These findings support development of RSVpreF, which is being evaluated in a pivotal phase 3 study for maternal immunization. CLINICAL TRIALS REGISTRATION NCT03529773.
Collapse
Affiliation(s)
- Edward E Walsh
- Department of Medicine, Infectious Diseases Division, Rochester General Hospital and University of Rochester Medical Center, Rochester, New York, USA
| | - Ann R Falsey
- Department of Medicine, Infectious Diseases Division, Rochester General Hospital and University of Rochester Medical Center, Rochester, New York, USA
| | - Daniel A Scott
- Vaccine Research and Development, Pfizer Inc, Collegeville, Pennsylvania, USA
| | - Alejandra Gurtman
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - Agnieszka M Zareba
- Vaccine Research and Development, Pfizer Inc, Collegeville, Pennsylvania, USA
| | - Kathrin U Jansen
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - William C Gruber
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - Philip R Dormitzer
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - Kena A Swanson
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - David Radley
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - Emily Gomme
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | - David Cooper
- Vaccine Research and Development, Pfizer Inc, Pearl River, New York, USA
| | | |
Collapse
|
39
|
Nanishi E, Angelidou A, Rotman C, Dowling DJ, Levy O, Ozonoff A. Precision Vaccine Adjuvants for Older Adults: A Scoping Review. Clin Infect Dis 2022; 75:S72-S80. [PMID: 35439286 PMCID: PMC9376277 DOI: 10.1093/cid/ciac302] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Older adults, defined as those ≥60 years of age, are a growing population vulnerable to infections including severe acute respiratory syndrome coronavirus 2. Although immunization is a key to protecting this population, immunosenescence can impair responses to vaccines. Adjuvants can increase the immunogenicity of vaccine antigens but have not been systematically compared in older adults. We conducted a scoping review to assess the comparative effectiveness of adjuvants in aged populations. Adjuvants AS01, MF59, AS03, and CpG-oligodeoxynucleotide, included in licensed vaccines, are effective in older human adults. A growing menu of investigational adjuvants, such as Matrix-M and CpG plus alum, showed promising results in early phase clinical trials and preclinical studies. Most studies assessed only 1 or 2 adjuvants and no study has directly compared >3 adjuvants among older adults. Enhanced preclinical approaches enabling direct comparison of multiple adjuvants including human in vitro modeling and age-specific animal models may derisk and accelerate vaccine development for older adults.
Collapse
Affiliation(s)
| | | | - Chloe Rotman
- Medical Library, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - David J Dowling
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital,Boston, Massachusetts, USA,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Ofer Levy
- Correspondence: O. Levy, Precision Vaccines Program, Boston Children’s Hospital, Boston, MA 02115 ()
| | - Al Ozonoff
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children’s Hospital,Boston, Massachusetts, USA,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA,Broad Institute of MIT & Harvard, Cambridge, Massachusetts, USA
| |
Collapse
|
40
|
Falsey AR, Walsh EE, Scott DA, Gurtman A, Zareba A, Jansen KU, Gruber WC, Dormitzer PR, Swanson KA, Jiang Q, Gomme E, Cooper D, Schmoele-Thoma B. Phase 1/2 Randomized Study of the Immunogenicity, Safety and Tolerability of an RSV Prefusion F Vaccine in Adults With Concomitant Inactivated Influenza Vaccine. J Infect Dis 2021; 225:2056-2066. [PMID: 34931667 PMCID: PMC9200152 DOI: 10.1093/infdis/jiab611] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 12/20/2021] [Indexed: 12/05/2022] Open
Abstract
Background Respiratory syncytial virus (RSV) causes substantial morbidity and mortality in older adults and adults with comorbidities. An effective vaccine is needed. An investigational bivalent prefusion F vaccine (RSVpreF) was assessed in healthy adults. Methods This phase 1/2 study randomized adults 18–85 years old to receive placebo or 60, 120, or 240 µg RSVpreF (with or without aluminum hydroxide) alone or concomitantly with seasonal inactivated influenza vaccine (SIIV). Safety and immunogenicity were assessed. Results In older adults, reactogenicity events were predominantly mild or moderate among RSVpreF recipients; adverse events through 1 month postvaccination were similar across formulations. Coadministration with SIIV did not appear to affect safety among younger or older adults. All RSVpreF formulations with or without concomitant SIIV elicited robust RSV serum-neutralizing responses in adults aged 50–85 years 1 month postvaccination. Neutralizing titers 1 and 12 months postvaccination were 6.9–14.9 and 2.9–4.5 times, respectively, those before vaccination. SIIV immune responses trended lower when coadministered with RSVpreF. Conclusions RSVpreF formulations administered alone or with SIIV were well tolerated and highly immunogenic in older adults, supporting the potential for RSVpreF to protect older adults from RSV disease. Clinical Trials Registration NCT03529773.
