1
|
Lee PI, Huang YC, Chen CJ, Chiu CH, Chen PY, Lu CY, Liu CC, Chiu NC, Chi H, Lin CY, Lee CY, Chiu SN, Jeng MJ, Kuo KC, Tang RB, Huang YF, Pan HH, Cheng MF, Huang LM, Hu YL, Lin TY. Recommendation for immune prophylaxis of respiratory syncytial virus infection in children. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2025; 58:288-293. [PMID: 40032543 DOI: 10.1016/j.jmii.2025.02.007] [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: 10/03/2024] [Revised: 01/26/2025] [Accepted: 02/22/2025] [Indexed: 03/05/2025]
Abstract
Respiratory syncytial virus (RSV) is the most common pathogen for young children hospitalized with bronchiolitis and pneumonia. Most infections occur below 1 year of age, and almost all children have been infected before 2 years of age. Monoclonal antibodies targeting RSV, such as palivizumab and nirsevimab, are accessible for preventing infection. A committee, consisting of experts in infectious diseases, cardiovascular diseases, and neonatal diseases in children, was assembled by the Pediatric Infectious Diseases Society of Taiwan. Collaborating with the Child Health Research Center at the National Health Research Institutes, Taiwan Pediatric Association, and Taiwan Society of Neonatology, the committee worked to formulate recommendations for immune prophylaxis against RSV infection in children. Palivizumab is recommended for the prevention of RSV infection in high-risk infants under 1 year old with one of the following (1) premature infants with a gestational age <33 weeks, (2) premature infants with a gestational age <35 weeks with chronic lung disease or (3) infants with hemodynamically significant CHD. Nirsevimab is recommended for the prevention of RSV infection in all infants <12 months. The recommendation is not intended as a sole source of guidance in the prevention of RSV infection in children. The provisions listed in this recommendation are comprehensive suggestions made by pediatric experts in Taiwan based on existing medical evidence. This recommendation should be subject to modification in light of additional medical research findings in the future, and these provisions should not be cited as a basis for dispute resolution.
Collapse
Affiliation(s)
- Ping-Ing Lee
- Department of Pediatrics, National Taiwan University Children's Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.
| | - Yhu-Chering Huang
- Division of Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taiwan
| | - Chih-Jung Chen
- Division of Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taiwan
| | - Cheng-Hsun Chiu
- Division of Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taiwan
| | - Po-Yen Chen
- Section of Pediatric Infectious Diseases, Department of Pediatrics, Taichung Veterans General Hospital, Taiwan
| | - Chun-Yi Lu
- Department of Pediatrics, National Taiwan University Children's Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Chuan Liu
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Taiwan
| | - Nan-Chang Chiu
- Department of Pediatrics, MacKay Children's Hospital, Taiwan, Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Hsin Chi
- Department of Pediatrics, MacKay Children's Hospital, Taiwan, Department of Medicine, MacKay Medical College, New Taipei City, Taiwan
| | - Chien-Yu Lin
- Hsinchu Municipal MacKay Children's Hospital, Hsinchu City, Taiwan
| | - Chun Yi Lee
- Department of Pediatrics, Chang Bing Show Chwan Memorial Hospital, Taiwan
| | - Shuenn-Nan Chiu
- Department of Pediatrics, National Taiwan University Children's Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Mei-Jy Jeng
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, Neonatal Medical Care Center, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Kuang-Che Kuo
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ren-Bin Tang
- Department of Pediatrics, Cheng-hsin General Hospital, Taipei, Taiwan
| | - Yung-Feng Huang
- Department of Pediatrics, Fu Jen Catholic University Hospital, Taiwan
| | - Hui-Hsien Pan
- Section of Pediatric Infectious Diseases, Department of Pediatrics, Taichung Veterans General Hospital, Taiwan
| | - Ming-Fang Cheng
- Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung Taiwan, Taiwan
| | - Li-Min Huang
- Department of Pediatrics, National Taiwan University Children's Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ya-Li Hu
- Department of Pediatrics, Cathay General Hospital, Taipei, Taiwan
| | - Tzou-Yien Lin
- Division of Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Chang Gung University, College of Medicine, Taiwan
| |
Collapse
|
2
|
Lin M, Yin Y, Zhao X, Wang C, Zhu X, Zhan L, Chen L, Wang S, Lin X, Zhang J, Xia N, Zheng Z. A truncated pre-F protein mRNA vaccine elicits an enhanced immune response and protection against respiratory syncytial virus. Nat Commun 2025; 16:1386. [PMID: 39910047 PMCID: PMC11799228 DOI: 10.1038/s41467-025-56302-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
The Food and Drug Administration (FDA) has approved vaccines designed by GSK, Pfizer and Moderna to protect high-risk populations against respiratory syncytial virus (RSV). These vaccines employ the pre-fusion F (pre-F) protein as the immunogen. In this study, we explored an mRNA vaccine based on a modified pre-F protein called LC2DM-lipid nanoparticle (LC2DM-LNP). This vaccine features a truncated version of the pre-F protein that is anchored to the cell membrane. Our experiments in young and old female mice revealed that the LC2DM-LNP vaccine elicited robust neutralizing antibody titers. Moreover, LC2DM-LNP prompted a Th1-skewed T-cell immune response in female rodent models. Female cotton rats immunized with LC2DM-LNP demonstrated strong immunity to RSV, without signs of vaccine-enhanced respiratory disease (VERD), even in cases of breakthrough infection. Importantly, when administered to pregnant female cotton rats, LC2DM-LNP ensured the transfer of pre-F-specific antibodies to the offspring and provided protection against RSV without increasing lung inflammation. Our findings suggest that LC2DM-LNP could serve as an alternative RSV vaccine candidate for high-risk groups.
Collapse
Affiliation(s)
- Min Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Yifan Yin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Xiaomeng Zhao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Chen Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Xueqing Zhu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Letao Zhan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Li Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Siling Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Xue Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China
| | - Jun Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China.
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China.
| | - Zizheng Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian Province, PR China.
- National Institute of Diagnostics and Vaccine Development in Infectious Diseases, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, National Innovation Platform for Industry-Education Integration in Vaccine Research, Xiamen University, Xiamen, Fujian Province, PR China.
| |
Collapse
|
3
|
Zhang W, Lin X, Li ZY, Zhang LJ, Chen L, Sun YP, Si JY, Zhao M, Wu GH, Zhan LT, Yang KY, You RL, Wang YB, Xia NS, Zheng ZZ. Novel intercellular spread mode of respiratory syncytial virus contributes to neutralization escape. Antiviral Res 2024; 231:106023. [PMID: 39489301 DOI: 10.1016/j.antiviral.2024.106023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/05/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Developing widely used respiratory syncytial virus (RSV) vaccines remains a significant challenge, despite the recent authorization of two pre-F vaccines for elderly adults. Previous reports have suggested that even when vaccine-induced immunity generates high titers of potent neutralizing antibodies targeting the pre-F protein, it may not fully inhibit breakthrough of RSV infections. This incomplete inhibition of RSV breakthrough infections can lead to an increased risk of enhanced respiratory disease (ERD) in vaccinated individuals. The reasons why potent neutralizing antibodies cannot fully prevent RSV breakthrough infections are not yet clear. In an attempt to explain this phenomenon, we investigated the effect of potent neutralizing antibodies on the intercellular spread of RSV. Our findings indicated that a specific titer of potent neutralizing antibodies, such as 5C4, could block certain modes of intercellular spread, such as the diffusion of cell-free virions and the delivery of virions through filopodia. However, these antibodies did not fully inhibit the entire process of intercellular spread. Through the use of super-resolution imaging techniques, we observed a novel and efficient spread mode called the transition of viral materials through intercellular nanotubes (TVMIN), independent of virions and insensitive to the presence of antibodies. TVMIN allowed RSV-infected cells to directly transfer viral materials to neighboring cells via intercellular nanotubes that are rich in microfilaments. TVMIN began as early as 5 h post-infection (h.p.i.) and rapidly initiated infection in recipient cells. Our data provided new insights into the intercellular spread of RSV and might help explain the occurrence of breakthrough infections.
