1
|
Guan P, Qi C, Xu G, Sheng C, Sun S, Zhou Z, Jia S. Designing a T cell multi-epitope vaccine against hRSV with reverse vaccinology: An immunoinformatics approach. Colloids Surf B Biointerfaces 2025; 251:114599. [PMID: 40031111 DOI: 10.1016/j.colsurfb.2025.114599] [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: 12/14/2024] [Revised: 02/11/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025]
Abstract
As an infectious viral pathogen, human respiratory syncytial virus (hRSV) can cause severe respiratory infections and is recognized as one of the highest priority pathogens by the World Health Organization (WHO). Although vaccines play an important role in disease prevention and transmission, the wild-type virus is usually prone to immune escape due to the relatively high mutation rate of biological proteins. Therefore, designing a broad-spectrum hRSV vaccine is essential to provide extensive protection against multiple viral variants. Using a consensus sequence approach, we designed a broad-spectrum T-cell epitope vaccine composed of 385 amino acids, consisting of 12 CTLs and 5 HTLs from the fusion protein and glycoprotein. The designed multi-epitope vaccine was expected to have non-allergenicity, high population coverage, strong antigenicity and immunogenicity, appropriate physical and chemical properties, and high solubility. Meanwhile, the structure of the vaccine had a high similarity to that of the natural virus. In addition, through structural biology analysis, the constructed vaccine achieved robust structural compactness and binding stability. Computer-generated immunological simulations indicated that the vaccine could elicit realistic immune responses in humans. The designed vaccine showed good binding affinity and molecular and immune simulation. In conclusion, the broad-spectrum hRSV vaccine could be an excellent candidate for preventing hRSV infection. The employed prediction pipeline was proved to be an efficient method for screening immunogenic epitopes of additional pathogens.
Collapse
Affiliation(s)
- Peibin Guan
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China
| | - Congyan Qi
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei 071000, China
| | - Guojin Xu
- National Institute of Biological Science (NIBS),Beijing 102206, China
| | - Can Sheng
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining 272004, China
| | - Siqi Sun
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College, Beijing 100730, China
| | - Zhicheng Zhou
- Hubei Provincial Key Laboratory of Yeast Function, Angel Yeast Co., Ltd, Yichang 443003, China
| | - Shulei Jia
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
2
|
Oraby AK, Stojic A, Elawar F, Bilawchuk LM, McClelland RD, Erwin K, Granoski MJ, Griffiths CD, Frederick JD, Arutyunova E, Joanne Lemieux M, West FG, Ramilo O, Mejias A, McLellan JS, Marchant DJ. A single amino acid mutation alters multiple neutralization epitopes in the respiratory syncytial virus fusion glycoprotein. NPJ VIRUSES 2025; 3:33. [PMID: 40295799 PMCID: PMC12015481 DOI: 10.1038/s44298-025-00119-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 04/11/2025] [Indexed: 04/30/2025]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of infant hospitalization. All current RSV therapeutics, including antibody prophylaxis and adult vaccination, target the RSV fusion glycoprotein (RSV-F). The seven neutralization sites on RSV-F are highly conserved and infrequently mutate. Here, we show that a single amino acid mutation at position 305 in RSV-F significantly alters antigenic recognition of RSV-F binding sites and reduces the susceptibility of RSV to neutralizing antibodies. In an in vitro evolution assay, we show that RSV-F L305I occurs in a majority of RSV quasi-species. Computational modeling predicted that the L305I mutation altered the epitope landscape of RSV-F, resulting in changes to neutralizing antibody sensitivity and affinity towards the RSV-F glycoprotein. Screening of published RSV-F sequences revealed that position 305 in RSV-F was conserved with a leucine and isoleucine in RSV-A and RSV-B subtypes respectively. Our study suggests that select amino acids in RSV-F may act as 'conformational switches' for RSV to evade host serum antibodies. This work has important implications in understanding RSV evolution and resistance as it suggests that mutational resistance to neutralizing antibodies can occur at sites distal to antigenic epitopes, significantly altering antibody sensitivity to viral infection. These unique antigenic landscape changes should be considered in the context of vaccine and therapeutic development in order to better understand viral mechanisms of evasion and resistance.
Collapse
Affiliation(s)
- Ahmed K Oraby
- Department of Medical Microbiology and Immunology, Edmonton, AB, Canada
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada
- Department of Pharmaceutical Organic Chemistry, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science & Technology, Al-Motamayez District, 6th of October City, Egypt
| | - Aleksandra Stojic
- Department of Medical Microbiology and Immunology, Edmonton, AB, Canada
| | - Farah Elawar
- Department of Medical Microbiology and Immunology, Edmonton, AB, Canada
| | | | | | - Kaci Erwin
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | | | - Cameron D Griffiths
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, 22908, USA
| | | | - Elena Arutyunova
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - M Joanne Lemieux
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Frederick G West
- Department of Chemistry, University of Alberta, Edmonton, AB, T6G 2E1, Canada
| | - Octavio Ramilo
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, 43205, USA
| | - Asuncion Mejias
- Center for Vaccines and Immunity, The Abigail Wexner Research Institute at Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, OH, 43205, USA
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, 78712, USA
| | - David J Marchant
- Department of Medical Microbiology and Immunology, Edmonton, AB, Canada.
| |
Collapse
|
3
|
Sun Z, Lu L, Liu L, Liang R, Zhang Q, Liu Z, An J, Liu Q, Wu Q, Wei S, Zhang L, Peng W. Group IIC self-splicing intron-derived novel circular RNA vaccine elicits superior immune response against RSV. Front Immunol 2025; 16:1574568. [PMID: 40292280 PMCID: PMC12021820 DOI: 10.3389/fimmu.2025.1574568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction The remarkable commercial success of mRNA vaccines against COVID-19 and tumors, along with their potential as therapeutic drugs, has significantly boosted enthusiasm for circular RNAs (circRNA) as a promising next-generation therapeutic platform. The development of novel circRNA cyclization technologies represents a significant leap forward in RNA engineering and therapeutic applications. Recent advancements in group I and IIB self-splicing intron-based ribozymes have enabled precise cyclization of RNA molecules. However, this approach faces significant limitations, including low cyclization efficiency and the requirement for additional additives, which restrict its broader application. Group IIC self-splicing introns represent the shortest known selfsplicing ribozymes and employ a splicing mechanism that is fundamentally distinct from that of group IIB self-splicing introns. However, the potential of group IIC self-splicing introns to carry exogenous sequences for the development of circular RNA-based platforms remains an open question and warrants further investigation. Methods Here, we demonstrate that group IIC self-splicing introns can efficiently circularize and express exogenous proteins of varying lengths, as evidenced by luciferase and GFP reporter systems. Leveraging structural biology-based design, we engineered the RSV pre-F protein and validated the potential of IIC self-splicing introns as a vaccine platform for preventing infectious diseases. Results In mouse models, the novel nucleic acid vaccine developed using IIC self-splicing introns elicited superior immunogenicity and in vivo protective efficacy compared to protein-adjuvant vaccines. Discussion The development of the novel circular RNA vaccine platform holds significant promise for advancing next-generation therapeutics for disease treatment and prevention.
Collapse
Affiliation(s)
- Zeyun Sun
- KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China
| | - Lirong Lu
- Graduate School of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Lijie Liu
- Graduate School of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Ruoxu Liang
- Guangzhou National Laboratory, Guangzhou, China
| | - Qiqi Zhang
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Zhining Liu
- Institute of Pathogenic Biology, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jiahao An
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou, China
| | - Qian Liu
- Guangzhou National Laboratory, Guangzhou, China
| | - Qingxin Wu
- Graduate School of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| | - Shuai Wei
- Guangzhou National Laboratory, Guangzhou, China
| | - Long Zhang
- Guangzhou National Laboratory, Guangzhou, China
| | - Wei Peng
- Graduate School of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
- Guangzhou National Laboratory, Guangzhou, China
| |
Collapse
|
4
|
Chen YC, Chung NH, Lin YC, Yu SL, Liu CC, Chow YH. Enhanced construction of a bivalent adenovirus-based vaccine for preventing childhood pathogens: Human respiratory syncytial virus and norovirus. SLAS Technol 2025; 31:100255. [PMID: 39952326 DOI: 10.1016/j.slast.2025.100255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/10/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
To impede the spread of respiratory syncytial virus (RSV) and norovirus (NoV) in children, we developed novel recombinant adenoviruses expressing RSV fusion (F) and NoV VP1 proteins driven by different promoter elements (EF1α, SV40, and CMV), resulting in Ad-F/E/NoV, Ad-F/S/NoV, and Ad-F/C/NoV, respectively. The expressed F can be recognized by postfusion-specific antibody targeting to site II and prefusion-specific antibodies targeting to sites V and Ø in the Ad-F-infected cells. However, NoV VP1 is only expressed in Ad-F/E/NoV and Ad-F/C/NoV-infected cells. Intraperitoneal two-dose with monovalent Ad-F and all three bicistronic Ads individually in rodents induced anti-F neutralizing antibodies similarly. Ad-F/C/NoV and Ad-F/E/NoV but not Ad-F/S/NoV significantly induced NoV VP1-specific IgG. The serum from Ad-F/C/NoV-immunized subjects elicited superior anti-NoV activity, as evidenced by reduced binding of NoV VLPs to histo-blood group antigens. Ad-F/C/NoV and Ad-F/E/NoV-immunized mice exhibited elevated cellular immune-mediated splenic IFN-γ and IL-4 secretions. In the RSV challenge study, pulmonary virions were significantly decreased during the vaccination with each of the three bicistronic Ads, confirming their efficacy in preventing RSV infection. Finally, the mucosal (intranasal) immunization in mice with Ad-F/C/NoV induced superior anti-RSV and NoV IgA in both serum and vaginal washes specific to RSV and NoV were highly induced. These findings highlight the optimization of an Ad vaccine targeting two pathogens prevalent in childhood. Additionally, the results underscore the utility of mucosal delivery of Ad vaccines as a valuable strategy for public vaccination efforts.
Collapse
Affiliation(s)
- Ying-Chin Chen
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Nai-Hsiang Chung
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Yu-Ching Lin
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Shu-Ling Yu
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Chia-Chyi Liu
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Yen-Hung Chow
- Institute of Infectious Disease and Vaccinology, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
| |
Collapse
|
5
|
Lao G, Feng J, Wu L, Su W, Chen L, Yang L, Zhang S, Xu Y, Peng T. Development of a genetically modified full-length human respiratory syncytial virus preF protein vaccine. Vaccine 2025; 49:126799. [PMID: 39874917 DOI: 10.1016/j.vaccine.2025.126799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/07/2025] [Accepted: 01/22/2025] [Indexed: 01/30/2025]
Abstract
Human Respiratory Syncytial Virus (hRSV) is a major cause of acute lower respiratory tract infections (ALRTI) in infants, the elderly, and immunocompromised individuals. The recent approval of recombinant protein-based hRSV vaccines represents significant progress in combating hRSV. However, these vaccines utilized optimized preF ectodomain attached with an exogenous trimeric motif, which may induce immunological complications. Our research addresses these concerns by employing modified "full-length" preF proteins, preF-TMCT, designed to mimic the natural F protein structure and avoid potential immunological complications. We characterized a group of preF constructs and identified two candidates that exhibited desirable expression levels, high antigenicity and good stability. Immunization of Balb/c mice confirmed the robust immunogenicity and effective in induction of cross-reactive neutralizing antibodies of these antigens, particularly the lead-construct BR40. This investigation aims to contribute new insights to hRSV vaccine development. The near-native structure of the "full-length" preF-TMCT antigen also makes it valuable for producing therapeutic monoclonal antibodies (mAbs) and other biopharmaceuticals against hRSV infection.