Collapse
Affiliation(s)
- Ann R Falsey
- Department of Medicine, Infectious Diseases Division, Rochester General Hospital and University of Rochester Medical Center, Rochester, NY, 14642 USA
| | - Edward E Walsh
- Department of Medicine, Infectious Diseases Division, Rochester General Hospital and University of Rochester Medical Center, Rochester, NY, 14642 USA
| | - Daniel A Scott
- Vaccine Research and Development, Pfizer Inc, Collegeville, PA, 19426 USA
| | - Alejandra Gurtman
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, 10965 USA
| | - Agnieszka Zareba
- Vaccine Research and Development, Pfizer Inc, Collegeville, PA, 19426 USA
| | - Kathrin U Jansen
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, 10965 USA
| | - William C Gruber
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, 10965 USA
| | - Philip R Dormitzer
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, 10965 USA
| | - Kena A Swanson
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, 10965 USA
| | - Qin Jiang
- Vaccine Research and Development, Pfizer Inc, Collegeville, PA, 19426 USA
| | - Emily Gomme
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, 10965 USA
| | - David Cooper
- Vaccine Research and Development, Pfizer Inc, Pearl River, NY, 10965 USA
| | | | | |
Collapse
|
41
|
Brakel KA, Binjawadagi B, French-Kim K, Watts M, Harder O, Ma Y, Li J, Niewiesk S. Coexpression of respiratory syncytial virus (RSV) fusion (F) protein and attachment glycoprotein (G) in a vesicular stomatitis virus (VSV) vector system provides synergistic effects against RSV infection in a cotton rat model. Vaccine 2021; 39:6817-6828. [PMID: 34702618 PMCID: PMC8595748 DOI: 10.1016/j.vaccine.2021.10.042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 10/11/2021] [Accepted: 10/19/2021] [Indexed: 11/20/2022]
Abstract
Respiratory syncytial virus (RSV) is one of the most important causes of respiratory disease in infants, immunocompromised individuals, and the elderly. Natural infection does not result in long-term immunity, and there is no licensed vaccine. Vesicular stomatitis virus (VSV) is a commonly used vaccine vector platform against infectious diseases, and has been used as a vector for a licensed Ebola vaccine. In this study, we expressed the RSV fusion (F) protein, the RSV F protein stabilized in either a pre-fusion or a post-fusion configuration, the attachment glycoprotein (G), or the G and F proteins of RSV in combination in a VSV vector. Cotton rats were immunized with these recombinants intranasally or subcutaneously to test immunogenicity. RSV F stabilized in either a pre-fusion or a post-fusion configuration proved to be poorly immunogenic and protective when compared to unmodified F. RSV G provided partial protection and moderate levels of neutralizing antibody production, both of which improved with intranasal administration compared to subcutaneous inoculation. The most successful vaccine vector was VSV expressing both the G and F proteins after intranasal inoculation. Immunization with this recombinant induced neutralizing antibodies and provided protection from RSV challenge in the upper and lower respiratory tract for at least 80 days. Our results demonstrate that co-expression of F and G proteins in a VSV vector provides synergistic effects in inducing RSV-specific neutralizing antibodies and protection against RSV infection.
Collapse
Affiliation(s)
- Kelsey A Brakel
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States.