Collapse
Affiliation(s)
- Wei Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Xue Lin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Zhi-Yong Li
- The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Lu-Jing Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Li Chen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Yong-Peng Sun
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Jun-Yu Si
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Min Zhao
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Guang-Hua Wu
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Lu-Ting Zhan
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China
| | - Kun-Yu Yang
- The Clinical Laboratory of Xiamen International Travel Healthcare Center, Xiamen, Fujian, China
| | - Rui-Luan You
- The Clinical Laboratory of Xiamen International Travel Healthcare Center, Xiamen, Fujian, China
| | - Ying-Bin Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
| | - Ning-Shao Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China; The Research Unit of Frontier Technology of Structural Vaccinology of Chinese Academy of Medical Sciences, Xiamen, China.
| | - Zi-Zheng Zheng
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang an Biomedicine Laboratory, National Institute of Diagnostics and Vaccine Development in Infectious Diseases, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361102, China.
| |
Collapse
|
4
|
Peng R, Chen C, Chen Q, Zhang Y, Huang R, Zhang Y, Li J. Global progress in clinical research on human respiratory syncytial virus vaccines. Front Microbiol 2024; 15:1457703. [PMID: 39286350 PMCID: PMC11402711 DOI: 10.3389/fmicb.2024.1457703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 08/20/2024] [Indexed: 09/19/2024] Open
Abstract
Human respiratory syncytial virus (hRSV) not only affects newborns but also older adults, contributing to a substantial worldwide burden of disease. However, only three approved hRSV vaccines remain commercially available to date. The development of a safe, practical and broad-spectrum vaccine suitable for all age groups remains extremely challenging. Using five different approaches-live-attenuated, recombinant-vector, subunit, particle-based, and mRNA-nearly 30 hRSV vaccine candidates are currently conducting clinical trials worldwide; moreover, > 30 vaccines are under preclinical evaluation. This review presents a comprehensive overview of these hRSV vaccines along with prospects for the development of infectious disease vaccines in the post-COVID-19 pandemic era.
Collapse
Affiliation(s)
- Ruofan Peng
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenghao Chen
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Qian Chen
- School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Yuwen Zhang
- School of Public Health, Hangzhou Medical College, Hangzhou, China
| | - Renjin Huang
- School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Yanjun Zhang
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| | - Jianhua Li
- Key Laboratory of Public Health Detection and Etiological Research of Zhejiang Province, Department of Microbiology, Zhejiang Provincial Center for Disease Control and Prevention, Hangzhou, China
| |
Collapse
|
5
|
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
|
6
|
Sanchez-Martinez ZV, Alpuche-Lazcano SP, Stuible M, Durocher Y. CHO cells for virus-like particle and subunit vaccine manufacturing. Vaccine 2024; 42:2530-2542. [PMID: 38503664 DOI: 10.1016/j.vaccine.2024.03.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/27/2024] [Accepted: 03/13/2024] [Indexed: 03/21/2024]
Abstract
Chinese Hamster Ovary (CHO) cells, employed primarily for manufacturing monoclonal antibodies and other recombinant protein (r-protein) therapeutics, are emerging as a promising host for vaccine antigen production. This is exemplified by the recently approved CHO cell-derived subunit vaccines (SUV) against respiratory syncytial virus (RSV) and varicella-zoster virus (VZV), as well as the enveloped virus-like particle (eVLP) vaccine against hepatitis B virus (HBV). Here, we summarize the design, production, and immunogenicity features of these vaccine and review the most recent progress of other CHO-derived vaccines in pre-clinical and clinical development. We also discuss the challenges associated with vaccine production in CHO cells, with a focus on ensuring viral clearance for eVLP products.
Collapse
Affiliation(s)
- Zalma V Sanchez-Martinez
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Sergio P Alpuche-Lazcano
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Matthew Stuible
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council of Canada, Montreal, QC H4P 2R2, Canada; Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada; PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université du Québec à Montréal, 201 Avenue du Président Kennedy, Montréal, QC H2X 3Y7, Canada.
| |
Collapse
|
7
|
Rzymski P, Gwenzi W. Respiratory syncytial virus immunoprophylaxis: Novel opportunities and a call for equity. J Med Virol 2024; 96:e29453. [PMID: 38305000 DOI: 10.1002/jmv.29453] [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: 10/25/2023] [Revised: 12/20/2023] [Accepted: 01/24/2024] [Indexed: 02/03/2024]
Abstract
With the approval of the first vaccines against respiratory syncytial virus (RSV) and a novel RSV-neutralizing antibody, 2023 has been perceived as a game-changing year in preventing severe outcomes of RSV infections in infants and the elderly. However, the costs of these pharmaceuticals are high, while RSV disproportionately impacts populations of low-to-middle-income regions, which may continue to suffer from a lack of pharmaceutical measures for RSV prevention under health and socioeconomic disparities. This paper presents an overview of the characteristics, clinical results, and approval status of various RSV vaccines and anti-RSV antibodies. It posits that wealthy nations cannot monopolize RSV immunoprophylaxis and should work jointly to make it available to lower-income countries. An approach toward RSV immunoprophylaxis equity based on five points is offered: (1) integration of RSV vaccines and antibodies into the existing global humanitarian distribution systems, (2) using affordable RSV vaccine pricing models, (3) enforcing equity as a part of national and global public health strategy, (4) implementing equitable allocation frameworks for RSV immunoprophylaxis, and (5) promoting local manufacturing. Such a plan needs to be put into action as soon as possible to avoid delays in serving the populations with the highest needs related to RSV burden.
Collapse
Affiliation(s)
- Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznań, Poland
| | - Willis Gwenzi
- Biosystems and Environmental Enginering Research Group, Harare, Zimbabwe
- Alexander von Humboldt Fellow and Guest Professor at Grassland Science and Renewable Plant Resources, Faculty of Organic Agricultural Sciences, Universität Kassel, Witzenhausen, Germany
| |
Collapse
|
8
|
Guo F, Wei J, Song Y, Li B, Qian Z, Wang X, Wang H, Xu T. Immunological effects of the PE/PPE family proteins of Mycobacterium tuberculosis and related vaccines. Front Immunol 2023; 14:1255920. [PMID: 37841250 PMCID: PMC10569470 DOI: 10.3389/fimmu.2023.1255920] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 08/25/2023] [Indexed: 10/17/2023] Open
Abstract
Tuberculosis (TB) is a chronic infectious disease caused by Mycobacterium tuberculosis (Mtb), and its incidence and mortality are increasing. The BCG vaccine was developed in the early 20th century. As the most widely administered vaccine in the world, approximately 100 million newborns are vaccinated with BCG every year, which has saved tens of millions of lives. However, due to differences in region and race, the average protective rate of BCG in preventing tuberculosis in children is still not high in some areas. Moreover, because the immune memory induced by BCG will weaken with the increase of age, it is slightly inferior in preventing adult tuberculosis, and BCG revaccination cannot reduce the incidence of tuberculosis again. Research on the mechanism of Mtb and the development of new vaccines against TB are the main strategies for preventing and treating TB. In recent years, Pro-Glu motif-containing (PE) and Pro-Pro-Glu motif-containing (PPE) family proteins have been found to have an increasingly important role in the pathogenesis and chronic protracted infection observed in TB. The development and clinical trials of vaccines based on Mtb antigens are in progress. Herein, we review the immunological effects of PE/PPE proteins and the development of common PE/PPE vaccines.
Collapse
Affiliation(s)
- Fangzheng Guo
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
| | - Jing Wei
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
| | - Yamin Song
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
| | - Baiqing Li
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical College, Bengbu, China
| | - Zhongqing Qian
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical College, Bengbu, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Bengbu Medical College, Bengbu, China
| | - Hongtao Wang
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
- Department of Immunology, School of Laboratory, Bengbu Medical College, Bengbu, China
| | - Tao Xu
- Research Center of Laboratory, Bengbu Medical College, Bengbu, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases , Bengbu Medical College, Bengbu, China
- Department of Clinical Laboratory, School of Laboratory, Bengbu Medical College, Bengbu, China
| |
Collapse
|
9
|
Chen F, Park HR, Ji HJ, Kwon Y, Kim MK, Song JY, Ahn KB, Seo HS. Gamma Irradiation-Inactivated Respiratory Syncytial Virus Vaccine Provides Protection but Exacerbates Pulmonary Inflammation by Switching from Prefusion to Postfusion F Protein. Microbiol Spectr 2023; 11:e0135823. [PMID: 37272801 PMCID: PMC10434263 DOI: 10.1128/spectrum.01358-23] [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: 04/22/2023] [Accepted: 04/30/2023] [Indexed: 06/06/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a common respiratory pathogen that causes lower respiratory diseases among infants and elderly people. Moreover, formalin-inactivated RSV (FI-RSV) vaccine induces serious enhanced respiratory disease (ERD). Radiation has been investigated as an alternative approach for producing inactivated or live-attenuated vaccines, which enhance the antigenicity and heterogeneous protective effects of vaccines compared with conventional formalin inactivation. In this study, we developed an RSV vaccine using gamma irradiation and analyzed its efficacy against RSV vaccine-induced ERD in a mouse model. Although gamma irradiation-inactivated RSV (RI-RSV) carbonylation was lower than FI-RSV carbonylation and RI-RSV showed a significant antibody production and viral clearance, RI-RSV caused more obvious body weight loss, pulmonary eosinophil infiltration, and pulmonary mucus secretion. Further, the conversion of prefusion F (pre-F) to postfusion F (post-F) was significant for both RI-RSV and FI-RSV, while that of RI-RSV was significantly higher than that of FI-RSV. We found that the conversion from pre- to post-F during radiation was caused by radiation-induced reactive oxygen species. Although we could not propose an effective RSV vaccine manufacturing method, we found that ERD was induced by RSV vaccine by various biochemical effects that affect antigen modification during RSV vaccine manufacturing, rather than simply by the combination of formalin and alum. Therefore, these biochemical actions should be considered in future developments of RSV vaccine. IMPORTANCE Radiation inactivation for viral vaccine production has been known to elicit a better immune response than other inactivation methods due to less surface protein damage. However, we found in this study that radiation-inactivated RSV (RI-RSV) vaccine induced a level of immune response similar to that induced by formalin-inactivated RSV (FI-RSV). Although RI-RSV vaccine showed less carbonylation than FI-RSV, it induced more conformational changes from pre-F to post-F due to the gamma radiation-induced reactive oxygen species response, which may be a key factor in RI-RSV-induced ERD. Therefore, ERD induced by RSV vaccine may be due to pre-F to post-F denaturation by random protein modifications caused by external stress. Our findings provide new ideas for inactivated vaccines for RSV and other viruses and confirm the importance of pre-F in RSV vaccines.