Collapse
Affiliation(s)
- Geqi Lao
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jin Feng
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong South China Vaccine Co., Ltd., Guangzhou 510530, China
| | - Liping Wu
- Guangdong South China Vaccine Co., Ltd., Guangzhou 510530, China
| | - Wenhan Su
- Guangdong South China Vaccine Co., Ltd., Guangzhou 510530, China
| | - Liyun Chen
- Guangdong South China Vaccine Co., Ltd., Guangzhou 510530, China
| | - Lejun Yang
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Songchen Zhang
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yuhua Xu
- Guangdong South China Vaccine Co., Ltd., Guangzhou 510530, China.
| | - Tao Peng
- State Key Laboratory of Respiratory Diseases, Sino-French Hoffmann Institute, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Guangdong South China Vaccine Co., Ltd., Guangzhou 510530, China.
| |
Collapse
|
6
|
Cadena-Cruz C, De-Avila-Arias M, Costello HM, Hurtado-Gomez L, Martínez-De-La-Rosa W, Macchia-Ceballos G, Rosales-Rada W, Valencia-Villa G, Villalba-Amarís P, Kararoudi MN, Peeples ME, San-Juan-Vergara H. Respiratory syncytial virus fuses with plasma membrane to infect primary cultures of bronchial epithelial cells. Front Microbiol 2025; 16:1498955. [PMID: 40099186 PMCID: PMC11911548 DOI: 10.3389/fmicb.2025.1498955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025] Open
Abstract
Background Respiratory syncytial virus (RSV) is a common cause of bronchiolitis in children under the age of five. RSV infection proceeds by fusion of the viral envelope with the target cell membrane, but it is unclear whether fusion occurs with plasma or endosomal membranes. Methods Entry and/or infection was studied in undifferentiated primary cultures of human bronchial epithelial cells. Synchronization of viral entry or infection was achieved by attaching the virus to the plasma membrane at temperatures of 4°C or 22°C. Cells in which entry events had occurred were identified by the enzymatic action of beta-lactamase M (BlaM) fused to the RSV P protein (BlaM-P) carried by rgRSV virions. BlaM cleaves the beta-lactam ring of CCF2 loaded into the cells, disrupting FRET and allowing blue light to be emitted. Green fluorescent protein (GFP) expression, encoded by the rgRSV genome, was used to identify infected cells. Results We found that adsorption of RSV at 4°C favors entry via endocytosis, whereas binding of the virus to the membrane at 22°C favors RSV entry via the plasma membrane. The induction of endocytosis by synchronization at 4°C is, therefore, an artifact. In addition, we found that all drugs that interfered with RSV infection reduced cell membrane deformations such as filopodia and lamellipodia, suggesting a mechanism by which they may interfere with RSV fusion with the cell membrane. Discussion In conclusion, RSV enters the cell by direct fusion of its envelope with the plasma membrane.
Collapse
Affiliation(s)
- Christian Cadena-Cruz
- Departamento de Medicina, Universidad del Norte, Barranquilla, Colombia
- Programa de Bacteriología, Universidad Libre Seccional, Barranquilla, Colombia
| | | | - Heather M Costello
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | | | | | | | - Wendy Rosales-Rada
- Departamento de Medicina, Universidad del Norte, Barranquilla, Colombia
- Grupo de Investigación Avanzada en Biomedicina, Programa de Microbiología, Universidad Libre de Colombia, Barranquilla, Atlántico, Colombia
| | | | | | - Meisam Naeimi Kararoudi
- Center for Childhood Cancer and Blood Diseases, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Mark E Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, United States
- Infectious Disease Institute, The Ohio State University, Columbus, OH, United States
| | | |
Collapse
|
7
|
Zhao F, Zhang Y, Zhang Z, Chen Z, Wang X, Wang S, Li R, Li Y, Zhang Z, Zheng W, Wang Y, Zhang Z, Wu S, Yang Y, Zhang J, Zai X, Xu J, Chen W. Epitope-focused vaccine immunogens design using tailored horseshoe-shaped scaffold. J Nanobiotechnology 2025; 23:119. [PMID: 39966941 PMCID: PMC11834273 DOI: 10.1186/s12951-025-03200-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
The continuous emergence of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants highlights the need to update coronavirus 2019 disease (COVID-19) vaccine components. Epitope-based vaccine designs targeting conserved and immunorecessive regions of SARS-CoV-2 are critically needed. Here, we report an engineered epitope-focused immunogen design based on a novel horseshoe-shaped natural protein scaffold, named ribonuclease inhibitor 1 (RNH1), that can multiply display of conserved neutralizing epitopes from SARS-CoV-2 S2 stem helix. The designed immunogen RNH1-S1139 demonstrates high binding affinity to S2-specific neutralizing antibodies and elicits robust epitope-targeted antibody responses either through homologous or heterologous vaccination regimens. RNH1-S1139 immune serum has been proven to have similar binding ability against SARS-CoV, SARS-CoV-2 and its variants, providing broad-spectrum protection as a membrane fusion inhibitor. Further studies showed that RNH1 has the potential to serve as a versatile scaffold that displays other helical epitopes from various antigens, including respiratory syncytial virus (RSV) F glycoprotein. Our proposed immunogen engineering strategy via tailored horseshoe-shape nano-scaffold supports the continued development of epitope-focused vaccines as part of a next-generation vaccine design.
Collapse
Affiliation(s)
- Fangxin Zhao
- School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yue Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Zhiling Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Zhengshan Chen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiaolin Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Shaoyan Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Ruihua Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yaohui Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Zhang Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Wanru Zheng
- School of Medicine, Zhejiang University, Hangzhou, 310058, China
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yudong Wang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Zhe Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Shipo Wu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yilong Yang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Jun Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiaodong Zai
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Junjie Xu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China.
| | - Wei Chen
- School of Medicine, Zhejiang University, Hangzhou, 310058, China.
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China.
- Lead Contact, Beijing, China.
| |
Collapse
|
8
|
Ridelfi M, Pierleoni G, Zucconi Galli Fonseca V, Batani G, Rappuoli R, Sala C. State of the Art and Emerging Technologies in Vaccine Design for Respiratory Pathogens. Semin Respir Crit Care Med 2025. [PMID: 39870103 DOI: 10.1055/a-2500-1878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
In this review, we present the efforts made so far in developing effective solutions to prevent infections caused by seven major respiratory pathogens: influenza virus, respiratory syncytial virus (RSV), the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), Bordetella pertussis, Streptococcus pneumoniae (pneumococcus), Mycobacterium tuberculosis, and Pseudomonas aeruginosa. Advancements driven by the recent coronavirus disease 2019 (COVID-19) crisis have largely focused on viruses, but effective prophylactic solutions for bacterial pathogens are also needed, especially in light of the antimicrobial resistance (AMR) phenomenon. Here, we discuss various innovative key technologies that can help address this critical need, such as (a) the development of Lung-on-Chip ex vivo models to gain a better understanding of the pathogenesis process and the host-microbe interactions; (b) a more thorough investigation of the mechanisms behind mucosal immunity as the first line of defense against pathogens; (c) the identification of correlates of protection (CoPs) which, in conjunction with the Reverse Vaccinology 2.0 approach, can push a more rational and targeted design of vaccines. By focusing on these critical areas, we expect substantial progress in the development of new vaccines against respiratory bacterial pathogens, thereby enhancing global health protection in the framework of the increasingly concerning AMR emergence.
Collapse
Affiliation(s)
- Matteo Ridelfi
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | - Giulio Pierleoni
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
| | | | - Giampiero Batani
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| | | | - Claudia Sala
- Monoclonal Antibody Discovery (MAD) Lab, Fondazione Toscana Life Sciences, Siena, Italy
| |
Collapse
|
9
|
Babawale PI, Martínez-Espinoza I, Mitchell AM, Guerrero-Plata A. Preventing RSV Infection in Children: Current Passive Immunizations and Vaccine Development. Pathogens 2025; 14:104. [PMID: 40005481 PMCID: PMC11858734 DOI: 10.3390/pathogens14020104] [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: 12/18/2024] [Revised: 01/12/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
Human respiratory syncytial virus (RSV) is a leading cause of acute respiratory tract infection and lower respiratory tract infection, associated with high morbidity and mortality in young children, the elderly, and immunocompromised individuals. Initial attempts to develop an RSV vaccine in the 1960s were faced with a setback due to the enhanced RSV disease developed by vaccinated children. More recent advancements have led to the generation of RSV vaccines for older adults and pregnant women. However, there are still no commercially available RSV vaccines for infants. This work summarizes the current passive immunizations and the ongoing efforts to develop an RSV vaccine for infants.
Collapse
Affiliation(s)
| | | | | | - Antonieta Guerrero-Plata
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA; (P.I.B.); (I.M.-E.); (A.M.M.)
| |
Collapse
|
10
|
Li J, Long H, Chen S, Zhang Z, Li S, Liu Q, Liu J, Cai J, Luo L, Peng Y. An mRNA-Based Respiratory Syncytial Virus Vaccine Elicits Strong Neutralizing Antibody Responses and Protects Rodents Without Vaccine-Associated Enhanced Respiratory Disease. Vaccines (Basel) 2025; 13:52. [PMID: 39852831 PMCID: PMC11768429 DOI: 10.3390/vaccines13010052] [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: 11/23/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) causes the most common type of severe lower respiratory tract infection worldwide, and the fusion (F) protein is a target for neutralizing antibodies and vaccine development. This study aimed to investigate the immunogenicity and efficacy of an mRNA-based RSV vaccine with an F protein sequence. METHODS We designed an mRNA construct encoding a modified RSV F protein, which was further developed into an LNP-encapsulated mRNA vaccine (LVRNA007). LVRNA007 was administered to mice and cotton rats, followed by immunogenicity analysis and viral challenge studies. Protection of rodents from the viral infection was evaluated based on the presence of the virus in the lung and pathological examination of respiratory tissues. RESULTS LVRNA007 induced robust humoral and cellular immune responses in both mice and cotton rats, with neutralization antibody levels in the immunized animals maintained at high levels for over one year. Vaccination of LVRNA007 also protected the rodents from RSV challenge, judged by the much decreased virus titer and the pathological score in the lung tissue. In addition, no vaccine-enhanced disease (VED) phenomenon was observed with LVRNA007 vaccination. CONCLUSIONS Based on the preclinical immunogenicity and efficacy data, LVRNA007 could be a potential promising vaccine for prophylaxis of RSV infection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yucai Peng
- Liverna Therapeutics Inc., Zhuhai 519000, China; (J.L.); (H.L.); (S.C.); (Z.Z.); (S.L.); (Q.L.); (J.L.); (J.C.); (L.L.)
| |
Collapse
|
11
|
Mukhopadhyay S, Manolaridis I, Warren C, Tang A, O’Donnell G, Luo B, Staupe RP, Vora KA, Chen Z. Anti-Idiotypic Antibody as a Booster Vaccine Against Respiratory Syncytial Virus. Vaccines (Basel) 2025; 13:35. [PMID: 39852814 PMCID: PMC11768756 DOI: 10.3390/vaccines13010035] [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: 11/20/2024] [Revised: 12/10/2024] [Accepted: 12/28/2024] [Indexed: 01/26/2025] Open
Abstract
Background/Objectives: The respiratory syncytial virus (RSV) is a major cause of lower respiratory tract infections in children and adults. With nearly everyone infected by the age of five, there is an opportunity to develop booster vaccines that enhance B-cell immunity, promoting potent and broadly neutralizing antibodies. One potential approach involves using anti-idiotypic antibodies (anti-IDs) to mimic specific antigenic sites and enhance preexisting immunity in an epitope-specific manner. RB1, a monoclonal antibody (mAb) that binds to site IV of the RSV fusion (RSV F) protein, is a potent and broadly neutralizing against RSV A and B viruses. It is the precursor for MK1654 (clesrovimab), which successfully completed a Phase III clinical trial. Methods: In this study, we isolated two anti-IDs, 1A6 and 1D4, targeting RB1 CDR regions, demonstrating that 1A6 competes fully with RSV F in binding to RB1. Results: We resolved the RB1-1A6 and RB1-1D4 Fab-Fab complex structures and proved that 1A6 mimics the RSV F site IV better than 1D4. In an immunogenicity study, mice primed with RSV F and boosted with 1A6 Fab showed a site IV-specific antibody response with a concurrent increase in RSV virus neutralization. Conclusions: These results suggest that anti-IDs could be potentially used as booster vaccines for specific epitopes.
Collapse
Affiliation(s)
- Shreya Mukhopadhyay
- Infectious Diseases and Vaccine Research, Merck & Co., Inc., Rahway, NJ 07065, USA; (S.M.); (C.W.); (A.T.)
| | | | - Christopher Warren
- Infectious Diseases and Vaccine Research, Merck & Co., Inc., Rahway, NJ 07065, USA; (S.M.); (C.W.); (A.T.)
| | - Aimin Tang
- Infectious Diseases and Vaccine Research, Merck & Co., Inc., Rahway, NJ 07065, USA; (S.M.); (C.W.); (A.T.)
| | - Gregory O’Donnell
- Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ 07065, USA; (G.O.); (B.L.)
| | - Bin Luo
- Quantitative Biosciences, Merck & Co., Inc., Rahway, NJ 07065, USA; (G.O.); (B.L.)
| | - Ryan P. Staupe
- Infectious Diseases and Vaccine Research, Merck & Co., Inc., Rahway, NJ 07065, USA; (S.M.); (C.W.); (A.T.)
| | - Kalpit A. Vora
- Infectious Diseases and Vaccine Research, Merck & Co., Inc., Rahway, NJ 07065, USA; (S.M.); (C.W.); (A.T.)
| | - Zhifeng Chen
- Infectious Diseases and Vaccine Research, Merck & Co., Inc., Rahway, NJ 07065, USA; (S.M.); (C.W.); (A.T.)
| |
Collapse
|
12
|
Pisuttinusart N, Rattanapisit K, Srisaowakarn C, Thitithanyanont A, Strasser R, Shanmugaraj B, Phoolcharoen W. Neutralizing activity of anti-respiratory syncytial virus monoclonal antibody produced in Nicotiana benthamiana. Hum Vaccin Immunother 2024; 20:2327142. [PMID: 38508690 PMCID: PMC10956629 DOI: 10.1080/21645515.2024.2327142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/03/2024] [Indexed: 03/22/2024] Open
Abstract
Respiratory syncytial virus (RSV) is a highly contagious virus that affects the lungs and respiratory passages of many vulnerable people. It is a leading cause of lower respiratory tract infections and clinical complications, particularly among infants and elderly. It can develop into serious complications such as pneumonia and bronchiolitis. The development of RSV vaccine or immunoprophylaxis remains highly active and a global health priority. Currently, GSK's Arexvy™ vaccine is approved for the prevention of lower respiratory tract disease in older adults (>60 years). Palivizumab and currently nirsevimab are the approved monoclonal antibodies (mAbs) for RSV prevention in high-risk patients. Many studies are ongoing to develop additional therapeutic antibodies for preventing RSV infections among newborns and other susceptible groups. Recently, additional antibodies have been discovered and shown greater potential for development as therapeutic alternatives to palivizumab and nirsevimab. Plant expression platforms have proven successful in producing recombinant proteins, including antibodies, offering a potential cost-effective alternative to mammalian expression platforms. Hence in this study, an attempt was made to use a plant expression platform to produce two anti-RSV fusion (F) mAbs 5C4 and CR9501. The heavy-chain and light-chain sequences of both these antibodies were transiently expressed in Nicotiana benthamiana plants using a geminiviral vector and then purified using single-step protein A affinity column chromatography. Both these plant-produced mAbs showed specific binding to the RSV fusion protein and demonstrate effective viral neutralization activity in vitro. These preliminary findings suggest that plant-produced anti-RSV mAbs are able to neutralize RSV in vitro.