| | - Basavaraj Binjawadagi
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States; Ceva Sante Animale, Lenexa, KS, United States
| | - Kristen French-Kim
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Mauria Watts
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Olivia Harder
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Yuanmei Ma
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Jianrong Li
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| | - Stefan Niewiesk
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
42
|
Dong Y, Dai T, Wang B, Zhang L, Zeng LH, Huang J, Yan H, Zhang L, Zhou F. The way of SARS-CoV-2 vaccine development: success and challenges. Signal Transduct Target Ther 2021; 6:387. [PMID: 34753918 PMCID: PMC8575680 DOI: 10.1038/s41392-021-00796-w] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/10/2021] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of coronavirus disease 2019 (COVID-19). To halt the pandemic, multiple SARS-CoV-2 vaccines have been developed and several have been allowed for emergency use and rollout worldwide. With novel SARS-CoV-2 variants emerging and circulating widely, whether the original vaccines that were designed based on the wild-type SARS-CoV-2 were effective against these variants has been a contentious discussion. Moreover, some studies revealed the long-term changes of immune responses post SARS-CoV-2 infection or vaccination and the factors that might impact the vaccine-induced immunity. Thus, in this review, we have summarized the influence of mutational hotspots on the vaccine efficacy and characteristics of variants of interest and concern. We have also discussed the reasons that might result in discrepancies in the efficacy of different vaccines estimated in different trials. Furthermore, we provided an overview of the duration of immune responses after natural infection or vaccination and shed light on the factors that may affect the immunity induced by the vaccines, such as special disease conditions, sex, and pre-existing immunity, with the aim of aiding in combating COVID-19 and distributing SARS-CoV-2 vaccines under the prevalence of diverse SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Yetian Dong
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, Zhejiang, China
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Rui'an, China
| | - Tong Dai
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Bin Wang
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Lei Zhang
- Department of Orthopaedic Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Rui'an, China
| | - Ling-Hui Zeng
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, Zhejiang, China
| | - Jun Huang
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Haiyan Yan
- School of Medicine, Zhejiang University City College, Hangzhou, 310015, Zhejiang, China
| | - Long Zhang
- Life Sciences Institute and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China.
| |
Collapse
|
43
|
Falsey AR, Walsh EE, Osborne RH, Vandendijck Y, Ren X, Witek J, Kang D, Chan E, Scott J, Ispas G. Comparative assessment of reported symptoms of influenza, respiratory syncytial virus, and human metapneumovirus infection during hospitalization and post-discharge assessed by Respiratory Intensity and Impact Questionnaire. Influenza Other Respir Viruses 2021; 16:79-89. [PMID: 34472708 PMCID: PMC8692817 DOI: 10.1111/irv.12903] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/15/2021] [Indexed: 11/28/2022] Open
Abstract
Background The hospitalized acute respiratory tract infection (HARTI) study used the Respiratory Intensity and Impact Questionnaire (RiiQ™) Symptom Scale, derived from FluiiQ™, to assess and compare the burden of respiratory infection symptoms for patients with influenza, respiratory syncytial virus (RSV), and human metapneumovirus (hMPV) infection, with or without core risk factors (CRF) (age ≥65; chronic heart, renal, obstructive pulmonary disease; asthma). Methods This was a prospective cohort study in adult patients hospitalized with acute respiratory tract infection (40 centers, 12 countries) during two consecutive influenza/RSV/hMPV seasons (2017–2019). The RiiQ™ Symptom Scale and EuroQol 5‐Dimensions 5‐Levels (EQ‐5D‐5L) were assessed by interview at two timepoints during hospitalization and at 1, 2, and 3 months post‐discharge. Results Mean lower respiratory tract (LRT) symptom scores were higher for RSV and hMPV participants compared to influenza at 48 h after enrollment/early discharge (p = 0.001) and 3 months post‐discharge (p = 0.007). This was driven by LRT symptoms, including shortness of breath (SOB) (p < 0.01) and wheezing (p < 0.01) during hospitalization, and SOB (p < 0.05) and cough (p < 0.05) post‐discharge. Participants with CRF reported more moderate‐to‐severe SOB (p < 0.05) and wheezing (p < 0.05) compared to CRF(−) participants post‐discharge. EQ‐5D‐5L scores were moderately associated with RiiQ™ LRT and systemic symptoms domains. Conclusions Results from the HARTI study suggest that in the study population, LRT symptoms were more severe for RSV and hMPV groups and for patients with CRF. RiiQ™ Symptom Scale scores shows a moderate association with EQ‐5D‐5L indicating that the RiiQ™ may provide useful insights and offer advantages over other measures for use in interventional RSV adult clinical studies.