Collapse
Affiliation(s)
- Fengjia Chen
- Accelerator Radioisotope Research Section, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
| | - Hae-Ran Park
- Accelerator Radioisotope Research Section, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
| | - Hyun Jung Ji
- Accelerator Radioisotope Research Section, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
- Department of Oral Microbiology and Immunology, DRI, and BK21 Plus Program, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Yeongkag Kwon
- Accelerator Radioisotope Research Section, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
| | - Min-Kyu Kim
- Accelerator Radioisotope Research Section, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
| | - Joon Young Song
- Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Ki Bum Ahn
- Accelerator Radioisotope Research Section, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
- Animal Production and Health Laboratory, Joint FAO/IAEA Centre for Nuclear Applications in Food and Agriculture, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Seibersdorf, Austria
| | - Ho Seong Seo
- Accelerator Radioisotope Research Section, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup, Republic of Korea
- Department of Radiation Science, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
10
|
Blanco JCG, Cullen LM, Kamali A, Sylla FYD, Boukhvalova MS, Morrison TG. Development of Respiratory Syncytial Virus Vaccine Candidates for the Elderly. Viruses 2023; 15:1305. [PMID: 37376605 PMCID: PMC10304043 DOI: 10.3390/v15061305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a significant threat to elderly populations and repeated infections that occur throughout life are poorly protective. To assess the role of prior RSV infections as well as elderly immune senescence on vaccine efficacy, we compared immune responses after virus-like particle (VLP) immunization of elderly cotton rats and young cotton rats, both previously RSV infected, in order to mimic the human population. We show that immunization of RSV-experienced young or elderly animals resulted in the same levels of anti-pre-F IgG, anti-G IgG, neutralizing antibody titers, and protection from challenge indicating that the delivery of F and G proteins in a VLP is equally effective in activation of protective responses in both elderly and young populations. Our results suggest that F and G protein-containing VLPs induce anti-RSV memory established in prior RSV infections equally well in both young and elderly animals and thus can be an effective vaccine for the elderly.
Collapse
Affiliation(s)
| | - Lori M. Cullen
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| | - Arash Kamali
- Sigmovir Biosystems Inc., Rockville, MD 20850, USA
| | | | | | - Trudy G. Morrison
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01655, USA
| |
Collapse
|
11
|
Rzymski P, Szuster-Ciesielska A, Dzieciątkowski T, Gwenzi W, Fal A. mRNA vaccines: The future of prevention of viral infections? J Med Virol 2023; 95:e28572. [PMID: 36762592 DOI: 10.1002/jmv.28572] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023]
Abstract
Messenger RNA (mRNA) vaccines against COVID-19 are the first authorized biological preparations developed using this platform. During the pandemic, their administration has been proven to be a life-saving intervention. Here, we review the main advantages of using mRNA vaccines, identify further technological challenges to be met during the development of the mRNA platform, and provide an update on the clinical progress on leading mRNA vaccine candidates against different viruses that include influenza viruses, human immunodeficiency virus 1, respiratory syncytial virus, Nipah virus, Zika virus, human cytomegalovirus, and Epstein-Barr virus. The prospects and challenges of manufacturing mRNA vaccines in low-income countries are also discussed. The ongoing interest and research in mRNA technology are likely to overcome some existing challenges for this technology (e.g., related to storage conditions and immunogenicity of some components of lipid nanoparticles) and enhance the portfolio of vaccines against diseases for which classical formulations are already authorized. It may also open novel pathways of protection against infections and their consequences for which no safe and efficient immunization methods are currently available.
Collapse
Affiliation(s)
- Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznań, Poland.,Integrated Science Association (ISA), Universal Scientific Education and Research Network (USERN), Poznań, Poland
| | - Agnieszka Szuster-Ciesielska
- Department of Virology and Immunology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | | | - Willis Gwenzi
- Alexander von Humboldt Fellow & Guest Professor, Grassland Science and Renewable Plant Resources, Faculty of Organic Agricultural Sciences, Universität Kassel, Witzenhausen, Germany.,Alexander von Humboldt Fellow & Guest Professor, Leibniz Institute for Agricultural Engineering and Bioeconomy (ATB), Potsdam, Germany
| | - Andrzej Fal
- Collegium Medicum, Warsaw Faculty of Medicine, Cardinal Stefan Wyszynski University, Warsaw, Poland.,Department of Public Health, Wrocław Medical University, Wrocław, Poland
| |
Collapse
|
12
|
Powell TJ, Jacobs A, Tang J, Cardenas E, Palath N, Daniels J, Boyd JG, Bergeron HC, Jorquera PA, Tripp RA. Microparticle RSV Vaccines Presenting the G Protein CX3C Chemokine Motif in the Context of TLR Signaling Induce Protective Th1 Immune Responses and Prevent Pulmonary Eosinophilia Post-Challenge. Vaccines (Basel) 2022; 10:vaccines10122078. [PMID: 36560488 PMCID: PMC9785538 DOI: 10.3390/vaccines10122078] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Layer-by-layer microparticle (LbL-MP) fabrication was used to produce synthetic vaccines presenting a fusion peptide containing RSV G protein CX3C chemokine motif and a CD8 epitope of the RSV matrix protein 2 (GM2) with or without a covalently linked TLR2 agonist (Pam3.GM2). Immunization of BALB/c mice with either GM2 or Pam3.GM2 LbL-MP in the absence of adjuvant elicited G-specific antibody responses and M2-specific CD8+ T-cell responses. Following challenge with RSV, mice immunized with the GM2 LbL-MP vaccine developed a Th2-biased immune response in the lungs with elevated levels of IL-4, IL-5, IL-13, and eotaxin in the bronchoalveolar lavage (BAL) fluid and a pulmonary influx of eosinophils. By comparison, mice immunized with the Pam3.GM2 LbL-MP vaccine had considerably lower to non-detectable levels of the Th2 cytokines and chemokines and very low numbers of eosinophils in the BAL fluid post-RSV challenge. In addition, mice immunized with the Pam3.GM2 LbL-MP also had higher levels of RSV G-specific IgG2a and IgG2b in the post-challenge BAL fluid compared to those immunized with the GM2 LbL-MP vaccine. While both candidates protected mice from infection following challenge, as evidenced by the reduction or elimination of RSV plaques, the inclusion of the TLR2 agonist yielded a more potent antibody response, greater protection, and a clear shift away from Th2/eosinophil responses. Since the failure of formalin-inactivated RSV (FI-RSV) vaccines tested in the 1960s has been hypothesized to be partly due to the ablation of host TLR engagement by the vaccine and inappropriate Th2 responses upon subsequent viral infection, these findings stress the importance of appropriate engagement of the innate immune response during initial exposure to RSV G CX3C.