Collapse
Affiliation(s)
- Nuttapat Pisuttinusart
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Plant-Produced Pharmaceuticals, Chulalongkorn University, Bangkok, Thailand
| | - Kaewta Rattanapisit
- Department of Research and Development, Baiya Phytopharm Co., Ltd., Bangkok, Thailand
| | - Chanya Srisaowakarn
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Richard Strasser
- Department of Applied Genetics and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Balamurugan Shanmugaraj
- Department of Research and Development, Baiya Phytopharm Co., Ltd., Bangkok, Thailand
- Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - Waranyoo Phoolcharoen
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Plant-Produced Pharmaceuticals, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
13
|
Terstappen J, Hak SF, Bhan A, Bogaert D, Bont LJ, Buchholz UJ, Clark AD, Cohen C, Dagan R, Feikin DR, Graham BS, Gupta A, Haldar P, Jalang'o R, Karron RA, Kragten L, Li Y, Löwensteyn YN, Munywoki PK, Njogu R, Osterhaus A, Pollard AJ, Nazario LR, Sande C, Satav AR, Srikantiah P, Stein RT, Thacker N, Thomas R, Bayona MT, Mazur NI. The respiratory syncytial virus vaccine and monoclonal antibody landscape: the road to global access. THE LANCET. INFECTIOUS DISEASES 2024; 24:e747-e761. [PMID: 39326422 DOI: 10.1016/s1473-3099(24)00455-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/18/2024] [Accepted: 07/12/2024] [Indexed: 09/28/2024]
Abstract
Respiratory syncytial virus (RSV) is the second most common pathogen causing infant mortality. Additionally, RSV is a major cause of morbidity and mortality in older adults (age ≥60 years) similar to influenza. A protein-based maternal vaccine and monoclonal antibody (mAb) are now market-approved to protect infants, while an mRNA and two protein-based vaccines are approved for older adults. First-year experience protecting infants with nirsevimab in high-income countries shows a major public health benefit. It is expected that the RSV vaccine landscape will continue to develop in the coming years to protect all people globally. The vaccine and mAb landscape remain active with 30 candidates in clinical development using four approaches: protein-based, live-attenuated and chimeric vector, mRNA, and mAbs. Candidates in late-phase trials aim to protect young infants using mAbs, older infants and toddlers with live-attenuated vaccines, and children and adults using protein-based and mRNA vaccines. This Review provides an overview of RSV vaccines highlighting different target populations, antigens, and trial results. As RSV vaccines have not yet reached low-income and middle-income countries, we outline urgent next steps to minimise the vaccine delay.
Collapse
Affiliation(s)
- Jonne Terstappen
- Department of Paediatric Infectious Disease & Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Sarah F Hak
- Department of Paediatric Infectious Disease & Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Anant Bhan
- Yenepoya Medical College & Centre for Ethics, Yenepoya University, Mangalore, India
| | - Debby Bogaert
- Paediatric Medicine, University of Edinburgh, Edinburgh, UK
| | - Louis J Bont
- Department of Paediatric Infectious Disease & Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands; ReSViNET Foundation, Zeist, Netherlands
| | - Ursula J Buchholz
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrew D Clark
- Department of Health Services Research and Policy, London School of Hygiene and Tropical Medicine, London, UK
| | - Cheryl Cohen
- Center for Respiratory Diseases and Meningitis, University of the Witwatersrand and National Institute for Communicable Diseases, Johannesburg, South Africa
| | - Ron Dagan
- The Shraga Segal Department of Microbiology, Immunology, and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheba, Israel
| | - Daniel R Feikin
- Department of Immunization, Vaccines, and Biologicals, WHO, Geneva, Switzerland
| | - Barney S Graham
- Department of Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Anuradha Gupta
- Global Immunization, Sabin Vaccine Institute, Washington, DC, USA
| | - Pradeep Haldar
- Government of India, Ministry of Health and Family Welfare, Delhi, India
| | - Rose Jalang'o
- National Vaccines and Immunization Program, Ministry of Health, Nairobi, Kenya
| | - Ruth A Karron
- Boomberg School of Public Health Department of International Health, Johns Hopkins Bloomberg Baltimore, MD, USA
| | | | - You Li
- Centre for Global Health, University of Edinburgh, Edinburgh, UK; School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yvette N Löwensteyn
- Department of Paediatric Infectious Disease & Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | | | - Rosemary Njogu
- Department of International Health, Jhpiego, Nairobi, Kenya
| | - Ab Osterhaus
- Center of Infection Medicine and Zoonosis Research, University of Veterinary Medicine, Hannover, Germany
| | - Andrew J Pollard
- Oxford Vaccine Group, Department of Pediatrics, University of Oxford, Oxford, UK
| | | | - Charles Sande
- Centre for Tropical Medicine and Global Health, University of Oxford, Oxford, UK; KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Ashish R Satav
- MAHAN Trust Mahatma Gandhi Tribal Hospital, Melghat, India
| | - Padmini Srikantiah
- Global Health Division, Bill & Melinda Gates Foundation, Seattle, WA, USA
| | - Renato T Stein
- Pneumologia Pediátrica, Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Naveen Thacker
- International Pediatric Association, Webster Groves, MI, USA; Child Health Foundation, Mumbai, India
| | | | | | - Natalie I Mazur
- Department of Paediatric Infectious Disease & Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands.
| |
Collapse
|
14
|
Bakkers MJG, Cox F, Koornneef A, Yu X, van Overveld D, Le L, van den Hoogen W, Vaneman J, Thoma A, Voorzaat R, Tettero L, Juraszek J, van der Fits L, Zahn R, Langedijk JPM. A foldon-free prefusion F trimer vaccine for respiratory syncytial virus to reduce off-target immune responses. Nat Microbiol 2024; 9:3254-3267. [PMID: 39567664 PMCID: PMC11602707 DOI: 10.1038/s41564-024-01860-1] [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: 04/10/2024] [Accepted: 10/14/2024] [Indexed: 11/22/2024]
Abstract
Respiratory syncytial virus (RSV) is a major cause of severe respiratory disease in infants and older people. Current RSV subunit vaccines are based on a fusion protein that is stabilized in the prefusion conformation and linked to a heterologous foldon trimerization domain to obtain a prefusion F (preF) trimer. Here we show that current RSV vaccines induce undesirable anti-foldon antibodies in non-human primates, mice and humans. To overcome this, we designed a foldon-free RSV preF trimer by elucidating the structural basis of trimerization-induced preF destabilization through molecular dynamics simulations and by introducing amino acid substitutions that negate hotspots of charge repulsion. The highly stable prefusion conformation was validated using antigenic and cryo-electron microscopy analysis. The preF is immunogenic and protective in naive mouse models and boosts neutralizing antibody titres in RSV-pre-exposed mice and non-human primates, while achieving similar titres to approved RSV vaccines in mice. This stable preF design is a promising option as a foldon-independent candidate for a next-generation RSV vaccine immunogen.
Collapse
Affiliation(s)
- Mark J G Bakkers
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
- ForgeBio BV, Amsterdam, The Netherlands
| | - Freek Cox
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | | | - Xiaodi Yu
- Structural and Protein Science, Janssen Research and Development, Spring House, PA, USA
| | | | - Lam Le
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | | | - Joost Vaneman
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | - Anne Thoma
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | | | | | - Jarek Juraszek
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | | | - Roland Zahn
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | - Johannes P M Langedijk
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands.
- ForgeBio BV, Amsterdam, The Netherlands.
| |
Collapse
|
15
|
Girma A. Biology of human respiratory syncytial virus: Current perspectives in immune response and mechanisms against the virus. Virus Res 2024; 350:199483. [PMID: 39396572 PMCID: PMC11513633 DOI: 10.1016/j.virusres.2024.199483] [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: 08/08/2024] [Revised: 09/30/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
Human respiratory syncytial virus (hRSV) remains a leading cause of morbidity and mortality in infants, young children, and older adults. hRSV infection's limited treatment and vaccine options significantly increase bronchiolitis' morbidity rates. The severity and outcome of viral infection hinge on the innate immune response. Developing vaccines and identifying therapeutic interventions suitable for young children, older adults, and pregnant women relies on comprehending the molecular mechanisms of viral PAMP recognition, genetic factors of the inflammatory response, and antiviral defense. This review covers fundamental elements of hRSV biology, diagnosis, pathogenesis, and the immune response, highlighting prospective options for vaccine development.
Collapse
Affiliation(s)
- Abayeneh Girma
- Department of Biology, College of Natural and Computational Sciences, Mekdela Amba University, P.O. Box 32, Tulu Awuliya, Ethiopia.
| |
Collapse
|
16
|
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
|
17
|
Scharffenberger SC, Wan YH, Homad LJ, Kher G, Haynes AM, Poudel B, Sinha IR, Aldridge N, Pai A, Bibby M, Chhan CB, Davis AR, Moodie Z, Palacio MB, Escolano A, McElrath MJ, Boonyaratanakornkit J, Pancera M, McGuire AT. Targeting RSV-neutralizing B cell receptors with anti-idiotypic antibodies. Cell Rep 2024; 43:114811. [PMID: 39383036 PMCID: PMC11496930 DOI: 10.1016/j.celrep.2024.114811] [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: 04/05/2024] [Revised: 07/23/2024] [Accepted: 09/16/2024] [Indexed: 10/11/2024] Open
Abstract
Respiratory syncytial virus (RSV) causes lower respiratory tract infections with significant morbidity and mortality at the extremes of age. Vaccines based on the viral fusion protein are approved for adults over 60, but infant protection relies on passive immunity via antibody transfer or maternal vaccination. An infant vaccine that rapidly elicits protective antibodies would fulfill a critical unmet need. Antibodies arising from the VH3-21/VL1-40 gene pairing can neutralize RSV without the need for affinity maturation, making them attractive to target through vaccination. Here, we develop an anti-idiotypic monoclonal antibody (ai-mAb) immunogen that is specific for unmutated VH3-21/VL1-40 B cell receptors (BCRs). The ai-mAb efficiently engages B cells with bona fide target BCRs and does not activate off-target non-neutralizing B cells, unlike recombinant pre-fusion (preF) protein used in current RSV vaccines. These results establish proof of concept for using an ai-mAb-derived vaccine to target B cells hardwired to produce RSV-neutralizing antibodies.