Collapse
Affiliation(s)
- Ann R Falsey
- School of Medicine Rochester Regional Health, University of Rochester, Rochester, New York, USA
| | - Edward E Walsh
- School of Medicine Rochester Regional Health, University of Rochester, Rochester, New York, USA
| | - Richard H Osborne
- Department of Health and Medical Sciences, Swinburne University of Technology, Melbourne, Victoria, Australia
| | | | - Xiaohui Ren
- Janssen Infectious Diseases, Beerse, Belgium
| | - James Witek
- Janssen Research & Development, LLC, Titusville, New Jersey, USA
| | - Diye Kang
- Janssen Infectious Diseases, Beerse, Belgium
| | - Eric Chan
- Janssen Global Services, LLC, Raritan, New Jersey, USA
| | - Jane Scott
- Janssen Global Services, LLC, Raritan, New Jersey, USA
| | | |
Collapse
|
44
|
Harshbarger W, Abeyrathne PD, Tian S, Huang Y, Chandramouli S, Bottomley MJ, Malito E. Improved epitope resolution of the prefusion trimer-specific antibody AM14 bound to the RSV F glycoprotein. MAbs 2021; 13:1955812. [PMID: 34420474 PMCID: PMC8386734 DOI: 10.1080/19420862.2021.1955812] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the most common cause of acute lower respiratory tract infections resulting in medical intervention and hospitalizations during infancy and early childhood, and vaccination against RSV remains a public health priority. The RSV F glycoprotein is a major target of neutralizing antibodies, and the prefusion stabilized form of F (DS-Cav1) is under investigation as a vaccine antigen. AM14 is a human monoclonal antibody with the exclusive capacity of binding an epitope on prefusion F (PreF), which spans two F protomers. The quality of recognizing a trimer-specific epitope makes AM14 valuable for probing PreF-based immunogen conformation and functionality during vaccine production. Currently, only a low-resolution (5.5 Å) X-ray structure is available of the PreF-AM14 complex, revealing few reliable details of the interface. Here, we perform complementary structural studies using X-ray crystallography and cryo-electron microscopy (cryo-EM) to provide improved resolution structures at 3.6 Å and 3.4 Å resolutions, respectively. Both X-ray and cryo-EM structures provide clear side-chain densities, which allow for accurate mapping of the AM14 epitope on DS-Cav1. The structures help rationalize the molecular basis for AM14 loss of binding to RSV F monoclonal antibody-resistant mutants and reveal flexibility for the side chain of a key antigenic residue on PreF. This work provides the basis for a comprehensive understanding of RSV F trimer specificity with implications in vaccine design and quality assessment of PreF-based immunogens.
Collapse
Affiliation(s)
| | | | - Sai Tian
- GSK, Vaccine Design and Cellular Immunology, Rockville, MD, USA
| | - Ying Huang
- GSK, Vaccine Design and Cellular Immunology, Rockville, MD, USA
| | | | | | - Enrico Malito
- GSK, Vaccine Design and Cellular Immunology, Rockville, MD, USA
| |
Collapse
|
45
|
Narkhede YB, Gonzalez KJ, Strauch EM. Targeting Viral Surface Proteins through Structure-Based Design. Viruses 2021; 13:v13071320. [PMID: 34372526 PMCID: PMC8310314 DOI: 10.3390/v13071320] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 06/30/2021] [Accepted: 07/01/2021] [Indexed: 12/28/2022] Open
Abstract
The emergence of novel viral infections of zoonotic origin and mutations of existing human pathogenic viruses represent a serious concern for public health. It warrants the establishment of better interventions and protective therapies to combat the virus and prevent its spread. Surface glycoproteins catalyzing the fusion of viral particles and host cells have proven to be an excellent target for antivirals as well as vaccines. This review focuses on recent advances for computational structure-based design of antivirals and vaccines targeting viral fusion machinery to control seasonal and emerging respiratory viruses.
Collapse
Affiliation(s)
- Yogesh B Narkhede
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA;
| | - Karen J Gonzalez
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA;
| | - Eva-Maria Strauch
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA 30602, USA;
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602, USA;
- Correspondence:
| |
Collapse
|
46
|
Stephens LM, Varga SM. Considerations for a Respiratory Syncytial Virus Vaccine Targeting an Elderly Population. Vaccines (Basel) 2021; 9:vaccines9060624. [PMID: 34207770 PMCID: PMC8228432 DOI: 10.3390/vaccines9060624] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/22/2022] Open
Abstract
Respiratory syncytial virus (RSV) is most commonly associated with acute lower respiratory tract infections in infants and children. However, RSV also causes a high disease burden in the elderly that is often under recognized. Adults >65 years of age account for an estimated 80,000 RSV-associated hospitalizations and 14,000 deaths in the United States annually. RSV infection in aged individuals can result in more severe disease symptoms including pneumonia and bronchiolitis. Given the large disease burden caused by RSV in the aged, this population remains an important target for vaccine development. Aging results in lowered immune responsiveness characterized by impairments in both innate and adaptive immunity. This immune senescence poses a challenge when developing a vaccine targeting elderly individuals. An RSV vaccine tailored towards an elderly population will need to maximize the immune response elicited in order to overcome age-related defects in the immune system. In this article, we review the hurdles that must be overcome to successfully develop an RSV vaccine for use in the elderly, and discuss the vaccine candidates currently being tested in this highly susceptible population.