Collapse
Affiliation(s)
- Thomas J. Powell
- Artificial Cell Technologies, 5 Science Park, Suite 13, New Haven, CT 06511, USA
- Correspondence:
| | - Andrea Jacobs
- Artificial Cell Technologies, 5 Science Park, Suite 13, New Haven, CT 06511, USA
| | - Jie Tang
- Artificial Cell Technologies, 5 Science Park, Suite 13, New Haven, CT 06511, USA
| | - Edwin Cardenas
- Artificial Cell Technologies, 5 Science Park, Suite 13, New Haven, CT 06511, USA
| | - Naveen Palath
- Artificial Cell Technologies, 5 Science Park, Suite 13, New Haven, CT 06511, USA
| | - Jennifer Daniels
- Artificial Cell Technologies, 5 Science Park, Suite 13, New Haven, CT 06511, USA
| | - James G. Boyd
- Artificial Cell Technologies, 5 Science Park, Suite 13, New Haven, CT 06511, USA
| | - Harrison C. Bergeron
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Patricia A. Jorquera
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
13
|
Soto JA, Galvez NMS, Rivera DB, Díaz FE, Riedel CA, Bueno SM, Kalergis AM. From animal studies into clinical trials: the relevance of animal models to develop vaccines and therapies to reduce disease severity and prevent hRSV infection. Expert Opin Drug Discov 2022; 17:1237-1259. [PMID: 36093605 DOI: 10.1080/17460441.2022.2123468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Human respiratory syncytial virus (hRSV) is an important cause of lower respiratory tract infections in the pediatric and the geriatric population worldwide. There is a substantial economic burden resulting from hRSV disease during winter. Although no vaccines have been approved for human use, prophylactic therapies are available for high-risk populations. Choosing the proper animal models to evaluate different vaccine prototypes or pharmacological treatments is essential for developing efficient therapies against hRSV. AREAS COVERED This article describes the relevance of using different animal models to evaluate the effect of antiviral drugs, pharmacological molecules, vaccine prototypes, and antibodies in the protection against hRSV. The animal models covered are rodents, mustelids, bovines, and nonhuman primates. Animals included were chosen based on the available literature and their role in the development of the drugs discussed in this manuscript. EXPERT OPINION Choosing the correct animal model is critical for exploring and testing treatments that could decrease the impact of hRSV in high-risk populations. Mice will continue to be the most used preclinical model to evaluate this. However, researchers must also explore the use of other models such as nonhuman primates, as they are more similar to humans, prior to escalating into clinical trials.
Collapse
Affiliation(s)
- J A Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - N M S Galvez
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - D B Rivera
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - F E Díaz
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - C A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - S M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - A M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
14
|
Eichinger KM, Kosanovich JL, Perkins T, Oury TD, Petrovsky N, Marshall CP, Yondola MA, Empey KM. Prior respiratory syncytial virus infection reduces vaccine-mediated Th2-skewed immunity, but retains enhanced RSV F-specific CD8 T cell responses elicited by a Th1-skewing vaccine formulation. Front Immunol 2022; 13:1025341. [PMID: 36268035 PMCID: PMC9577258 DOI: 10.3389/fimmu.2022.1025341] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Respiratory syncytial virus (RSV) remains the most common cause of lower respiratory tract infections in children worldwide. Development of a vaccine has been hindered due the risk of enhanced respiratory disease (ERD) following natural RSV exposure and the young age (<6 months) at which children would require protection. Risk factors linked to the development of ERD include poorly neutralizing antibody, seronegative status (never been exposed to RSV), and a Th2-type immune response. Stabilization of the more antigenic prefusion F protein (PreF) has reinvigorated hope for a protective RSV vaccine that elicits potent neutralizing antibody. While anecdotal evidence suggests that children and adults previously exposed to RSV (seropositive) are not at risk for developing vaccine associated ERD, differences in host immune responses in seropositive and seronegative individuals that may protect against ERD remain unclear. It is also unclear if vaccine formulations that skew towards Th1- versus Th2-type immune responses increase pathology or provide greater protection in seropositive individuals. Therefore, the goal of this work was to compare the host immune response to a stabilized prefusion RSV antigen formulated alone or with Th1 or Th2 skewing adjuvants in seronegative and seropositive BALB/c mice. We have developed a novel BALB/c mouse model whereby mice are first infected with RSV (seropositive) and then vaccinated during pregnancy to recapitulate maternal immunization strategies. Results of these studies show that prior RSV infection mitigates vaccine-mediated skewing by Th1- and Th2-polarizing adjuvants that was observed in seronegative animals. Moreover, vaccination with PreF plus the Th1-skewing adjuvant, Advax, increased RSV F85-93-specific CD8 T cells in both seronegative and seropositive dams. These data demonstrate the importance of utilizing seropositive animals in preclinical vaccine studies to assess both the safety and efficacy of candidate RSV vaccines.
Collapse
Affiliation(s)
- Katherine M. Eichinger
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jessica L. Kosanovich
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Timothy N. Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburg, Pittsburgh, PA, United States
| | - Tim D. Oury
- Department of Pathology, University of Pittsburgh School of Medicine, University of Pittsburg, Pittsburgh, PA, United States
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Bedford Park, SA, Australia
- College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | | | | | - Kerry M. Empey
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
- Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
15
|
Chen JW, Yang L, Santos C, Hassan SA, Collins PL, Buchholz UJ, Le Nouën C. Reversion mutations in phosphoprotein P of a codon-pair-deoptimized human respiratory syncytial virus confer increased transcription, immunogenicity, and genetic stability without loss of attenuation. PLoS Pathog 2021; 17:e1010191. [PMID: 34965283 PMCID: PMC8751989 DOI: 10.1371/journal.ppat.1010191] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 01/11/2022] [Accepted: 12/09/2021] [Indexed: 11/18/2022] Open
Abstract
Recoding viral genomes by introducing numerous synonymous nucleotide substitutions that create suboptimal codon pairs provides new live-attenuated vaccine candidates. Because recoding typically involves a large number of nucleotide substitutions, the risk of de-attenuation is presumed to be low. However, this has not been thoroughly studied. We previously generated human respiratory syncytial virus (RSV) in which the NS1, NS2, N, P, M and SH ORFs were codon-pair deoptimized (CPD) by 695 synonymous nucleotide changes (Min A virus). Min A exhibited a global reduction in transcription and protein synthesis, was restricted for replication in vitro and in vivo, and exhibited moderate temperature sensitivity. Here, we show that under selective pressure by serial passage at progressively increasing temperatures, Min A regained replication fitness and lost its temperature sensitivity. Whole-genome deep sequencing identified numerous missense mutations in several genes, in particular ones accumulating between codons 25 and 34 of the phosphoprotein (P), a polymerase cofactor and chaperone. When re-introduced into Min A, these P mutations restored viral transcription to wt level, resulting in increased protein expression and RNA replication. Molecular dynamic simulations suggested that these P mutations increased the flexibility of the N-terminal domain of P, which might facilitate its interaction with the nucleoprotein N, and increase the functional efficiency of the RSV transcription/replication complex. Finally, we evaluated the effect of the P mutations on Min A replication and immunogenicity in hamsters. Mutation P[F28V] paradoxically reduced Min A replication but not its immunogenicity. The further addition of one missense mutation each in M and L generated a version of Min A with increased genetic stability. Thus, this study provides further insight into the adaptability of large-scale recoded RNA viruses under selective pressure and identified an improved CPD RSV vaccine candidate. Synonymous recoding of viral genomes by codon-pair deoptimization (CPD) generates live-attenuated vaccines presumed to be genetically stable due to the high number of nucleotide substitutions. However, their actual genetic stability under selective pressure was largely unknown. In a recoded human respiratory syncytial virus (RSV) mutant called Min A, six of 11 ORFs were CPD, reducing protein expression and inducing moderate temperature sensitivity and attenuation. When passaged in vitro under selective pressure, Min A lost its temperature-sensitive phenotype and regained fitness by the acquisition of numerous mutations, in particular missense mutations in the viral phosphoprotein (P), a polymerase cofactor and a chaperone for soluble nucleoprotein. These P mutations increased RSV gene transcription globally, thereby increasing RSV protein expression, RNA replication, and virus particle production. Thus, the P mutations increased the efficiency of the RSV transcription/replication complex, compensating for the reduced protein expression due to CPD. In addition, introduction of the P mutations into Min A generated a live-attenuated vaccine candidate with increased genetic stability. Surprisingly, this vaccine candidate exhibited increased attenuation and, paradoxically, exhibited increased immunogenicity per plaque-forming unit in hamsters. This study provides insights into the adaptability of large-scale recoded RNA viruses and identified an improved CPD RSV vaccine candidate.
Collapse
Affiliation(s)
- Jessica W. Chen
- RNA Viruses Section, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Lijuan Yang
- RNA Viruses Section, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Celia Santos
- RNA Viruses Section, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Sergio A. Hassan
- Bioinformatics and Computational Biosciences Branch, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Peter L. Collins
- RNA Viruses Section, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Ursula J. Buchholz
- RNA Viruses Section, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
| | - Cyril Le Nouën
- RNA Viruses Section, Laboratory of Infectious Diseases, NIAID, NIH, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
16
|
Bergeron HC, Tripp RA. Immunopathology of RSV: An Updated Review. Viruses 2021; 13:2478. [PMID: 34960746 PMCID: PMC8703574 DOI: 10.3390/v13122478] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022] Open
Abstract
RSV is a leading cause of respiratory tract disease in infants and the elderly. RSV has limited therapeutic interventions and no FDA-approved vaccine. Gaps in our understanding of virus-host interactions and immunity contribute to the lack of biological countermeasures. This review updates the current understanding of RSV immunity and immunopathology with a focus on interferon responses, animal modeling, and correlates of protection.