Collapse
MESH Headings
- Antibodies, Neutralizing/immunology
- Animals
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Humans
- Antibodies, Anti-Idiotypic/immunology
- Antibodies, Anti-Idiotypic/pharmacology
- Mice
- B-Lymphocytes/immunology
- Respiratory Syncytial Virus Infections/immunology
- Respiratory Syncytial Viruses/immunology
- Antibodies, Monoclonal/immunology
- Antibodies, Viral/immunology
- Female
- Respiratory Syncytial Virus, Human/immunology
- Mice, Inbred BALB C
Collapse
Affiliation(s)
- Samuel C Scharffenberger
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA
| | - Yu-Hsin Wan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Leah J Homad
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Gargi Kher
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Austin M Haynes
- Department of Global Health, University of Washington, Seattle, WA 98195, USA
| | - Bibhav Poudel
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Irika R Sinha
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Nicholas Aldridge
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Ayana Pai
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Madeleine Bibby
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Crystal B Chhan
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Seattle, WA 98195, USA
| | - Amelia R Davis
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Zoe Moodie
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Maria Belen Palacio
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104, USA
| | - Amelia Escolano
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA 19104, USA
| | - M Juliana McElrath
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Jim Boonyaratanakornkit
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Department of Global Health, University of Washington, Seattle, WA 98195, USA; Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Marie Pancera
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Andrew T McGuire
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA; Department of Global Health, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
18
|
Grego E, Kelly SM, McGill JL, Wannemuehler M, Narasimhan B. Bovine Respiratory Syncytial Virus Nanovaccine Induces Long-Lasting Humoral Immunity in Mice. ACS Pharmacol Transl Sci 2024; 7:3205-3215. [PMID: 39421663 PMCID: PMC11480889 DOI: 10.1021/acsptsci.4c00375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024]
Abstract
With limited therapies and vaccines available, human respiratory syncytial virus (HRSV) has a significant negative health impact on all age groups but particularly on infants, young children, and older adults. Bovine respiratory syncytial virus (BRSV) is pathogenically and antigenically similar to HRSV. Building upon previous studies using a BRSV nanovaccine coencapsulating multiple proteins, this work demonstrates the development and comparative evaluation of a coencapsulated nanovaccine to a cocktail nanovaccine formulation composed of polyanhydride nanoparticles encapsulating BRSV postfusion (F) glycoprotein and CpG ODN 1668 coadjuvant delivered simultaneously with nanoparticles encapsulating BRSV attachment glycoprotein (G) and CpG ODN 1668. These nanovaccine formulations were administered to C57BL/6 mice by one of two prime-boost regimens (i.e., intranasal/intranasal or intranasal/subcutaneous) followed by assessment of humoral immunity. The cocktail nanovaccine induced sustained anti-F and anti-G serum IgG antibody responses for 12 weeks postprimary immunization. Using polyanhydride particles to deliver G protein in a prime-boost regime also significantly induced serum anti-G antibodies compared to protein and coadjuvant alone. Serum IgG induced by the nanovaccine demonstrated virus-neutralizing capability from 42 to 119 days postprimary immunization. Further, anti-F IgG antibodies were detected in the bronchoalveolar lavage fluid of vaccinated animals. Finally, the nanovaccine induced long-lived anti-F antibody secreting plasma cells that were detectable in the bone marrow 205 days postprimary immunization. Overall, the BRSV nanovaccine(s) successfully induced long-lived humoral immune responses capable of virus neutralization, making this a promising vaccine candidate for further evaluation in other relevant animal models.
Collapse
Affiliation(s)
- Elizabeth Grego
- Chemical
& Biological Engineering, Iowa State
University, Ames, Iowa 50011, United States
- Nanovaccine
Institute, Ames, Iowa 50011, United States
| | - Sean M. Kelly
- Chemical
& Biological Engineering, Iowa State
University, Ames, Iowa 50011, United States
- Nanovaccine
Institute, Ames, Iowa 50011, United States
| | - Jodi L. McGill
- Nanovaccine
Institute, Ames, Iowa 50011, United States
- Veterinary
Microbiology & Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States
| | - Michael Wannemuehler
- Nanovaccine
Institute, Ames, Iowa 50011, United States
- Veterinary
Microbiology & Preventive Medicine, Iowa State University, Ames, Iowa 50011, United States
| | - Balaji Narasimhan
- Chemical
& Biological Engineering, Iowa State
University, Ames, Iowa 50011, United States
- Nanovaccine
Institute, Ames, Iowa 50011, United States
| |
Collapse
|
19
|
Liang Y, Shao S, Li XY, Zhao ZX, Liu N, Liu ZM, Shen FJ, Zhang H, Hou JW, Zhang XF, Jin YQ, Du LF, Li X, Zhang J, Su JG, Li QM. Mutating a flexible region of the RSV F protein can stabilize the prefusion conformation. Science 2024; 385:1484-1491. [PMID: 39325881 DOI: 10.1126/science.adp2362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024]
Abstract
The respiratory syncytial virus (RSV) fusion (F) glycoprotein is highly immunogenic in its prefusion (pre-F) conformation. However, the protein is unstable, and its conformation must be stabilized for it to function effectively as an immunogen in vaccines. We present a mutagenesis strategy to arrest the RSV F protein in its pre-F state by blocking localized changes in protein structure that accompany large-scale conformational rearrangements. We generated a series of mutants and screened them in vitro to assess their potential for forming a stable pre-F. In animals, the immunogenicity of a representative mutant F protein, with a conformation confirmed by cryo-electron microscopy, elicited levels of neutralizing antibodies and protection against RSV-induced lung damage that were comparable to those of DS-Cav1, a pre-F used in a licensed vaccine.
Collapse
Affiliation(s)
- Yu Liang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Shuai Shao
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Xin Yu Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Zi Xin Zhao
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Ning Liu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Zhao Ming Liu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Fu Jie Shen
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Hao Zhang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Jun Wei Hou
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Xue Feng Zhang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Yu Qin Jin
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Li Fang Du
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Xin Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Jing Zhang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Ji Guo Su
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| | - Qi Ming Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing 101111, China
- National Engineering Center for New Vaccine Research, Beijing 101111, China
| |
Collapse
|
20
|
Bakkers MJG, Ritschel T, Tiemessen M, Dijkman J, Zuffianò AA, Yu X, van Overveld D, Le L, Voorzaat R, van Haaren MM, de Man M, Tamara S, van der Fits L, Zahn R, Juraszek J, Langedijk JPM. Efficacious human metapneumovirus vaccine based on AI-guided engineering of a closed prefusion trimer. Nat Commun 2024; 15:6270. [PMID: 39054318 PMCID: PMC11272930 DOI: 10.1038/s41467-024-50659-5] [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: 02/20/2024] [Accepted: 07/12/2024] [Indexed: 07/27/2024] Open
Abstract
The prefusion conformation of human metapneumovirus fusion protein (hMPV Pre-F) is critical for eliciting the most potent neutralizing antibodies and is the preferred immunogen for an efficacious vaccine against hMPV respiratory infections. Here we show that an additional cleavage event in the F protein allows closure and correct folding of the trimer. We therefore engineered the F protein to undergo double cleavage, which enabled screening for Pre-F stabilizing substitutions at the natively folded protomer interfaces. To identify these substitutions, we developed an AI convolutional classifier that successfully predicts complex polar interactions often overlooked by physics-based methods and visual inspection. The combination of additional processing, stabilization of interface regions and stabilization of the membrane-proximal stem, resulted in a Pre-F protein vaccine candidate without the need for a heterologous trimerization domain that exhibited high expression yields and thermostability. Cryo-EM analysis shows the complete ectodomain structure, including the stem, and a specific interaction of the newly identified cleaved C-terminus with the adjacent protomer. Importantly, the protein induces high and cross-neutralizing antibody responses resulting in near complete protection against hMPV challenge in cotton rats, making the highly stable, double-cleaved hMPV Pre-F trimer an attractive vaccine candidate.
Collapse
Affiliation(s)
- Mark J G Bakkers
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
- ForgeBio B.V., Amsterdam, The Netherlands
| | - Tina Ritschel
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
- J&J Innovative Medicine Technology, R&D, New Brunswick, NJ, USA
| | | | - Jacobus Dijkman
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
- Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Machine Learning Lab, Informatics Institute, University of Amsterdam, Amsterdam, The Netherlands
| | - Angelo A Zuffianò
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
- Promaton BV, Amsterdam, The Netherlands
| | - Xiaodi Yu
- Structural & Protein Science, Janssen Research and Development, Spring House, PA, 19044, USA
| | | | - Lam Le
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | | | | | - Martijn de Man
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | - Sem Tamara
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | | | - Roland Zahn
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | - Jarek Juraszek
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands
| | - Johannes P M Langedijk
- Janssen Vaccines & Prevention BV, Leiden, The Netherlands.
- ForgeBio B.V., Amsterdam, The Netherlands.
| |
Collapse
|
21
|
Felicetti T, Sarnari C, Gaito R, Tabarrini O, Manfroni G. Recent Progress toward the Discovery of Small Molecules as Novel Anti-Respiratory Syncytial Virus Agents. J Med Chem 2024; 67:11543-11579. [PMID: 38970494 DOI: 10.1021/acs.jmedchem.4c00630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2024]
Abstract
Respiratory syncytial virus (RSV) stands as the foremost cause of infant hospitalization globally, ranking second only to malaria in terms of infant mortality. Although three vaccines have recently been approved for the prophylaxis of adults aged 60 and above, and pregnant women, there is currently no effective antiviral drug for treating RSV infections. The only preventive measure for infants at high risk of severe RSV disease is passive immunization through monoclonal antibodies. This Perspective offers an overview of the latest advancements in RSV drug discovery of small molecule antivirals, with particular focus on the promising findings from agents targeting the fusion and polymerase proteins. A comprehensive reflection on the current state of RSV research is also given, drawing inspiration from the lessons gleaned from HCV and HIV, while also considering the impact of the recent approval of the three vaccines.
Collapse
Affiliation(s)
- Tommaso Felicetti
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Chiara Sarnari
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Roberta Gaito
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| | - Giuseppe Manfroni
- Department of Pharmaceutical Sciences, University of Perugia, Via Del Liceo, 1-06123, Perugia, Italy
| |
Collapse
|
22
|
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
|
23
|
Sibert BS, Kim JY, Yang JE, Ke Z, Stobart CC, Moore ML, Wright ER. Assembly of respiratory syncytial virus matrix protein lattice and its coordination with fusion glycoprotein trimers. Nat Commun 2024; 15:5923. [PMID: 39004634 PMCID: PMC11247094 DOI: 10.1038/s41467-024-50162-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Respiratory syncytial virus (RSV) is an enveloped, filamentous, negative-strand RNA virus that causes significant respiratory illness worldwide. RSV vaccines are available, however there is still significant need for research to support the development of vaccines and therapeutics against RSV and related Mononegavirales viruses. Individual virions vary in size, with an average diameter of ~130 nm and ranging from ~500 nm to over 10 µm in length. Though the general arrangement of structural proteins in virions is known, we use cryo-electron tomography and sub-tomogram averaging to determine the molecular organization of RSV structural proteins. We show that the peripheral membrane-associated RSV matrix (M) protein is arranged in a packed helical-like lattice of M-dimers. We report that RSV F glycoprotein is frequently observed as pairs of trimers oriented in an anti-parallel conformation to support potential interactions between trimers. Our sub-tomogram averages indicate the positioning of F-trimer pairs is correlated with the underlying M lattice. These results provide insight into RSV virion organization and may aid in the development of RSV vaccines and anti-viral targets.
Collapse
Affiliation(s)
- Bryan S Sibert
- Department of Biochemistry, University of Wisconsin, Madison, WI, USA
- Cryo-Electron Microscopy Research Center, Department of Biochemistry, University of Wisconsin, Madison, WI, USA
- Midwest Center for Cryo-Electron Tomography, Department of Biochemistry, University of Wisconsin, Madison, WI, USA
| | - Joseph Y Kim
- Department of Biochemistry, University of Wisconsin, Madison, WI, USA
- Department of Chemistry, University of Wisconsin, Madison, WI, USA
| | - Jie E Yang
- Department of Biochemistry, University of Wisconsin, Madison, WI, USA
- Cryo-Electron Microscopy Research Center, Department of Biochemistry, University of Wisconsin, Madison, WI, USA
- Midwest Center for Cryo-Electron Tomography, Department of Biochemistry, University of Wisconsin, Madison, WI, USA
| | - Zunlong Ke
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, USA
| | | | | | - Elizabeth R Wright
- Department of Biochemistry, University of Wisconsin, Madison, WI, USA.
- Cryo-Electron Microscopy Research Center, Department of Biochemistry, University of Wisconsin, Madison, WI, USA.
- Midwest Center for Cryo-Electron Tomography, Department of Biochemistry, University of Wisconsin, Madison, WI, USA.
- Morgridge Institute for Research, Madison, WI, USA.
| |
Collapse
|
24
|
Mejias A, Ramilo O. RSV Prevention Within Reach for Older Infants and Toddlers: The Role of Active Immunization. J Pediatric Infect Dis Soc 2024; 13:S125-S130. [PMID: 38995087 DOI: 10.1093/jpids/piae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 05/17/2024] [Indexed: 07/13/2024]
Abstract
This review article will summarize the vaccines and monoclonal antibodies currently under evaluation for the prevention of RSV disease in older infants, toddlers and young children. We will review the rationale for passive protection during the first months of life, and the role of active immunization afterwards, either with live attenuated, protein-based or mRNA vaccines.