Collapse
Affiliation(s)
- Laura M. Stephens
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA;
| | - Steven M. Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA;
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
- Correspondence:
| |
Collapse
|
47
|
Andreano E, Paciello I, Bardelli M, Tavarini S, Sammicheli C, Frigimelica E, Guidotti S, Torricelli G, Biancucci M, D’Oro U, Chandramouli S, Bottomley MJ, Rappuoli R, Finco O, Buricchi F. The respiratory syncytial virus (RSV) prefusion F-protein functional antibody repertoire in adult healthy donors. EMBO Mol Med 2021; 13:e14035. [PMID: 33998144 PMCID: PMC8185550 DOI: 10.15252/emmm.202114035] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/10/2021] [Accepted: 04/13/2021] [Indexed: 12/27/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of death from lower respiratory tract infection in infants and children, and is responsible for considerable morbidity and mortality in older adults. Vaccines for pregnant women and elderly which are in phase III clinical studies target people with pre-existing natural immunity against RSV. To investigate the background immunity which will be impacted by vaccination, we single cell-sorted human memory B cells and dissected functional and genetic features of neutralizing antibodies (nAbs) induced by natural infection. Most nAbs recognized both the prefusion and postfusion conformations of the RSV F-protein (cross-binders) while a smaller fraction bound exclusively to the prefusion conformation. Cross-binder nAbs used a wide array of gene rearrangements, while preF-binder nAbs derived mostly from the expansion of B-cell clonotypes from the IGHV1 germline. This latter class of nAbs recognizes an epitope located between Site Ø, Site II, and Site V on the F-protein, identifying an important site of pathogen vulnerability.
Collapse
Affiliation(s)
- Emanuele Andreano
- Department of Life SciencesUniversity of SienaSienaItaly
- GSK VaccinesSienaItaly
- Present address:
Monoclonal Antibody Discovery (MAD) LabFondazione Toscana Life SciencesSienaItaly
| | - Ida Paciello
- GSK VaccinesSienaItaly
- Present address:
Monoclonal Antibody Discovery (MAD) LabFondazione Toscana Life SciencesSienaItaly
| | | | | | | | | | | | | | | | | | - Sumana Chandramouli
- GSK VaccinesRockvilleMDUSA
- Present address:
Moderna Therapeutics IncCambridgeMAUSA
| | | | - Rino Rappuoli
- GSK VaccinesSienaItaly
- Faculty of MedicineImperial CollegeLondonUK
- Monoclonal Antibody Discovery (MAD) LabFondazione Toscana Life SciencesSienaItaly
| | | | | |
Collapse
|
48
|
Kalnin KV, Plitnik T, Kishko M, Zhang J, Zhang D, Beauvais A, Anosova NG, Tibbitts T, DiNapoli J, Ulinski G, Piepenhagen P, Cummings SM, Bangari DS, Ryan S, Huang PWD, Huleatt J, Vincent D, Fries K, Karve S, Goldman R, Gopani H, Dias A, Tran K, Zacharia M, Gu X, Boeglin L, Abysalh J, Vargas J, Beaulieu A, Shah M, Jeannotte T, Gillis K, Chivukula S, Swearingen R, Landolfi V, Fu TM, DeRosa F, Casimiro D. Immunogenicity and efficacy of mRNA COVID-19 vaccine MRT5500 in preclinical animal models. NPJ Vaccines 2021; 6:61. [PMID: 33875658 DOI: 10.1101/2020.10.14.337535] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 03/17/2021] [Indexed: 05/28/2023] Open
Abstract
Emergency use authorization of COVID vaccines has brought hope to mitigate pandemic of coronavirus disease 2019 (COVID-19). However, there remains a need for additional effective vaccines to meet the global demand and address the potential new viral variants. mRNA technologies offer an expeditious path alternative to traditional vaccine approaches. Here we describe the efforts to utilize an mRNA platform for rational design and evaluations of mRNA vaccine candidates based on the spike (S) glycoprotein of SARS-CoV-2. Several mRNA constructs of S-protein, including wild type, a pre-fusion stabilized mutant (2P), a furin cleavage-site mutant (GSAS) and a double mutant form (2P/GSAS), as well as others, were tested in animal models for their capacity to elicit neutralizing antibodies (nAbs). The lead 2P/GSAS candidate was further assessed in dose-ranging studies in mice and Cynomolgus macaques, and for efficacy in a Syrian golden hamster model. The selected 2P/GSAS vaccine formulation, designated MRT5500, elicited potent nAbs as measured in neutralization assays in all three preclinical models and more importantly, protected against SARS-CoV-2-induced weight loss and lung pathology in hamsters. In addition, MRT5500 elicited TH1-biased responses in both mouse and non-human primate (NHP), thus alleviating a hypothetical concern of potential vaccine-associated enhanced respiratory diseases known associated with TH2-biased responses. These data position MRT5500 as a viable vaccine candidate for entering clinical development.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Susan Ryan