Collapse
Affiliation(s)
| | - Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
17
|
Intranasal vaccination with a recombinant protein CTA1-DD-RBF protects mice against hRSV infection. Sci Rep 2021; 11:18641. [PMID: 34545126 PMCID: PMC8452643 DOI: 10.1038/s41598-021-97535-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/04/2021] [Indexed: 12/05/2022] Open
Abstract
Human respiratory syncytial virus (hRSV) infection is a major pediatric health concern worldwide. Despite more than half a century of efforts, there is still no commercially available vaccine. In this study, we constructed and purified the recombinant protein CTA1-DD-RBF composed of a CTA1-DD mucosal adjuvant and prefusion F protein (RBF) using Escherichia coli BL21 cells. We studied the immunogenicity of CTA1-DD-RBF in mice. Intranasal immunization with CTA1-DD-RBF stimulated hRSV F-specific IgG1, IgG2a, sIgA, and neutralizing antibodies as well as T cell immunity without inducing lung immunopathology upon hRSV challenge. Moreover, the protective immunity of CTA1-DD-RBF was superior to that of the RBF protein, as confirmed by the assessment of serum-neutralizing activity and viral clearance after challenge. Compared to formalin-inactivated hRSV (FI-RSV), intranasal immunization with CTA1-DD-RBF induced a Th1 immune response. In summary, intranasal immunization with CTA1-DD-RBF is safe and effective in mice. Therefore, CTA1-DD-RBF represents a potential mucosal vaccine candidate for the prevention of human infection with hRSV.
Collapse
|
18
|
Zuniga A, Rassek O, Vrohlings M, Marrero-Nodarse A, Moehle K, Robinson JA, Ghasparian A. An epitope-specific chemically defined nanoparticle vaccine for respiratory syncytial virus. NPJ Vaccines 2021; 6:85. [PMID: 34145291 PMCID: PMC8213762 DOI: 10.1038/s41541-021-00347-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 06/02/2021] [Indexed: 12/05/2022] Open
Abstract
Respiratory syncytial virus (RSV) can cause severe respiratory disease in humans, particularly in infants and the elderly. However, attempts to develop a safe and effective vaccine have so far been unsuccessful. Atomic-level structures of epitopes targeted by RSV-neutralizing antibodies are now known, including that bound by Motavizumab and its clinically used progenitor Palivizumab. We developed a chemically defined approach to RSV vaccine design, that allows control of both immunogenicity and safety features of the vaccine. Structure-guided antigen design and a synthetic nanoparticle delivery platform led to a vaccine candidate that elicits high titers of palivizumab-like, epitope-specific neutralizing antibodies. The vaccine protects preclinical animal models from RSV infection and lung pathology typical of vaccine-derived disease enhancement. The results suggest that the development of a safe and effective synthetic epitope-specific RSV vaccine may be feasible by combining this conformationally stabilized peptide and synthetic nanoparticle delivery system.
Collapse
Affiliation(s)
- Armando Zuniga
- Virometix AG, Schlieren, Switzerland
- Shape Biopharmaceuticals Inc, Cambridge, MA, USA
| | | | - Melissa Vrohlings
- Virometix AG, Schlieren, Switzerland
- CDR-Life, Schlieren, Switzerland
| | | | - Kerstin Moehle
- Chemistry Department, University of Zurich, Zurich, Switzerland
| | - John A Robinson
- Chemistry Department, University of Zurich, Zurich, Switzerland.
| | - Arin Ghasparian
- Virometix AG, Schlieren, Switzerland.
- Shape Biopharmaceuticals Inc, Cambridge, MA, USA.
| |
Collapse
|
19
|
Park Y, Kim KH, Lee Y, Lee YT, Kang SM, Ko EJ. Natural killer cells contribute to enhanced respiratory disease after oil-in-water emulsion adjuvanted vaccination against respiratory syncytial virus and infection. Hum Vaccin Immunother 2021; 17:3806-3817. [PMID: 33877948 DOI: 10.1080/21645515.2021.1915039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection caused severe acute respiratory disease in children and the elderly. There is no licensed vaccine. It has been a challenging problem to avoid vaccine enhanced respiratory disease in developing a safe and effective RSV vaccine. Here, we investigated the impact of MF59-like oil-in-water emulsion adjuvant Addavax on the vaccine efficacy of inactivated split RSV (sRSV) and the roles of natural killer (NK) cells in enhanced respiratory disease in sRSV vaccinated mice after RSV infection. Addavax-adjuvanted sRSV vaccination induced higher levels of IgG1 isotype antibodies and more effective lung viral clearance upon RSV infection but promoted enhanced respiratory disease of weight loss, pulmonary inflammation, and NK and NK T (NKT) cell infiltrations in the lungs. Antibody treatment depleting NK cells prior to RSV infection resulted in preventing severe weight loss and histopathology, as well as attenuating infiltration of dendritic cell subsets and TNF-α+ T cells in the lungs. This study demonstrated the impacts of oil-in-water emulsion adjuvant on sRSV vaccination and the potential roles of NK and NKT cells in protection and respiratory disease after adjuvanted RSV vaccination and infection in a mouse model.
Collapse
Affiliation(s)
- Yoonsuh Park
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA.,College of Veterinary Medicine and Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, South Korea
| |
Collapse
|
20
|
Stephens LM, Ross KA, Waldstein KA, Legge KL, McLellan JS, Narasimhan B, Varga SM. Prefusion F-Based Polyanhydride Nanovaccine Induces Both Humoral and Cell-Mediated Immunity Resulting in Long-Lasting Protection against Respiratory Syncytial Virus. THE JOURNAL OF IMMUNOLOGY 2021; 206:2122-2134. [PMID: 33827894 DOI: 10.4049/jimmunol.2100018] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 02/19/2021] [Indexed: 11/19/2022]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of lower respiratory tract infection in both young children and in older adults. Despite the morbidity, mortality, and high economic burden caused by RSV worldwide, no licensed vaccine is currently available. We have developed a novel RSV vaccine composed of a prefusion-stabilized variant of the fusion (F) protein (DS-Cav1) and a CpG oligodeoxynucleotide adjuvant encapsulated within polyanhydride nanoparticles, termed RSVNanoVax. A prime-boost intranasal administration of RSVNanoVax in BALB/c mice significantly alleviated weight loss and pulmonary dysfunction in response to an RSV challenge, with protection maintained up to at least 6 mo postvaccination. In addition, vaccinated mice exhibited rapid viral clearance in the lungs as early as 2 d after RSV infection in both inbred and outbred populations. Vaccination induced tissue-resident memory CD4 and CD8 T cells in the lungs, as well as RSV F-directed neutralizing Abs. Based on the robust immune response elicited and the high level of durable protection observed, our prefusion RSV F nanovaccine is a promising new RSV vaccine candidate.
Collapse
Affiliation(s)
- Laura M Stephens
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA
| | - Kathleen A Ross
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA.,Nanovaccine Institute, Ames, IA
| | - Kody A Waldstein
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA
| | - Kevin L Legge
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA.,Nanovaccine Institute, Ames, IA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA.,Department of Pathology, University of Iowa, Iowa City, IA; and
| | - Jason S McLellan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX
| | - Balaji Narasimhan
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA.,Nanovaccine Institute, Ames, IA
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA; .,Nanovaccine Institute, Ames, IA.,Department of Microbiology and Immunology, University of Iowa, Iowa City, IA.,Department of Pathology, University of Iowa, Iowa City, IA; and
| |
Collapse
|
21
|
Single-Shot Vaccines against Bovine Respiratory Syncytial Virus (BRSV): Comparative Evaluation of Long-Term Protection after Immunization in the Presence of BRSV-Specific Maternal Antibodies. Vaccines (Basel) 2021; 9:vaccines9030236. [PMID: 33803302 PMCID: PMC8001206 DOI: 10.3390/vaccines9030236] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/25/2021] [Accepted: 03/03/2021] [Indexed: 11/28/2022] Open
Abstract
The induction of long-lasting clinical and virological protection is needed for a successful vaccination program against the bovine respiratory syncytial virus (BRSV). In this study, calves with BRSV-specific maternally derived antibodies were vaccinated once, either with (i) a BRSV pre-fusion protein (PreF) and MontanideTM ISA61 VG (ISA61, n = 6), (ii) BRSV lacking the SH gene (ΔSHrBRSV, n = 6), (iii) a commercial vaccine (CV, n = 6), or were injected with ISA61 alone (n = 6). All calves were challenged with BRSV 92 days later and were euthanized 13 days post-infection. Based on clinical, pathological, and proteomic data, all vaccines appeared safe. Compared to the controls, PreF induced the most significant clinical and virological protection post-challenge, followed by ΔSHrBRSV and CV, whereas the protection of PreF-vaccinated calves was correlated with BRSV-specific serum immunoglobulin (Ig)G antibody responses 84 days post-vaccination, and the IgG antibody titers of ΔSHrBRSV- and CV-vaccinated calves did not differ from the controls on this day. Nevertheless, strong anamnestic BRSV- and PreF-specific IgG responses occurred in calves vaccinated with either of the vaccines, following a BRSV challenge. In conclusion, PreF and ΔSHrBRSV are two efficient one-shot candidate vaccines. By inducing a protection for at least three months, they could potentially improve the control of BRSV in calves.