Collapse
Affiliation(s)
- Asuncion Mejias
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Octavio Ramilo
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, Tennessee, USA
- Department of Pediatrics, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
25
|
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
|
26
|
Bissett C, Belij-Rammerstorfer S, Ulaszewska M, Smith H, Kailath R, Morris S, Powers C, Sebastian S, Sharpe HR, Allen ER, Wang Z, Cunliffe RF, Sallah HJ, Spencer AJ, Gilbert S, Tregoning JS, Lambe T. Systemic prime mucosal boost significantly increases protective efficacy of bivalent RSV influenza viral vectored vaccine. NPJ Vaccines 2024; 9:118. [PMID: 38926455 PMCID: PMC11208422 DOI: 10.1038/s41541-024-00912-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Although licensed vaccines against influenza virus have been successful in reducing pathogen-mediated disease, they have been less effective at preventing viral infection of the airways and current seasonal updates to influenza vaccines do not always successfully accommodate viral drift. Most licensed influenza and recently licensed RSV vaccines are administered via the intramuscular route. Alternative immunisation strategies, such as intranasal vaccinations, and "prime-pull" regimens, may deliver a more sterilising form of protection against respiratory viruses. A bivalent ChAdOx1-based vaccine (ChAdOx1-NP + M1-RSVF) encoding conserved nucleoprotein and matrix 1 proteins from influenza A virus and a modified pre-fusion stabilised RSV A F protein, was designed, developed and tested in preclinical animal models. The aim was to induce broad, cross-protective tissue-resident T cells against heterotypic influenza viruses and neutralising antibodies against RSV in the respiratory mucosa and systemically. When administered via an intramuscular prime-intranasal boost (IM-IN) regimen in mice, superior protection was generated against challenge with either RSV A, Influenza A H3N2 or H1N1. These results support further clinical development of a pan influenza & RSV vaccine administered in a prime-pull regimen.
Collapse
Affiliation(s)
- Cameron Bissett
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK.
| | | | - Marta Ulaszewska
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Holly Smith
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Reshma Kailath
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Susan Morris
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Claire Powers
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Sarah Sebastian
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Hannah R Sharpe
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Elizabeth R Allen
- The Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Ziyin Wang
- Department of Infectious Disease, Imperial College London, London, UK
| | - Robert F Cunliffe
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - Alexandra J Spencer
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Sarah Gilbert
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - John S Tregoning
- Department of Infectious Disease, Imperial College London, London, UK
| | - Teresa Lambe
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| |
Collapse
|
27
|
Hönemann M, Maier M, Frille A, Thiem S, Bergs S, Williams TC, Mas V, Lübbert C, Pietsch C. Respiratory Syncytial Virus in Adult Patients at a Tertiary Care Hospital in Germany: Clinical Features and Molecular Epidemiology of the Fusion Protein in the Severe Respiratory Season of 2022/2023. Viruses 2024; 16:943. [PMID: 38932235 PMCID: PMC11209376 DOI: 10.3390/v16060943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
Following an interseasonal rise in mainly pediatric respiratory syncytial virus (RSV) cases in Germany in 2021, an exceptionally high number of adult cases was observed in the subsequent respiratory season of 2022/2023. The aim of this study was to compare the clinical presentation of RSV infections in the pre- and post-SARS-CoV-2 pandemic periods. Additionally, the local epidemiology of the RSV fusion protein was analyzed at a molecular genetic and amino acid level. RSV detections in adults peaked in calendar week 1 of 2023, 8 weeks earlier than the earliest peak observed in the three pre-pandemic seasons. Although the median age of the adult patients was not different (66.5 vs. 65 years), subtle differences between both periods regarding comorbidities and the clinical presentation of RSV cases were noted. High rates of comorbidities prevailed; however, significantly lower numbers of patients with a history of lung transplantation (p = 0.009), chronic kidney disease (p = 0.013), and immunosuppression (p = 0.038) were observed in the 2022/2023 season. In contrast, significantly more lower respiratory tract infections (p < 0.001), in particular in the form of pneumonia (p = 0.015) and exacerbations of obstructive lung diseases (p = 0.008), were detected. An ICU admission was noted for 23.7% of all patients throughout the study period. Sequence analysis of the fusion protein gene revealed a close phylogenetic relatedness, regardless of the season of origin. However, especially for RSV-B, an accumulation of amino acid point substitutions was noted, including in antigenic site Ø. The SARS-CoV-2 pandemic had a tremendous impact on the seasonality of RSV, and the introduction of new vaccination and immunization strategies against RSV warrants further epidemiologic studies of this important pathogen.
Collapse
Affiliation(s)
- Mario Hönemann
- Virology Department, Institute of Medical Microbiology and Virology, Leipzig University Hospital, Johannisalle 30, 04103 Leipzig, Germany
- Interdisciplinary Center for Infectious Diseases, Leipzig University Hospital, Liebigstrasse 20, 04103 Leipzig, Germany
| | - Melanie Maier
- Virology Department, Institute of Medical Microbiology and Virology, Leipzig University Hospital, Johannisalle 30, 04103 Leipzig, Germany
- Interdisciplinary Center for Infectious Diseases, Leipzig University Hospital, Liebigstrasse 20, 04103 Leipzig, Germany
| | - Armin Frille
- Department of Respiratory Medicine, Leipzig University Hospital, Liebigstrasse 20, 04103 Leipzig, Germany
| | - Stephanie Thiem
- Virology Department, Institute of Medical Microbiology and Virology, Leipzig University Hospital, Johannisalle 30, 04103 Leipzig, Germany
| | - Sandra Bergs
- Virology Department, Institute of Medical Microbiology and Virology, Leipzig University Hospital, Johannisalle 30, 04103 Leipzig, Germany
| | - Thomas C. Williams
- Child Life and Health, University of Edinburgh, Royal Hospital for Children and Young People, 50 Little France Crescent, Edinburgh EH16 4TJ, UK
| | - Vicente Mas
- Centro Nacional de Microbiología and CIBER de Enfermedades Respiratorias, Instituto de Salud Carlos III, C/ Sinesio Delgado, 4, 28029 Madrid, Spain
| | - Christoph Lübbert
- Interdisciplinary Center for Infectious Diseases, Leipzig University Hospital, Liebigstrasse 20, 04103 Leipzig, Germany
- Division of Infectious Diseases and Tropical Medicine, Department of Medicine I, Leipzig University Hospital, Liebigstrasse 20, 04103 Leipzig, Germany
| | - Corinna Pietsch
- Virology Department, Institute of Medical Microbiology and Virology, Leipzig University Hospital, Johannisalle 30, 04103 Leipzig, Germany
- Interdisciplinary Center for Infectious Diseases, Leipzig University Hospital, Liebigstrasse 20, 04103 Leipzig, Germany
| |
Collapse
|
28
|
Yang Q, Xue B, Liu F, Lu Y, Tang J, Yan M, Wu Q, Chen R, Zhou A, Liu L, Liu J, Qu C, Wu Q, Fu M, Zhong J, Dong J, Chen S, Wang F, Zhou Y, Zheng J, Peng W, Shang J, Chen X. Farnesyltransferase inhibitor lonafarnib suppresses respiratory syncytial virus infection by blocking conformational change of fusion glycoprotein. Signal Transduct Target Ther 2024; 9:144. [PMID: 38853183 PMCID: PMC11163014 DOI: 10.1038/s41392-024-01858-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/28/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024] Open
Abstract
Respiratory syncytial virus (RSV) is the major cause of bronchiolitis and pneumonia in young children and the elderly. There are currently no approved RSV-specific therapeutic small molecules available. Using high-throughput antiviral screening, we identified an oral drug, the prenylation inhibitor lonafarnib, which showed potent inhibition of the RSV fusion process. Lonafarnib exhibited antiviral activity against both the RSV A and B genotypes and showed low cytotoxicity in HEp-2 and human primary bronchial epithelial cells (HBEC). Time-of-addition and pseudovirus assays demonstrated that lonafarnib inhibits RSV entry, but has farnesyltransferase-independent antiviral efficacy. Cryo-electron microscopy revealed that lonafarnib binds to a triple-symmetric pocket within the central cavity of the RSV F metastable pre-fusion conformation. Mutants at the RSV F sites interacting with lonafarnib showed resistance to lonafarnib but remained fully sensitive to the neutralizing monoclonal antibody palivizumab. Furthermore, lonafarnib dose-dependently reduced the replication of RSV in BALB/c mice. Collectively, lonafarnib could be a potential fusion inhibitor for RSV infection.
Collapse
Affiliation(s)
- Qi Yang
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Bao Xue
- Guangzhou National Laboratory, Guangzhou, 510005, China
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Fengjiang Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yongzhi Lu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jielin Tang
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Mengrong Yan
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Qiong Wu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Ruyi Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Anqi Zhou
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Lijie Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Junjun Liu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Changbin Qu
- Guangzhou National Laboratory, Guangzhou, 510005, China
| | - Qingxin Wu
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Muqing Fu
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jiayi Zhong
- Guangzhou National Laboratory, Guangzhou, 510005, China
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jianwei Dong
- Guangzhou National Laboratory, Guangzhou, 510005, China
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Sijie Chen
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Fan Wang
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Yuan Zhou
- Guangzhou National Laboratory, Guangzhou, 510005, China
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jie Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou lnstitute for Advanced Study, UCAS, Hangzhou, 310024, China
| | - Wei Peng
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Jinsai Shang
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China.
| | - Xinwen Chen
- Guangzhou National Laboratory, Guangzhou, 510005, China.
- State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, 511436, China.
| |
Collapse
|
29
|
Sanz-Muñoz I, Sánchez-de Prada L, Castrodeza-Sanz J, Eiros JM. Microbiological and epidemiological features of respiratory syncytial virus. REVISTA ESPANOLA DE QUIMIOTERAPIA : PUBLICACION OFICIAL DE LA SOCIEDAD ESPANOLA DE QUIMIOTERAPIA 2024; 37:209-220. [PMID: 38515332 PMCID: PMC11094634 DOI: 10.37201/req/006.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/09/2024] [Accepted: 02/23/2024] [Indexed: 03/23/2024]
Abstract
The properties of the main surface proteins and the viral cycle of the respiratory syncytial virus (RSV) make it an attractive pathogen from the perspective of microbiology. The virus gets its name from the manner it infects cells, which enables it to produce syncytia, which allow the virus' genetic material to move across cells without having to release viral offspring to the cellular exterior, reducing immune system identification. This causes a disease with a high impact in both children and adults over 60, which has sparked the development of several preventive interventions based on vaccines and monoclonal antibodies for both age groups. The epidemiological characteristics of this virus, which circulates in epidemics throughout the coldest months of the year and exhibits a marked genetic and antigenic drift due to its high mutation capability, must be taken into consideration while using these preventive methods. The most important microbiological and epidemiological elements of RSV are covered in this study, along with how they have affected the creation of preventive medications and their use in the future.
Collapse
Affiliation(s)
- I Sanz-Muñoz
- Dr. Iván Sanz-Muñoz, National Influenza Centre, Valladolid, Calle Rondilla de Santa Teresa s/n, Edificio Rondilla, Hospital Clínico Universitario de Valladolid, Valladolid, Spain.
| | | | | | | |
Collapse
|
30
|
Holfeld A, Schuster D, Sesterhenn F, Gillingham AK, Stalder P, Haenseler W, Barrio-Hernandez I, Ghosh D, Vowles J, Cowley SA, Nagel L, Khanppnavar B, Serdiuk T, Beltrao P, Korkhov VM, Munro S, Riek R, de Souza N, Picotti P. Systematic identification of structure-specific protein-protein interactions. Mol Syst Biol 2024; 20:651-675. [PMID: 38702390 PMCID: PMC11148107 DOI: 10.1038/s44320-024-00037-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/12/2024] [Accepted: 04/15/2024] [Indexed: 05/06/2024] Open
Abstract
The physical interactome of a protein can be altered upon perturbation, modulating cell physiology and contributing to disease. Identifying interactome differences of normal and disease states of proteins could help understand disease mechanisms, but current methods do not pinpoint structure-specific PPIs and interaction interfaces proteome-wide. We used limited proteolysis-mass spectrometry (LiP-MS) to screen for structure-specific PPIs by probing for protease susceptibility changes of proteins in cellular extracts upon treatment with specific structural states of a protein. We first demonstrated that LiP-MS detects well-characterized PPIs, including antibody-target protein interactions and interactions with membrane proteins, and that it pinpoints interfaces, including epitopes. We then applied the approach to study conformation-specific interactors of the Parkinson's disease hallmark protein alpha-synuclein (aSyn). We identified known interactors of aSyn monomer and amyloid fibrils and provide a resource of novel putative conformation-specific aSyn interactors for validation in further studies. We also used our approach on GDP- and GTP-bound forms of two Rab GTPases, showing detection of differential candidate interactors of conformationally similar proteins. This approach is applicable to screen for structure-specific interactomes of any protein, including posttranslationally modified and unmodified, or metabolite-bound and unbound protein states.