- Global Discovery Pathology, Sanofi, Framingham, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Safety, tolerability, and immunogenicity of the respiratory syncytial virus prefusion F subunit vaccine DS-Cav1: a phase 1, randomised, open-label, dose-escalation clinical trial. THE LANCET RESPIRATORY MEDICINE 2021; 9:1111-1120. [PMID: 33864736 DOI: 10.1016/s2213-2600(21)00098-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Multiple active vaccination approaches have proven ineffective in reducing the substantial morbidity and mortality caused by respiratory syncytial virus (RSV) in infants and older adults (aged ≥65 years). A vaccine conferring a substantial and sustainable boost in neutralising activity is required to protect against severe RSV disease. To that end, we evaluated the safety and immunogenicity of DS-Cav1, a prefusion F subunit vaccine. METHODS In this randomised, open-label, phase 1 clinical trial, the stabilised prefusion F vaccine DS-Cav1 was evaluated for dose, safety, tolerability, and immunogenicity in healthy adults aged 18-50 years at a single US site. Participants were assigned to receive escalating doses of either 50 μg, 150 μg, or 500 μg DS-Cav1 at weeks 0 and 12, and were randomly allocated in a 1:1 ratio within each dose group to receive the vaccine with or without aluminium hydroxide (AlOH) adjuvant. After 71 participants had been randomised, the protocol was amended to allow some participants to receive a single vaccination at week 0. The primary objectives evaluated the safety and tolerability at every dose within 28 days following each injection. Neutralising activity and RSV F-binding antibodies were evaluated from week 0 to week 44 as secondary and exploratory objectives. Safety was assessed in all participants who received at least one vaccine dose; secondary and exploratory immunogenicity analysis included all participants with available data at a given visit. The trial is registered with ClinicalTrials.gov, NCT03049488, and is complete and no longer recruiting. FINDINGS Between Feb 21, 2017, and Nov 29, 2018, 244 participants were screened for eligibility and 95 were enrolled to receive DS-Cav1 at the 50 μg (n=30, of which n=15 with AlOH), 150 μg (n=35, of which n=15 with AlOH), or 500 μg (n=30, of which n=15 with AlOH) doses. DS-Cav1 was safe and well tolerated and no serious vaccine-associated adverse events deemed related to the vaccine were identified. DS-Cav1 vaccination elicited robust neutralising activity and binding antibodies by 4 weeks after a single vaccination (p<0·0001 for F-binding and neutralising antibodies). In analyses of exploratory endpoints at week 44, pre-F-binding IgG and neutralising activity were significantly increased compared with baseline in all groups. At week 44, RSV A neutralising activity was 3·1 fold above baseline in the 50 μg group, 3·8 fold in the 150 μg group, and 4·5 fold in the 500 μg group (p<0·0001). RSV B neutralising activity was 2·8 fold above baseline in the 50 μg group, 3·4 fold in the 150 μg group, and 3·7 fold in the 500 μg group (p<0·0001). Pre-F-binding IgG remained significantly 3·2 fold above baseline in the 50 μg group, 3·4 fold in the 150 μg group, and 4·0 fold in the 500 μg group (p<0·0001). Pre-F-binding serum IgA remained 4·1 fold above baseline in the 50 μg group, 4·3 fold in the 150 μg group, and 4·8 fold in the 500 μg group (p<0·0001). Although a higher vaccine dose or second immunisation elicited a transient advantage compared with lower doses or a single immunisation, neither significantly impacted long-term neutralisation. There was no long-term effect of dose, number of vaccinations, or adjuvant on neutralising activity. INTERPRETATION In this phase 1 study, DS-Cav1 vaccination was safe and well tolerated. DS-Cav1 vaccination elicited a robust boost in RSV F-specific antibodies and neutralising activity that was sustained above baseline for at least 44 weeks. A single low-dose of pre-F immunisation of antigen-experienced individuals might confer protection that extends throughout an entire RSV season. FUNDING The National Institutes of Allergy and Infectious Diseases.