Collapse
|
22
|
Su S, Du L, Jiang S. Learning from the past: development of safe and effective COVID-19 vaccines. Nat Rev Microbiol 2021; 19:211-219. [PMID: 33067570 PMCID: PMC7566580 DOI: 10.1038/s41579-020-00462-y] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2020] [Indexed: 01/29/2023]
Abstract
The rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has elicited an equally rapid response aiming to develop a COVID-19 vaccine. These efforts are encouraging; however, comprehensive efficacy and safety evaluations are essential in the development of a vaccine, and we can learn from previous vaccine development campaigns. In this Perspective, we summarize examples of vaccine-associated disease enhancement in the history of developing vaccines against respiratory syncytial virus, dengue virus, SARS-CoV and Middle East respiratory syndrome coronavirus, which highlight the importance of a robust safety and efficacy profile, and present recommendations for preclinical and clinical evaluation of COVID-19 vaccine candidates as well as for vaccine design and optimization.
Collapse
Affiliation(s)
- Shan Su
- Key Laboratory of Medical Molecular Virology (MOE/MOH/CAM), School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/MOH/CAM), School of Basic Medical Sciences, Fudan University, Shanghai, China.
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA.
| |
Collapse
|
23
|
Karron RA, Luongo C, Mateo JS, Wanionek K, Collins PL, Buchholz UJ. Safety and Immunogenicity of the Respiratory Syncytial Virus Vaccine RSV/ΔNS2/Δ1313/I1314L in RSV-Seronegative Children. J Infect Dis 2021; 222:82-91. [PMID: 31605113 PMCID: PMC7199783 DOI: 10.1093/infdis/jiz408] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 08/08/2019] [Indexed: 01/01/2023] Open
Abstract
Background Respiratory syncytial virus (RSV) is the leading global cause of severe pediatric acute respiratory tract illness, and a vaccine is needed. RSV/ΔNS2/Δ1313/I1314L contains 2 attenuating elements: (1) deletion of the interferon antagonist NS2 gene and (2) deletion of codon 1313 of the RSV polymerase gene and the stabilizing missense mutation I1314L. This live vaccine candidate was temperature-sensitive, genetically stable, replication restricted, and immunogenic in nonhuman primates. Methods A single intranasal dose of RSV/ΔNS2/Δ1313/I1314L was evaluated in a double-blind, placebo-controlled trial (vaccine-placebo ratio, 2:1) at 106 plaque-forming units (PFU) in 15 RSV-seropositive children and at 105 and 106 PFU in 21 and 30 RSV-seronegative children, respectively. Results In RSV-seronegative children, the 105 PFU dose was overattenuated, but the 106 PFU dose was well tolerated, infectious (RSV/ΔNS2/Δ1313/I1314L replication detected in 90% of vaccinees), and immunogenic (geometric mean serum RSV plaque-reduction neutralizing antibody titer, 1:64). After the RSV season, 9 of 20 vaccinees had increases in the RSV titer that were significantly greater than those in 8 of 10 placebo recipients (1:955 vs 1:69, respectively), indicating that the vaccine primed for anamnestic responses after natural RSV exposure. Conclusion Rational design yielded a genetically stable candidate RSV vaccine that is attenuated yet immunogenic in RSV-seronegative children, warranting further evaluation. Clinical Trials Registration NCT01893554.
Collapse
Affiliation(s)
- Ruth A Karron
- Center for Immunization Research, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore
| | - Cindy Luongo
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy, Immunology, and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jocelyn San Mateo
- Center for Immunization Research, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore
| | - Kimberli Wanionek
- Center for Immunization Research, Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore
| | - Peter L Collins
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy, Immunology, and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Ursula J Buchholz
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy, Immunology, and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
24
|
Jenkins T, Wang R, Harder O, Xue M, Chen P, Corry J, Walker C, Teng M, Mejias A, Ramilo O, Niewiesk S, Li J, Peeples ME. A Novel Live Attenuated Respiratory Syncytial Virus Vaccine Candidate with Mutations in the L Protein SAM Binding Site and the G Protein Cleavage Site Is Protective in Cotton Rats and a Rhesus Macaque. J Virol 2021; 95:e01568-20. [PMID: 33177201 PMCID: PMC7925107 DOI: 10.1128/jvi.01568-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/06/2020] [Indexed: 11/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of acute lower respiratory tract infections in children of <5 years of age worldwide, infecting the majority of infants in their first year of life. Despite the widespread impact of this virus, no vaccine is currently available. For more than 50 years, live attenuated vaccines (LAVs) have been shown to protect against other childhood viral infections, offering the advantage of presenting all viral proteins to the immune system for stimulation of both B and T cell responses and memory. The RSV LAV candidate described here, rgRSV-L(G1857A)-G(L208A), contains two modifications: an attenuating mutation in the S-adenosylmethionine (SAM) binding site of the viral mRNA cap methyltransferase (MTase) within the large (L) polymerase protein and a mutation in the attachment (G) glycoprotein that inhibits its cleavage during production in Vero cells, resulting in virus with a "noncleaved G" (ncG). RSV virions containing the ncG have an increased ability to infect primary well-differentiated human bronchial epithelial (HBE) cultures which model the in vivo site of immunization, the ciliated airway epithelium. This RSV LAV candidate is produced efficiently in Vero cells, is highly attenuated in HBE cultures, efficiently induces neutralizing antibodies that are long lasting, and provides protection against an RSV challenge in the cotton rat, without causing enhanced disease. Similar results were obtained in a rhesus macaque.IMPORTANCE Globally, respiratory syncytial virus (RSV) is a major cause of death in children under 1 year of age, yet no vaccine is available. We have generated a novel RSV live attenuated vaccine candidate containing mutations in the L and G proteins. The L polymerase mutation does not inhibit virus yield in Vero cells, the cell type required for vaccine production, but greatly reduces virus spread in human bronchial epithelial (HBE) cultures, a logical in vitro predictor of in vivo attenuation. The G attachment protein mutation reduces its cleavage in Vero cells, thereby increasing vaccine virus yield, making vaccine production more economical. In cotton rats, this RSV vaccine candidate is highly attenuated at a dose of 105 PFU and completely protective following immunization with 500 PFU, 200-fold less than the dose usually used in such studies. It also induced long-lasting antibodies in cotton rats and protected a rhesus macaque from RSV challenge. This mutant virus is an excellent RSV live attenuated vaccine candidate.