Collapse
Affiliation(s)
- Aleš Holfeld
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Dina Schuster
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, Villigen, Switzerland
| | - Fabian Sesterhenn
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | | | - Patrick Stalder
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Walther Haenseler
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
- University Research Priority Program AdaBD (Adaptive Brain Circuits in Development and Learning), University of Zurich, Zurich, Switzerland
| | - Inigo Barrio-Hernandez
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Open Targets, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Dhiman Ghosh
- Laboratory of Physical Chemistry, ETH Zurich, Zurich, Switzerland
| | - Jane Vowles
- James and Lillian Martin Centre for Stem Cell Research, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Sally A Cowley
- James and Lillian Martin Centre for Stem Cell Research, Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Luise Nagel
- Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Basavraj Khanppnavar
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, Villigen, Switzerland
| | - Tetiana Serdiuk
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Pedro Beltrao
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Volodymyr M Korkhov
- Institute of Molecular Biology and Biophysics, Department of Biology, ETH Zurich, Zurich, Switzerland
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, Villigen, Switzerland
| | - Sean Munro
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Roland Riek
- Laboratory of Physical Chemistry, ETH Zurich, Zurich, Switzerland
| | - Natalie de Souza
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland
- Department of Quantitative Biomedicine, University of Zurich, Zurich, Switzerland
| | - Paola Picotti
- Institute of Molecular Systems Biology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
31
|
Trinité B, Durr E, Pons-Grífols A, O'Donnell G, Aguilar-Gurrieri C, Rodriguez S, Urrea V, Tarrés F, Mane J, Ortiz R, Rovirosa C, Carrillo J, Clotet B, Zhang L, Blanco J. VLPs generated by the fusion of RSV-F or hMPV-F glycoprotein to HIV-Gag show improved immunogenicity and neutralizing response in mice. Vaccine 2024; 42:3474-3485. [PMID: 38641492 DOI: 10.1016/j.vaccine.2024.04.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/26/2024] [Accepted: 04/14/2024] [Indexed: 04/21/2024]
Abstract
Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) vaccines have been long overdue. Structure-based vaccine design created a new momentum in the last decade, and the first RSV vaccines have finally been approved in older adults and pregnant individuals. These vaccines are based on recombinant stabilized pre-fusion F glycoproteins administered as soluble proteins. Multimeric antigenic display could markedly improve immunogenicity and should be evaluated in the next generations of vaccines. Here we tested a new virus like particles-based vaccine platform which utilizes the direct fusion of an immunogen of interest to the structural human immunodeficient virus (HIV) protein Gag to increase its surface density and immunogenicity. We compared, in mice, the immunogenicity of RSV-F or hMPV-F based immunogens delivered either as soluble proteins or displayed on the surface of our VLPs. VLP associated F-proteins showed better immunogenicity and induced superior neutralizing responses. Moreover, when combining both VLP associated and soluble immunogens in a heterologous regimen, VLP-associated immunogens provided added benefits when administered as the prime immunization.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Neutralizing/blood
- Mice
- Metapneumovirus/immunology
- Vaccines, Virus-Like Particle/immunology
- Vaccines, Virus-Like Particle/administration & dosage
- Female
- Viral Fusion Proteins/immunology
- Viral Fusion Proteins/genetics
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Mice, Inbred BALB C
- gag Gene Products, Human Immunodeficiency Virus/immunology
- gag Gene Products, Human Immunodeficiency Virus/genetics
- Respiratory Syncytial Virus, Human/immunology
- Immunogenicity, Vaccine
- Humans
- Respiratory Syncytial Virus Vaccines/immunology
- Respiratory Syncytial Virus Vaccines/administration & dosage
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/genetics
- Respiratory Syncytial Virus Infections/prevention & control
- Respiratory Syncytial Virus Infections/immunology
- Viral Vaccines/immunology
- Viral Vaccines/administration & dosage
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Bonaventura Clotet
- IrsiCaixa, Badalona, Spain; University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain
| | | | - Julià Blanco
- IrsiCaixa, Badalona, Spain; University of Vic-Central University of Catalonia (UVic-UCC), Vic, Spain; Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain; CIBERINFEC, Madrid, Spain.
| |
Collapse
|
32
|
Wang Z, McCallum M, Yan L, Gibson CA, Sharkey W, Park YJ, Dang HV, Amaya M, Person A, Broder CC, Veesler D. Structure and design of Langya virus glycoprotein antigens. Proc Natl Acad Sci U S A 2024; 121:e2314990121. [PMID: 38593070 PMCID: PMC11032465 DOI: 10.1073/pnas.2314990121] [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: 08/31/2023] [Accepted: 03/04/2024] [Indexed: 04/11/2024] Open
Abstract
Langya virus (LayV) is a recently discovered henipavirus (HNV), isolated from febrile patients in China. HNV entry into host cells is mediated by the attachment (G) and fusion (F) glycoproteins which are the main targets of neutralizing antibodies. We show here that the LayV F and G glycoproteins promote membrane fusion with human, mouse, and hamster target cells using a different, yet unknown, receptor than Nipah virus (NiV) and Hendra virus (HeV) and that NiV- and HeV-elicited monoclonal and polyclonal antibodies do not cross-react with LayV F and G. We determined cryoelectron microscopy structures of LayV F, in the prefusion and postfusion states, and of LayV G, revealing their conformational landscape and distinct antigenicity relative to NiV and HeV. We computationally designed stabilized LayV G constructs and demonstrate the generalizability of an HNV F prefusion-stabilization strategy. Our data will support the development of vaccines and therapeutics against LayV and closely related HNVs.
Collapse
Affiliation(s)
- Zhaoqian Wang
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - Matthew McCallum
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - Lianying Yan
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD20814
| | - Cecily A. Gibson
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - William Sharkey
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - Young-Jun Park
- Department of Biochemistry, University of Washington, Seattle, WA98195
- HHMI, Seattle, WA98195
| | - Ha V. Dang
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - Moushimi Amaya
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD20814
| | - Ashley Person
- Department of Biochemistry, University of Washington, Seattle, WA98195
| | - Christopher C. Broder
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD20814
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA98195
- HHMI, Seattle, WA98195
| |
Collapse
|
33
|
Sanz-Muñoz I, Castrodeza-Sanz J, Eiros JM. Potential Effects on Elderly People From Nirsevimab Use in Infants. OPEN RESPIRATORY ARCHIVES 2024; 6:100320. [PMID: 38617129 PMCID: PMC11015503 DOI: 10.1016/j.opresp.2024.100320] [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: 01/30/2024] [Accepted: 03/10/2024] [Indexed: 04/16/2024] Open
Abstract
Nirsevimab therapy has the potential to revolutionize infant respiratory syncytial virus (RSV) prophylaxis. But other populations suffering RSV, such the elderly or those over 60, may also be protected by using this novel antibody in the infant group. It is true that some studies link the use of nirsevimab to a reduction in the virus's ability to spread by lowering the viral load in infants as a result of the drug's long half-life. However, this protective effect may not be very significant because RSV transmission in the elderly typically comes from other elderly people or from school-aged children. Furthermore, RSV may be transmitted at any time of the year and not just during the period of nirsevimab protection due to its existence in human reservoirs. The reasons made here show that, even though nirsevimab treatment in infants may protect the elderly, this benefit would be limited and testimonial. Therefore, immunizing the elderly with currently licensed and developing vaccines should be a priority.
Collapse
Affiliation(s)
- Iván Sanz-Muñoz
- National Influenza Centre, Valladolid, Spain
- Instituto de Estudios de Ciencias de la Salud de Castilla y León, ICSCYL, Soria, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFECC), Spain
| | - Javier Castrodeza-Sanz
- National Influenza Centre, Valladolid, Spain
- Preventive Medicine and Public Health Unit, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
- Preventive Medicine and Public Department, Faculty of Medicine, Universidad de Valladolid, Valladolid, Spain
| | - José M. Eiros
- National Influenza Centre, Valladolid, Spain
- Microbiology Unit, Hospital Universitario Río Hortega, Valladolid, Spain
- Microbiology Unit, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
- Microbiology Department, Faculty of Medicine, Universidad de Valladolid, Valladolid, Spain
| |
Collapse
|
34
|
Hak SF, Venekamp RP, Wildenbeest JG, Bont LJ. Outpatient respiratory syncytial virus infections and novel preventive interventions. Curr Opin Pediatr 2024; 36:171-181. [PMID: 38085019 PMCID: PMC10919273 DOI: 10.1097/mop.0000000000001323] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
PURPOSE OF REVIEW With interventions to prevent respiratory syncytial virus (RSV) infection within reach, this review aims to provide healthcare professionals with the latest information necessary to inform parents and assess the potential impact of RSV prevention on everyday practice. We address frequently asked questions for parental counseling. RECENT FINDINGS Numerous studies emphasize the major burden of RSV on young children, parents, healthcare and society. In the first year of life, about 14% of healthy term infants visit a doctor and 2% require hospitalization due to RSV. In older children (1--5 years), RSV infections and associated morbidity (wheeze, acute otitis media) are major drivers of outpatient visits. A novel maternal RSV vaccine and long-acting mAb can provide protection during infants' first months of life. This maternal vaccine showed 70.9% efficacy against severe RSV infection within 150 days after birth; the mAb nirsevimab reduces medically attended RSV infections by 79.5% within 150 days after administration. Both gained regulatory approval in the USA (FDA) and Europe (EMA). SUMMARY Novel RSV immunizations hold promise to reduce the RSV burden in infants, with substantial impact on everyday practice. Tailored parental guidance will be instrumental for successful implementation. Awaiting pediatric vaccines, RSV infections beyond infancy will still pose a significant outpatient burden.
Collapse
Affiliation(s)
- Sarah F. Hak
- Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital/University Medical Center Utrecht
| | - Roderick P. Venekamp
- Department of General Practice & Nursing Science, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht
| | - Joanne G. Wildenbeest
- Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital/University Medical Center Utrecht
| | - Louis J. Bont
- Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital/University Medical Center Utrecht
- Respiratory Syncytial Virus NETwork (ReSViNET) Foundation, Zeist, The Netherlands
| |
Collapse
|
35
|
Shao S, Zhang XF, Hou JW, Yang SS, Han ZB, Wu HL, Tang F, Li XY, Lei ZH, Zhao ZX, Li SX, Liu ZM, Shan P, Jin YQ, Su JG, Liang Y, Zhang J, Li QM. Design of hepadnavirus core protein-based chimeric virus-like particles carrying epitopes from respiratory syncytial virus. NPJ Vaccines 2024; 9:62. [PMID: 38503757 PMCID: PMC10951352 DOI: 10.1038/s41541-024-00855-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 02/26/2024] [Indexed: 03/21/2024] Open
Abstract
Respiratory syncytial virus (RSV) is one of the most important pathogens causing respiratory tract infection in humans, especially in infants and the elderly. The identification and structural resolution of the potent neutralizing epitopes on RSV fusion (F) protein enable an "epitope-focused" vaccine design. However, the display of RSV F epitope II on the surface of the widely-used human hepatitis B virus core antigen (HBcAg) has failed to induce neutralizing antibody response in mice. Here, we used the hepadnavirus core protein (HcAg) from different mammalian hosts as scaffolds to construct chimeric virus-like particles (VLPs) presenting the RSV F epitope II. Mouse immunization showed that different HcAg-based chimeric VLPs elicited significantly different neutralizing antibody responses, among which the HcAg derived from roundleaf bat (RBHcAg) is the most immunogenic. Furthermore, RBHcAg was used as the scaffold platform to present multiple RSV F epitopes, and the immunogenicity was further improved in comparison to that displaying a single epitope II. The designed RBHcAg-based multiple-epitope-presenting VLP formulated with MF59-like adjuvant elicited a potent and balanced Th1/Th2 immune response, and offered substantial protection in mice against the challenge of live RSV A2 virus. The designed chimeric VLPs may serve as the potential starting point for developing epitope-focused vaccines against RSV. Our study also demonstrated that RBHcAg is an effective VLP carrier for presenting foreign epitopes, providing a promising platform for epitope-focused vaccine design.
Collapse
Affiliation(s)
- Shuai Shao
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
- National Engineering Center for New Vaccine Research, Beijing, China
| | - Xue Feng Zhang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
- National Engineering Center for New Vaccine Research, Beijing, China
| | - Jun Wei Hou
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
- National Engineering Center for New Vaccine Research, Beijing, China
| | - Sen Sen Yang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
- National Engineering Center for New Vaccine Research, Beijing, China
| | - Zi Bo Han
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
- National Engineering Center for New Vaccine Research, Beijing, China
| | - Hai Lan Wu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
- National Engineering Center for New Vaccine Research, Beijing, China
| | - Fang Tang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
- National Engineering Center for New Vaccine Research, Beijing, China
| | - Xin Yu Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
- National Engineering Center for New Vaccine Research, Beijing, China
| | - Ze Hua Lei
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
- National Engineering Center for New Vaccine Research, Beijing, China
| | - Zi Xin Zhao
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
- National Engineering Center for New Vaccine Research, Beijing, China
| | - Shu Xiang Li
- National Engineering Center for New Vaccine Research, Beijing, China
- The Third Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
| | - Zhao Ming Liu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
- National Engineering Center for New Vaccine Research, Beijing, China
| | - Pu Shan
- National Engineering Center for New Vaccine Research, Beijing, China
- The Third Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
| | - Yu Qin Jin
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
- National Engineering Center for New Vaccine Research, Beijing, China
| | - Ji Guo Su
- National Engineering Center for New Vaccine Research, Beijing, China
- High Performance Computing Center, National Vaccine and Serum Institute (NVSI), Beijing, China
| | - Yu Liang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China
- National Engineering Center for New Vaccine Research, Beijing, China
| | - Jing Zhang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China.