Collapse
|
50
|
Korsten K, Adriaenssens N, Coenen S, Butler C, Ravanfar B, Rutter H, Allen J, Falsey A, Pirçon JY, Gruselle O, Pavot V, Vernhes C, Balla-Jhagjhoorsingh S, Öner D, Ispas G, Aerssens J, Shinde V, Verheij T, Bont L, Wildenbeest J. Burden of respiratory syncytial virus infection in community-dwelling older adults in Europe (RESCEU): an international prospective cohort study. Eur Respir J 2021; 57:13993003.02688-2020. [PMID: 33060153 DOI: 10.1183/13993003.02688-2020] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 09/20/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Respiratory syncytial virus (RSV) infection in older adults is recognised as an important health issue. We aimed to assess the community burden of RSV in Europe in older adults aged ≥60 years. METHODS This international, prospective, observational cohort study is part of work by the REspiratory Syncytial virus Consortium in EUrope (RESCEU). Participants were recruited through general practitioners' (GPs) offices before two independent RSV seasons. Participants reported weekly about symptoms of acute respiratory tract infection (ARTI) during one RSV season. ARTI patients were tested for RSV during home visits and completed a daily symptom diary. RSV illness included PCR-confirmed ARTI and those showing seroconversion over the season. RSV ARTI was based on PCR alone (ClinicalTrials.gov, NCT03621930). RESULTS We recruited 1040 participants (527 in season 2017-2018 and 513 in season 2018-2019) with a median age of 75 years (range 60-100 years). Of these, 1023 (99%) lived independently at home at baseline. RSV illness incidence was 22 out of 527 (4.2%) and 37 out of 513 (7.2%) in the respective seasons. RSV illness did not affect frailty or cardiopulmonary status during the course of the study. No patients were hospitalised or died from RSV illness. In the 36 patients with PCR confirmed RSV ARTI, symptom duration averaged 19 days, while a doctor's visit took place in 11 out of 36 cases (31%). RSV ARTI could not be differentiated clinically from all other ARTIs based on symptoms. CONCLUSION This European study showed that RSV is prevalent in community-dwelling older adults and rarely causes severe disease. This suggests that watchful waiting, using a continuity of care approach to identify those who do need more intensive care, is often justified when RSV is suspected in family practice.
Collapse
Affiliation(s)
- Koos Korsten
- Dept of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Niels Adriaenssens
- Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.,Dept of Primary and Interdisciplinary Care (ELIZA)-Centre for General Practice, University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Samuel Coenen
- Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.,Dept of Primary and Interdisciplinary Care (ELIZA)-Centre for General Practice, University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium.,Dept of Epidemiology and Social Medicine (ESOC), University of Antwerp, Faculty of Medicine and Health Sciences, Antwerp, Belgium
| | - Christopher Butler
- Nuffield Dept of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Behnaz Ravanfar
- Nuffield Dept of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Heather Rutter
- Nuffield Dept of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Julie Allen
- Nuffield Dept of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Ann Falsey
- Dept of Medicine-Infectious Diseases, University of Rochester Medical Center, Rochester, NY, USA
| | | | | | - Vincent Pavot
- Sanofi Pasteur R&D, Sanofi Pasteur Campus Mérieux, Marcy l'Etoile, France
| | | | | | - Deniz Öner
- Infectious Diseases Translational Biomarkers, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Gabriela Ispas
- Infectious Diseases Translational Biomarkers, Janssen Pharmaceutica NV, Beerse, Belgium
| | - Jeroen Aerssens
- Infectious Diseases Translational Biomarkers, Janssen Pharmaceutica NV, Beerse, Belgium
| | | | - Theo Verheij
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Louis Bont
- Dept of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Joanne Wildenbeest
- Dept of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|