Collapse
Affiliation(s)
- Tiffany Jenkins
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Rongzhang Wang
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Olivia Harder
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Miaoge Xue
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Phylip Chen
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jacqueline Corry
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Christopher Walker
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Michael Teng
- Department of Internal Medicine, University of South Florida Morsani College of Medicine, Tampa, Florida, USA
| | - Asuncion Mejias
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Octavio Ramilo
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Stefan Niewiesk
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Jianrong Li
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Mark E Peeples
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
25
|
Liang B, Matsuoka Y, Le Nouën C, Liu X, Herbert R, Swerczek J, Santos C, Paneru M, Collins PL, Buchholz UJ, Munir S. A Parainfluenza Virus Vector Expressing the Respiratory Syncytial Virus (RSV) Prefusion F Protein Is More Effective than RSV for Boosting a Primary Immunization with RSV. J Virol 2020; 95:e01512-20. [PMID: 33115876 PMCID: PMC7944453 DOI: 10.1128/jvi.01512-20] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/20/2020] [Indexed: 12/11/2022] Open
Abstract
Live-attenuated pediatric vaccines for intranasal administration are being developed for human respiratory syncytial virus (RSV), an important worldwide pediatric respiratory pathogen that lacks a licensed vaccine or suitable antiviral drug. We evaluated a prime-boost strategy in which primary immunization with RSV was boosted by secondary immunization with RSV or with a chimeric recombinant bovine/human parainfluenza virus type 3 (rB/HPIV3) vector expressing the RSV fusion F protein. The vector-expressed F protein had been engineered (DS-Cav1 mutations) for increased stability in the highly immunogenic prefusion (pre-F) conformation, with or without replacement of its transmembrane and cytoplasmic tail domains with their counterparts from bovine parainfluenza virus type 3 (BPIV3) F protein to direct incorporation into the vector virion for increased immunogenicity. In hamsters that received a primary infection with RSV, a booster infection with RSV ∼6 weeks later was completely restricted for producing infectious virus but induced a significant increase in the serum RSV-plaque-reduction neutralizing antibody titer (RSV-PRNT). Boosting instead with the rB/HPIV3-RSV-pre-F vectors resulted in efficient replication and induced significantly higher RSV-PRNTs than RSV. In African green monkeys that received a primary infection with RSV, a booster infection with RSV ∼2, ∼6, or ∼15 months later was highly restricted, whereas booster infections with the vectors had robust replication. Compared with RSV, boosts with the vectors induced 7- to 15-fold higher titers of RSV-specific serum antibodies with high neutralizing activity, as well as significantly higher titers of RSV-specific mucosal IgA antibodies. These findings support further development of this heterologous prime-boost strategy.IMPORTANCE Immune responses to RSV in infants can be reduced due to immunological immaturity and immunosuppression by RSV-specific maternal antibodies. In infants and young children, two infections with wild-type RSV typically are needed to achieve the titers of RSV-specific serum antibodies and protection against illness that are observed in adults. Therefore, a boost might substantially improve the performance of live pediatric RSV vaccines presently being developed. Hamsters and African green monkeys received a primary intranasal infection with RSV and were given a boost with RSV or a parainfluenza virus (PIV) vector expressing RSV fusion protein engineered for enhanced immunogenicity. The RSV boost was highly restricted but induced a significant increase in serum RSV-neutralizing antibodies. The PIV vectors replicated efficiently and induced significantly higher antibody responses. The use of an attenuated PIV vector expressing RSV antigen to boost a primary immunization with an attenuated RSV warrants further evaluation.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Chlorocebus aethiops
- Cricetinae
- Immunization, Secondary/methods
- Immunogenicity, Vaccine
- Mutation
- Respiratory Syncytial Virus Infections/prevention & control
- Respiratory Syncytial Virus Vaccines/administration & dosage
- Respiratory Syncytial Virus Vaccines/genetics
- Respiratory Syncytial Virus Vaccines/immunology
- Respiratory Syncytial Virus, Human/genetics
- Respiratory Syncytial Virus, Human/immunology
- Respirovirus/genetics
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/genetics
- Vaccines, Attenuated/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Fusion Proteins/genetics
- Viral Fusion Proteins/immunology
Collapse
Affiliation(s)
- Bo Liang
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Yumiko Matsuoka
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Cyril Le Nouën
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Xueqiao Liu
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Richard Herbert
- Experimental Primate Virology Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Poolesville, Maryland, USA
| | - Joanna Swerczek
- Experimental Primate Virology Section, Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Poolesville, Maryland, USA
| | - Celia Santos
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Monica Paneru
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter L Collins
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ursula J Buchholz
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Shirin Munir
- RNA Viruses Section, Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
26
|
Stephens LM, Varga SM. Nanoparticle vaccines against respiratory syncytial virus. Future Virol 2020; 15:763-778. [PMID: 33343684 PMCID: PMC7737143 DOI: 10.2217/fvl-2020-0174] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022]
Abstract
Respiratory syncytial virus (RSV) is a leading cause of respiratory disease in infants, the elderly and immunocompromised individuals. Despite the global burden, there is no licensed vaccine for RSV. Recent advances in the use of nanoparticle technology have provided new opportunities to address some of the limitations of conventional vaccines. Precise control over particle size and surface properties enhance antigen stability and prolong antigen release. Particle size can also be modified to target specific antigen-presenting cells in order to induce specific types of effector T-cell responses. Numerous nanoparticle-based vaccines are currently being evaluated for RSV including inorganic, polymeric and virus-like particle-based formulations. Here, we review the potential advantages of using different nanoparticle formulations in a vaccine for RSV, and discuss many examples of safe, and effective vaccines currently in both preclinical and clinical stages of testing.
Collapse
Affiliation(s)
- Laura M Stephens
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
| | - Steven M Varga
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Microbiology & Immunology, University of Iowa, Iowa City, IA 52242, USA
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
27
|
Eichinger KM, Kosanovich JL, Lipp MA, Perkins TN, Petrovsky N, Marshall C, Yondola MA, Empey KM. Maternal immunization with adjuvanted RSV prefusion F protein effectively protects offspring from RSV challenge and alters innate and T cell immunity. Vaccine 2020; 38:7885-7891. [PMID: 33129608 DOI: 10.1016/j.vaccine.2020.10.065] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 10/17/2020] [Accepted: 10/20/2020] [Indexed: 11/16/2022]
Abstract
Respiratory syncytial virus (RSV) commonly causes severe respiratory tract infections in infants, peaking between 2 and 6 months of age; an age at which direct vaccination is unlikely to be effective. Maternal immunization can deliver high levels of antibodies to newborns, providing immediate protection. Following natural infection, antibodies targeting the prefusion conformation of RSV F protein (PreF) have the greatest neutralizing capacity and thus, may provide infants with a high degree of RSV protection when acquired through maternal vaccination. However, the influence of anti-PreF maternal antibodies on infant immunity following RSV exposure has not been elucidated. To address this knowledge gap, offspring born to dams immunized with a RSV PreF vaccine formulation were challenged with RSV and their immune responses were analyzed over time. These studies demonstrated safety and efficacy for RSV-challenged, maternally-immunized offspring but high and waning maternal antibody levels were associated with differential innate and T cell immunity.
Collapse
Affiliation(s)
- Katherine M Eichinger
- Department of Pharmacy and Therapeutics, University of Pittsburgh, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Center for Clinical Pharmaceutical Sciences, University of Pittsburgh, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Department of Medicine, Division of Internal Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jessica L Kosanovich
- Department of Pharmacy and Therapeutics, University of Pittsburgh, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Madeline A Lipp
- Department of Pharmacy and Therapeutics, University of Pittsburgh, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, Bedford Park, SA 5042, Australia; College of Medicine and Public Health, Flinders University, Bedford Park, SA 5042, Australia
| | | | | | - Kerry M Empey
- Department of Pharmacy and Therapeutics, University of Pittsburgh, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Center for Clinical Pharmaceutical Sciences, University of Pittsburgh, School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, USA; Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA, USA; Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
28
|
Kalergis AM, Soto JA, Gálvez NMS, Andrade CA, Fernandez A, Bohmwald K, Bueno SM. Pharmacological management of human respiratory syncytial virus infection. Expert Opin Pharmacother 2020; 21:2293-2303. [PMID: 32808830 DOI: 10.1080/14656566.2020.1806821] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Human respiratory syncytial virus (hRSV) is the primary viral cause of respiratory diseases, leading to bronchiolitis and pneumonia in vulnerable populations. The only current treatment against this virus is palliative, and no efficient and specific vaccine against this pathogen is available. AREAS COVERED The authors describe the disease symptoms caused by hRSV, the economic and social impact of this infection worldwide, and how this infection can be modulated using pharmacological treatments, preventing and limiting its dissemination. The authors discuss the use of antibodies as prophylactic tools -such as palivizumab- and the use of nonspecific drugs to decrease the symptoms associated with the infection -such as bronchodilators, corticoids, and antivirals. They also discuss current vaccine candidates, new prophylactic treatments, and new antivirals options, which are currently being tested. EXPERT OPINION Today, many researchers are focused on developing different strategies to modulate the symptoms induced by hRSV. However, to achieve this, understanding how current treatments are working and their shortcomings needs to be further elucidated.