- National Engineering Center for New Vaccine Research, Beijing, China.
| | - Qi Ming Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI), Beijing, China.
- National Engineering Center for New Vaccine Research, Beijing, China.
| |
Collapse
|
36
|
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
|
37
|
Huong TN, Lee ZQ, Lai SK, Lee HY, Tan BH, Sugrue RJ. Evidence that an interaction between the respiratory syncytial virus F and G proteins at the distal ends of virus filaments mediates efficient multiple cycle infection. Virology 2024; 591:109985. [PMID: 38227992 DOI: 10.1016/j.virol.2024.109985] [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: 08/21/2023] [Revised: 11/29/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024]
Abstract
Evidence for a stable interaction between the respiratory syncytial virus (RSV) F and G proteins on the surface of virus filaments was provided using antibody immunoprecipitation studies on purified RSV particles, and by the in situ analysis on the surface of RSV-infected cells using the proximity ligation assay. Imaging of the F and G protein distribution on virus filaments suggested that this protein complex was localised at the distal ends of the virus filaments, and suggested that this protein complex played a direct role in mediating efficient localised cell-to-cell virus transmission. G protein expression was required for efficient localised cell-to-cell transmission of RSV in cell monolayers which provided evidence that this protein complex mediates efficient multiple cycle infection. Collectively, these data provide evidence that F and G proteins form a complex on the surface of RSV particles, and that a role for this protein complex in promoting virus transmission is suggested.
Collapse
Affiliation(s)
- Tra Nguyen Huong
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore
| | - Zhi Qi Lee
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore
| | - Soak Kuan Lai
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore
| | - Hsin Yee Lee
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore
| | - Boon Huan Tan
- LKC School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Republic of Singapore
| | - Richard J Sugrue
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Republic of Singapore.
| |
Collapse
|
38
|
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
|
39
|
Loaiza RA, Ramírez RA, Sepúlveda-Alfaro J, Ramírez MA, Andrade CA, Soto JA, González PA, Bueno SM, Kalergis AM. A molecular perspective for the development of antibodies against the human respiratory syncytial virus. Antiviral Res 2024; 222:105783. [PMID: 38145755 DOI: 10.1016/j.antiviral.2023.105783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/27/2023]
Abstract
The human respiratory syncytial virus (hRSV) is the leading etiologic agent causing respiratory infections in infants, children, older adults, and patients with comorbidities. Sixty-seven years have passed since the discovery of hRSV, and only a few successful mitigation or treatment tools have been developed against this virus. One of these is immunotherapy with monoclonal antibodies against structural proteins of the virus, such as Palivizumab, the first prophylactic approach approved by the Food and Drug Administration (FDA) of the USA. In this article, we discuss different strategies for the prevention and treatment of hRSV infection, focusing on the molecular mechanisms against each target that underly the rational design of antibodies against hRSV. At the same time, we describe the latest results regarding currently approved therapies against hRSV and the challenges associated with developing new candidates.
Collapse
Affiliation(s)
- Ricardo A Loaiza
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Robinson A Ramírez
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Javiera Sepúlveda-Alfaro
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Mario A Ramírez
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Catalina A Andrade
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Jorge A Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de La Vida, Universidad Andrés Bello, Santiago, Chile
| | - Pablo A González
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile
| | - Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Chile; Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile.
| |
Collapse
|
40
|
Zou G, Cao S, Gao Z, Yie J, Wu JZ. Current state and challenges in respiratory syncytial virus drug discovery and development. Antiviral Res 2024; 221:105791. [PMID: 38160942 DOI: 10.1016/j.antiviral.2023.105791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/03/2024]
Abstract
Human respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infections (LRTI) in young children and elderly people worldwide. Recent significant progress in our understanding of the structure and function of RSV proteins has led to the discovery of several clinical candidates targeting RSV fusion and replication. These include both the development of novel small molecule interventions and the isolation of potent monoclonal antibodies. In this review, we summarize the state-of-the-art of RSV drug discovery, with a focus on the characteristics of the candidates that reached the clinical stage of development. We also discuss the lessons learned from failed and discontinued clinical developments and highlight the challenges that remain for development of RSV therapies.
Collapse
Affiliation(s)
- Gang Zou
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China.
| | - Sushan Cao
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China
| | - Zhao Gao
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China
| | - Junming Yie
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China
| | - Jim Zhen Wu
- Shanghai Ark Biopharmaceutical Co., Ltd, Shanghai, 201203, China
| |
Collapse
|
41
|
Mas V, Melero JA. Entry of Enveloped Viruses into Host Cells: Membrane Fusion. Subcell Biochem 2024; 105:567-592. [PMID: 39738958 DOI: 10.1007/978-3-031-65187-8_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
Abstract
Viruses are intracellular parasites that hijack the cellular machinery for their own replication. Therefore, an obligatory step in the virus life cycle is the delivery of the viral genome inside the cell. Enveloped viruses (i.e., viruses with a lipid envelope) use a two-step procedure to release their genetic material into the cell: (1) they first bind to specific surface receptors of the target cell membrane and then (2) they fuse the viral and cell membranes. This last step may occur at the cell surface or after internalization of the virus particle by endocytosis or by some other route (e.g., macropinocytosis). Remarkably, the virus-cell membrane fusion process goes essentially along the same intermediate steps than other membrane fusions that occur, for instance, in vesicular fusion at the nerve synapsis or cell-cell fusion in yeast mating. Specialized viral proteins, fusogens, promote virus-cell membrane fusion. The viral fusogens experience drastic structural rearrangements during fusion, releasing the energy required to overcome the repulsive forces that prevent spontaneous fusion of the two membranes. This chapter provides an overview of the different types of viral fusogens and their mode of action, as they are currently known. Furthermore, it outlines novel strategies for vaccine development related to stabilized viral fusogens.
Collapse
Affiliation(s)
- Vicente Mas
- Unidad de Biología Viral, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain.
| | - Jose Antonio Melero
- Unidad de Biología Viral, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
42
|
Xiao X, Wen Z, Chen Q, Shipman JM, Kostas J, Reid JC, Warren C, Tang A, Luo B, O’Donnell G, Fridman A, Chen Z, Vora KA, Zhang L, Su HP, Eddins MJ. Structural characterization of M8C10, a neutralizing antibody targeting a highly conserved prefusion-specific epitope on the metapneumovirus fusion trimerization interface. J Virol 2023; 97:e0105223. [PMID: 38032197 PMCID: PMC10734504 DOI: 10.1128/jvi.01052-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
IMPORTANCE Human metapneumovirus (hMPV) is a common pathogen causing lower respiratory tract infections worldwide and can develop severe symptoms in high-risk populations such as infants, the elderly, and immunocompromised patients. There are no approved hMPV vaccines or neutralizing antibodies available for therapeutic or prophylactic use. The trimeric hMPV fusion F protein is the major target of neutralizing antibodies in human sera. Understanding the immune recognition of antibodies to hMPV-F antigen will provide critical insights into developing efficacious hMPV monoclonal antibodies and vaccines.
Collapse
Affiliation(s)
- Xiao Xiao
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania, USA
- Discovery Biologics, Merck & Co., Inc., Boston, Massachusetts, USA
- MRL Postdoctoral Research Program, Merck & Co., Inc., Kenilworth, New Jersey, USA
| | - Zhiyun Wen
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Qing Chen
- Protein and Structural Chemistry, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Jennifer M. Shipman
- Protein and Structural Chemistry, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - James Kostas
- Protein and Structural Chemistry, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - John C. Reid
- Protein and Structural Chemistry, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Christopher Warren
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Aimin Tang
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Bin Luo
- Quantitative Biosciences, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Gregory O’Donnell
- Quantitative Biosciences, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Arthur Fridman
- Data Science and Scientific Informatics, Merck & Co., Inc., Rahway, New Jersey, USA
| | - Zhifeng Chen
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Kalpit A. Vora
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Lan Zhang
- Infectious Diseases and Vaccines Discovery, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Hua-Poo Su
- Protein and Structural Chemistry, Merck & Co., Inc., West Point, Pennsylvania, USA
| | - Michael J. Eddins
- Protein and Structural Chemistry, Merck & Co., Inc., West Point, Pennsylvania, USA
| |
Collapse
|
43
|
Kawahara E, Shibata T, Hirai T, Yoshioka Y. Non-glycosylated G protein with CpG ODN provides robust protection against respiratory syncytial virus without inducing eosinophilia. Front Immunol 2023; 14:1282016. [PMID: 38169867 PMCID: PMC10758452 DOI: 10.3389/fimmu.2023.1282016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Introduction Respiratory syncytial virus (RSV) vaccines targeting the fusion glycoprotein (F protein) are highly effective clinically in preventing RSV challenges. The attachment glycoprotein (G protein) is a potentially effective vaccine antigen candidate, as it is important for cell adhesion during infection. However, vaccine-associated enhanced diseases in mice, such as eosinophilic lung inflammation following RSV challenge, are a concern with G protein vaccines. This study aimed to design an effective G protein vaccine with enhanced safety and efficacy by evaluating the efficacy and adverse reactions of vaccines composed of different recombinant G proteins and adjuvants in mice. Methods Mice were subcutaneously immunized with glycosylated G protein expressed in mammalian cells (mG), non-glycosylated G protein expressed in Escherichia coli (eG), or F protein with or without aluminum salts (alum), CpG oligodeoxynucleotide (CpG ODN), or AddaVax. After vaccination, the levels of G-specific antibody and T-cell responses were measured. The immunized mice were challenged with RSV and examined for the viral load in the lungs and nasal turbinates, lung-infiltrating cells, and lung pathology. Results mG with any adjuvant was ineffective at inducing G-specific antibodies and had difficulty achieving both protection against RSV challenge and eosinophilia suppression. In particular, mG+CpG ODN induced G-specific T helper 1 (Th1) cells but only a few G-specific antibodies and did not protect against RSV challenge. However, eG+CpG ODN induced high levels of G-specific antibodies and Th1 cells and protected against RSV challenge without inducing pulmonary inflammation. Moreover, the combination vaccine of eG+F+CpG ODN showed greater protection against upper respiratory tract RSV challenge than using each single antigen vaccine alone. Discussion These results indicate that the efficacy of recombinant G protein vaccines can be enhanced without inducing adverse reactions by using appropriate antigens and adjuvants, and their efficacy is further enhanced in the combination vaccine with F protein. These data provide valuable information for the clinical application of G protein vaccines.
Collapse
Affiliation(s)
- Eigo Kawahara
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Vaccine Creation Group, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Takehiko Shibata
- Department of Microbiology, Tokyo Medical University, Tokyo, Japan
| | - Toshiro Hirai
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Vaccine Creation Group, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
| | - Yasuo Yoshioka
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
- Vaccine Creation Group, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Osaka, Japan
- Innovative Vaccine Research and Development Center, The Research Foundation for Microbial Diseases of Osaka University, Osaka, Japan
- Global Center for Medical Engineering and Informatics, Osaka University, Osaka, Japan
- Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| |
Collapse
|
44
|
Kawahara E, Yamamoto S, Shibata T, Hirai T, Yoshioka Y. CpG ODN enhances the efficacy of F protein vaccine against respiratory syncytial virus infection in the upper respiratory tract via CD4 + T cells. Biochem Biophys Res Commun 2023; 686:149143. [PMID: 37926041 DOI: 10.1016/j.bbrc.2023.149143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 10/22/2023] [Indexed: 11/07/2023]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of severe respiratory illness worldwide, particularly in infants and older adults. Vaccines targeting the fusion glycoprotein (F protein) -one of the surface antigens of RSV- are highly effective in preventing RSV-associated severe lower respiratory tract disease. However, the efficacy of these vaccines against upper respiratory tract challenge needs improvement. Here, we aimed to examine the efficacy of F protein vaccines with or without CpG oligodeoxynucleotide (CpG ODN) as an adjuvant in the upper and lower respiratory tracts in mice. F + CpG ODN induced higher levels of F-specific IgG than that induced by F alone; however, levels of neutralizing antibodies did not increase compared to those induced by F alone. F + CpG ODN induced T helper 1 (Th1) cells while F alone induced T helper 2 (Th2) cells. Moreover, F + CpG ODN improved the protection against RSV challenge in the upper respiratory tract compared to F alone, which was largely dependent on CD4+ T cells. Meanwhile, both F + CpG ODN and F alone protected the lower respiratory tract. In conclusion, we demonstrated that induction of F-specific Th1 cells is an effective strategy to prevent RSV challenge in the upper respiratory tract in F protein vaccines. These data support the development of novel F protein vaccines.
Collapse
Affiliation(s)
- Eigo Kawahara
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, Research Institute for Microbial Diseases, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shinya Yamamoto
- The Research Foundation for Microbial Diseases of Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takehiko Shibata
- Department of Microbiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku-ku, Tokyo, Japan
| | - Toshiro Hirai
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, Research Institute for Microbial Diseases, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yasuo Yoshioka
- Laboratory of Nano-design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; Vaccine Creation Group, Research Institute for Microbial Diseases, Osaka University, 1-6 Yamadaoka, Suita, Osaka, 565-0871, Japan; The Research Foundation for Microbial Diseases of Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Institute for Open and Transdisciplinary Research Initiatives, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Center for Advanced Modalities and DDS, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Center for Infectious Disease Education and Research, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan; Global Center for Medical Engineering and Informatics, Osaka University, 3-1 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
45
|
Kopera E, Czajka H, Zapolnik P, Mazur A. New Insights on Respiratory Syncytial Virus Prevention. Vaccines (Basel) 2023; 11:1797. [PMID: 38140201 PMCID: PMC10747926 DOI: 10.3390/vaccines11121797] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/20/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a well-known infant pathogen transmitted mainly by droplets. It is a leading cause of upper respiratory tract infections in children, usually with a mild course of illness. RSV has also been a threat to older people, especially those with underlying medical conditions. For a long time, prevention was limited to passive immunoprophylaxis with palivizumab for high-risk infants. There was a strong need to find other treatment or prevention methods against RSV infections. In addition, after the coronavirus disease 2019 (COVID-19) pandemic, some significant changes in RSV epidemiology have been observed. Researchers noticed the shift in RSV seasonality and age distribution and the increased number of cases in older infants and adults. All of these made the need to find other medical options even stronger. Fortunately, two protein-based vaccines against RSV have successfully passed all phases of clinical trials and have been approved for use by adults and older people. One of them is also approved for infants from birth to 6 months of age (after maternal immunisation during pregnancy) and for pregnant women between 24 and 36 weeks of pregnancy. Also, a new passive immunisation option named nirsevimab (a highly potent monoclonal antibody with a long half-life) is now available for the paediatric group. In this review, we will discuss the previous and current RSV prevention methods in the light of structural discoveries of RSV antigens.
Collapse
Affiliation(s)
| | - Hanna Czajka
- College of Medical Sciences, University of Rzeszów, 35-315 Rzeszów, Poland; (E.K.); (P.Z.); (A.M.)
| | | | | |
Collapse
|
46
|
McCool RS, Musayev M, Bush SM, Derrien-Colemyn A, Acreman CM, Wrapp D, Ruckwardt TJ, Graham BS, Mascola JR, McLellan JS. Vaccination with prefusion-stabilized respiratory syncytial virus fusion protein elicits antibodies targeting a membrane-proximal epitope. J Virol 2023; 97:e0092923. [PMID: 37737588 PMCID: PMC10617438 DOI: 10.1128/jvi.00929-23] [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: 06/24/2023] [Accepted: 07/31/2023] [Indexed: 09/23/2023] Open
Abstract
IMPORTANCE Respiratory syncytial virus (RSV) is the leading cause of bronchiolitis and pneumonia in infants, infecting all children by age 5. RSV also causes substantial morbidity and mortality in older adults, and a vaccine for older adults based on a prefusion-stabilized form of the viral F glycoprotein was recently approved by the FDA. Here, we investigate a set of antibodies that belong to the same public clonotype and were isolated from individuals vaccinated with a prefusion-stabilized RSV F protein. Our results reveal that these antibodies are highly potent and recognize a previously uncharacterized antigenic site on the prefusion F protein. Vaccination with prefusion RSV F proteins appears to boost the elicitation of these neutralizing antibodies, which are not commonly elicited by natural infection.
Collapse
Affiliation(s)
- Ryan S. McCool
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Maryam Musayev
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sabrina M. Bush
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexandrine Derrien-Colemyn
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Cory M. Acreman
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Daniel Wrapp
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| | - Tracy J. Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Barney S. Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - John R. Mascola
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jason S. McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas, USA
| |
Collapse
|
47
|
Nussbaum J, Cao X, Railkar RA, Sachs JR, Spellman DS, Luk J, Shaw CA, Cejas PJ, Citron MP, Al-Ibrahim M, Han D, Pagnussat S, Stoch SA, Lai E, Bett AJ, Espeseth AS. Evaluation of a stabilized RSV pre-fusion F mRNA vaccine: Preclinical studies and Phase 1 clinical testing in healthy adults. Vaccine 2023; 41:6488-6501. [PMID: 37777449 DOI: 10.1016/j.vaccine.2023.05.062] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/15/2023] [Accepted: 05/26/2023] [Indexed: 10/02/2023]
Abstract
Human respiratory syncytial virus (RSV) causes a substantial proportion of respiratory tract infections worldwide. Although RSV reinfections occur throughout life, older adults, particularly those with underlying comorbidities, are at risk for severe complications from RSV. There is no RSV vaccine available to date, and treatment of RSV in adults is largely supportive. A correlate of protection for RSV has not yet been established, but antibodies targeting the pre-fusion conformation of the RSV F glycoprotein play an important role in RSV neutralization. We previously reported a Phase 1 study of an mRNA-based vaccine (V171) expressing a pre-fusion-stabilized RSV F protein (mDS-Cav1) in healthy adults. Here, we evaluated an mRNA-based vaccine (V172) expressing a further stabilized RSV pre-fusion F protein (mVRC1). mVRC1 is a single chain version of RSV F with interprotomer disulfides in addition to the stabilizing mutations present in the mDS-Cav1 antigen. The immunogenicity of the two mRNA-based vaccines encoding mVRC1 (V172) or a sequence-optimized version of mDS-Cav1 to improve transcriptional fidelity (V171.2) were compared in RSV-naïve and RSV-experienced African green monkeys (AGMs). V172 induced higher neutralizing antibody titers than V171.2 and demonstrated protection in the AGM challenge model. We conducted a Phase 1, randomized, placebo-controlled, clinical trial of 25 μg, 100 μg, 200 μg, or 300 μg of V172 in healthy older adults (60-79 years old; N = 112) and 100 μg, 200 μg, or 300 μg of V172 in healthy younger adults (18-49 years old; N = 48). The primary clinical objectives were to evaluate the safety and tolerability of V172, and the secondary objective was to evaluate RSV serum neutralization titers. The most commonly reported solicited adverse events were injection-site pain, injection-site swelling, headache, and tiredness. V172 was generally well tolerated in older and younger adults and increased serum neutralizing antibody titers, pre-fusion F-specific competing antibody titers, and RSV F-specific T-cell responses.
Collapse
Affiliation(s)
| | - Xin Cao
- Merck & Co., Inc., Rahway, NJ, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Díaz FE, McGill JL. Modeling Human Respiratory Syncytial Virus (RSV) Infection: Recent Contributions and Future Directions Using the Calf Model of Bovine RSV Disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1180-1186. [PMID: 37782855 PMCID: PMC10558079 DOI: 10.4049/jimmunol.2300260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/24/2023] [Indexed: 10/04/2023]
Abstract
The human orthopneumovirus (human respiratory syncytial virus [RSV]) is a leading cause of respiratory disease in children worldwide and a significant cause of infant mortality in low- and middle-income countries. The natural immune response to the virus has a preponderant role in disease progression, with a rapid neutrophil infiltration and dysbalanced T cell response in the lungs associated with severe disease in infants. The development of preventive interventions against human RSV has been difficult partly due to the need to use animal models that only partially recapitulate the immune response as well as the disease progression seen in human infants. In this brief review, we discuss the contributions of the calf model of RSV infection to understanding immunity to RSV and in developing vaccine and drug candidates, focusing on recent research areas. We propose that the bovine model of RSV infection is a valuable alternative for assessing the translational potential of interventions aimed at the human population.
Collapse
Affiliation(s)
- Fabián E. Díaz
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| | - Jodi L. McGill
- Department of Veterinary Microbiology and Preventive Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
49
|
Ols S, Lenart K, Arcoverde Cerveira R, Miranda MC, Brunette N, Kochmann J, Corcoran M, Skotheim R, Philomin A, Cagigi A, Fiala B, Wrenn S, Marcandalli J, Hellgren F, Thompson EA, Lin A, Gegenfurtner F, Kumar A, Chen M, Phad GE, Graham BS, Perez L, Borst AJ, Karlsson Hedestam GB, Ruckwardt TJ, King NP, Loré K. Multivalent antigen display on nanoparticle immunogens increases B cell clonotype diversity and neutralization breadth to pneumoviruses. Immunity 2023; 56:2425-2441.e14. [PMID: 37689061 DOI: 10.1016/j.immuni.2023.08.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 05/19/2023] [Accepted: 08/16/2023] [Indexed: 09/11/2023]
Abstract
Nanoparticles for multivalent display and delivery of vaccine antigens have emerged as a promising avenue for enhancing B cell responses to protein subunit vaccines. Here, we evaluated B cell responses in rhesus macaques immunized with prefusion-stabilized respiratory syncytial virus (RSV) F glycoprotein trimer compared with nanoparticles displaying 10 or 20 copies of the same antigen. We show that multivalent display skews antibody specificities and drives epitope-focusing of responding B cells. Antibody cloning and repertoire sequencing revealed that focusing was driven by the expansion of clonally distinct B cells through recruitment of diverse precursors. We identified two antibody lineages that developed either ultrapotent neutralization or pneumovirus cross-neutralization from precursor B cells with low initial affinity for the RSV-F immunogen. This suggests that increased avidity by multivalent display facilitates the activation and recruitment of these cells. Diversification of the B cell response by multivalent nanoparticle immunogens has broad implications for vaccine design.
Collapse
Affiliation(s)
- Sebastian Ols
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Klara Lenart
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Rodrigo Arcoverde Cerveira
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marcos C Miranda
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Natalie Brunette
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jana Kochmann
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Martin Corcoran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Rebecca Skotheim
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Annika Philomin
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Alberto Cagigi
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Brooke Fiala
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Samuel Wrenn
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Jessica Marcandalli
- Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Fredrika Hellgren
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Elizabeth A Thompson
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Ang Lin
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Florian Gegenfurtner
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Azad Kumar
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Man Chen
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ganesh E Phad
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; Università della Svizzera italiana, Faculty of Biomedical Sciences, Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Laurent Perez
- University of Lausanne (UNIL), Lausanne University Hospital (CHUV), Department of Medicine, Service of Immunology and Allergy, and Center for Human Immunology (CHIL), Lausanne, Switzerland
| | - Andrew J Borst
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | | | - Tracy J Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Neil P King
- Department of Biochemistry, University of Washington, Seattle, WA, USA; Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Karin Loré
- Division of Immunology & Allergy, Department of Medicine Solna, Karolinska Institutet, and Karolinska University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
50
|
Jacobs L, Stobbelaar K, Heykers A, Cos P, Delputte P. Subtractive Immunization as a Method to Develop Respiratory Syncytial Virus (RSV)-Specific Monoclonal Antibodies. Antibodies (Basel) 2023; 12:62. [PMID: 37873859 PMCID: PMC10594476 DOI: 10.3390/antib12040062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/15/2023] [Accepted: 09/21/2023] [Indexed: 10/25/2023] Open
Abstract
Respiratory Syncytial Virus (RSV) is a significant cause of lower respiratory tract infections in the young, the elderly, and in immunodeficient patients. As such, the virus represents an important cause of morbidity and mortality worldwide. Development of monoclonal antibodies against RSV has resulted in a commercial prophylaxis, palivizumab (Synagis®), and different antibodies that have improved our understanding of the structure of the viral proteins. In this study, a different immunization technique, subtractive immunization, was evaluated for its applicability to develop RSV-specific antibodies. One hybridoma which produced antibodies with the strongest staining of RSV infected cells, ATAC-0025, was selected for further characterization. This antibody belongs to the IgG1 class, has neutralizing capacity and recognizes the envelope F-protein. The antibody has a broad reactivity against a range of RSV reference strains and clinical isolates.
Collapse
Affiliation(s)
- Lotte Jacobs
- Laboratory for Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (L.J.); (K.S.); (A.H.); (P.C.)
| | - Kim Stobbelaar
- Laboratory for Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (L.J.); (K.S.); (A.H.); (P.C.)
- Pediatrics Department, Antwerp University Hospital (UZA), Wilrijkstraat 10, 2650 Edegem, Belgium
| | - Annick Heykers
- Laboratory for Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (L.J.); (K.S.); (A.H.); (P.C.)
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (L.J.); (K.S.); (A.H.); (P.C.)
| | - Peter Delputte
- Laboratory for Microbiology, Parasitology and Hygiene, Infla-Med Centre of Excellence, University of Antwerp (UA), Universiteitsplein 1 S.7, 2610 Antwerp, Belgium; (L.J.); (K.S.); (A.H.); (P.C.)
| |
Collapse
|