Collapse
Affiliation(s)
- Alexis M Kalergis
- Millennium Institute on Immunology and Immunotherapy, Departamento De Genética Molecular Y Microbiología, Facultad De Ciencias Biológicas, Pontificia Universidad Católica De Chile , Santiago, Chile.,Departamento De Endocrinología, Facultad De Medicina, Pontificia Universidad Católica De Chile , Santiago, Chile
| | - Jorge A Soto
- Millennium Institute on Immunology and Immunotherapy, Departamento De Genética Molecular Y Microbiología, Facultad De Ciencias Biológicas, Pontificia Universidad Católica De Chile , Santiago, Chile
| | - Nicolás M S Gálvez
- Millennium Institute on Immunology and Immunotherapy, Departamento De Genética Molecular Y Microbiología, Facultad De Ciencias Biológicas, Pontificia Universidad Católica De Chile , Santiago, Chile
| | - Catalina A Andrade
- Millennium Institute on Immunology and Immunotherapy, Departamento De Genética Molecular Y Microbiología, Facultad De Ciencias Biológicas, Pontificia Universidad Católica De Chile , Santiago, Chile
| | - Ayleen Fernandez
- Millennium Institute on Immunology and Immunotherapy, Departamento De Genética Molecular Y Microbiología, Facultad De Ciencias Biológicas, Pontificia Universidad Católica De Chile , Santiago, Chile
| | - Karen Bohmwald
- Millennium Institute on Immunology and Immunotherapy, Departamento De Genética Molecular Y Microbiología, Facultad De Ciencias Biológicas, Pontificia Universidad Católica De Chile , Santiago, Chile
| | - Susan M Bueno
- Millennium Institute on Immunology and Immunotherapy, Departamento De Genética Molecular Y Microbiología, Facultad De Ciencias Biológicas, Pontificia Universidad Católica De Chile , Santiago, Chile
| |
Collapse
|
29
|
Eichinger KM, Kosanovich JL, Gidwani SV, Zomback A, Lipp MA, Perkins TN, Oury TD, Petrovsky N, Marshall CP, Yondola MA, Empey KM. Prefusion RSV F Immunization Elicits Th2-Mediated Lung Pathology in Mice When Formulated With a Th2 (but Not a Th1/Th2-Balanced) Adjuvant Despite Complete Viral Protection. Front Immunol 2020; 11:1673. [PMID: 32849580 PMCID: PMC7403488 DOI: 10.3389/fimmu.2020.01673] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/23/2020] [Indexed: 12/19/2022] Open
Abstract
Respiratory syncytial virus (RSV) remains the most common cause of lower respiratory tract infections in children worldwide. Development of a vaccine has been hindered by the risk of developing enhanced respiratory disease (ERD) upon natural exposure to the virus. Generation of higher quality neutralizing antibodies with stabilized pre-fusion F protein antigens has been proposed as a strategy to prevent ERD. We sought to test whether there was evidence of ERD in naïve BALB/c mice immunized with an unadjuvanted, stabilized pre-fusion F protein, and challenged with RSV line 19. We further sought to determine the extent to which formulation with a Th2-biased (alum) or a more Th1/Th2-balanced (Advax-SM) adjuvant influenced cellular responses and lung pathology. When exposed to RSV, mice immunized with pre-fusion F protein alone (PreF) exhibited increased airway eosinophilia and mucus accumulation. This was further exacerbated by formulation of PreF with Alum (aluminum hydroxide). Conversely, formulation of PreF with a Th1/Th2-balanced adjuvant, Advax-SM, not only suppressed RSV viral replication, but also inhibited airway eosinophilia and mucus accumulation. This was associated with lower numbers of lung innate lymphocyte cells (ILC2s) and CD4+ T cells producing IL-5+ or IL-13+ and increased IFNγ+ CD4+ and CD8+ T cells, in addition to RSV F-specific CD8+ T cells. These data suggest that in the absence of preimmunity, stabilized PreF antigens may still be associated with aberrant Th2 responses that induce lung pathology in response to RSV infection, and can be prevented by formulation with more Th1/Th2-balanced adjuvants that enhance CD4+ and CD8+ IFNγ+ T cell responses. This may support the use of stabilized PreF antigens with Th1/Th2-balanced adjuvants like, Advax-SM, as safer alternatives to alum in RSV vaccine candidates.
Collapse
Affiliation(s)
- Katherine M Eichinger
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Medicine, Division of Internal Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jessica L Kosanovich
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | | | - Aaron Zomback
- Calder Biosciences, New York City, NY, United States
| | - Madeline A Lipp
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States
| | - Timothy N Perkins
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Tim D Oury
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Nikolai Petrovsky
- Vaxine Pty Ltd., Bedford Park, SA, Australia.,College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | | | | | - Kerry M Empey
- Department of Pharmacy and Therapeutics, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Immunology, University of Pittsburgh School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
30
|
Riffault S, Hägglund S, Guzman E, Näslund K, Jouneau L, Dubuquoy C, Pietralunga V, Laubreton D, Boulesteix O, Gauthier D, Remot A, Boukaridi A, Falk A, Shevchenko G, Lind SB, Vargmar K, Zhang B, Kwong PD, Rodriguez MJ, Duran MG, Schwartz-Cornil I, Eléouët JF, Taylor G, Valarcher JF. A Single Shot Pre-fusion-Stabilized Bovine RSV F Vaccine is Safe and Effective in Newborn Calves with Maternally Derived Antibodies. Vaccines (Basel) 2020; 8:vaccines8020231. [PMID: 32443437 PMCID: PMC7349975 DOI: 10.3390/vaccines8020231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/11/2020] [Accepted: 05/11/2020] [Indexed: 01/21/2023] Open
Abstract
Achieving safe and protective vaccination against respiratory syncytial virus (RSV) in infants and in calves has proven a challenging task. The design of recombinant antigens with a conformation close to their native form in virus particles is a major breakthrough. We compared two subunit vaccines, the bovine RSV (BRSV) pre-fusion F (preF) alone or with nanorings formed by the RSV nucleoprotein (preF+N). PreF and N proteins are potent antigenic targets for neutralizing antibodies and T cell responses, respectively. To tackle the challenges of neonatal immunization, three groups of six one-month-old calves with maternally derived serum antibodies (MDA) to BRSV received a single intramuscular injection of PreF, preF+N with MontanideTM ISA61 VG (ISA61) as adjuvant or only ISA61 (control). One month later, all calves were challenged with BRSV and monitored for virus replication in the upper respiratory tract and for clinical signs of disease over one week, and then post-mortem examinations of their lungs were performed. Both preF and preF+N vaccines afforded safe, clinical, and virological protection against BRSV, with little difference between the two subunit vaccines. Analysis of immune parameters pointed to neutralizing antibodies and antibodies to preF as being significant correlates of protection. Thus, a single shot vaccination with preF appears sufficient to reduce the burden of BRSV disease in calves with MDA.
Collapse
Affiliation(s)
- Sabine Riffault
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
- Correspondence: ; Tel.: +33-(0)-134-652-620
| | - Sara Hägglund
- Host Pathogen Interaction Group, Unit of ruminant medicine, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Box 7054, 75007 Uppsala, Sweden; (S.H.); (K.N.); (J.F.V.)
| | - Efrain Guzman
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK; (E.G.); (G.T.)
| | - Katarina Näslund
- Host Pathogen Interaction Group, Unit of ruminant medicine, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Box 7054, 75007 Uppsala, Sweden; (S.H.); (K.N.); (J.F.V.)
| | - Luc Jouneau
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Catherine Dubuquoy
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Vincent Pietralunga
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Daphné Laubreton
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | | | | | - Aude Remot
- INRAE, University of Tours, ISP, 37380 Nouzilly, France;
| | - Abdelhak Boukaridi
- University Paris Saclay, INRAE, AgroParisTech, GABI, 78350 Jouy-en-Josas, France;
| | - Alexander Falk
- Department of Chemistry-BMC, Uppsala University, 875007 Uppsala, Sweden; (A.F.); (G.S.); (S.B.L.)
| | - Ganna Shevchenko
- Department of Chemistry-BMC, Uppsala University, 875007 Uppsala, Sweden; (A.F.); (G.S.); (S.B.L.)
| | - Sara Bergström Lind
- Department of Chemistry-BMC, Uppsala University, 875007 Uppsala, Sweden; (A.F.); (G.S.); (S.B.L.)
| | - Karin Vargmar
- Department of Biomedicine and veterinary public Health, Swedish University of Agricultural Sciences, Box 7054, SE-756 51, 875007 Uppsala, Sweden;
| | - Baoshan Zhang
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (B.Z.); (P.D.K.)
| | - Peter D. Kwong
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA; (B.Z.); (P.D.K.)
| | - María Jose Rodriguez
- Applied Immunology and Genetics, S.L. (INGENASA), 28037 Madrid, Spain; (M.J.R.); (M.G.D.)
| | - Marga Garcia Duran
- Applied Immunology and Genetics, S.L. (INGENASA), 28037 Madrid, Spain; (M.J.R.); (M.G.D.)
| | - Isabelle Schwartz-Cornil
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Jean-François Eléouët
- University Paris-Saclay, INRAE, UVSQ, VIM, 78350 Jouy-en-Josas, France; (L.J.); (C.D.); (V.P.); (D.L.); (I.S.-C.); (J.-F.E.)
| | - Geraldine Taylor
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey GU24 0NF, UK; (E.G.); (G.T.)
| | - Jean François Valarcher
- Host Pathogen Interaction Group, Unit of ruminant medicine, Department of Clinical Sciences, Swedish University of Agricultural Sciences, Box 7054, 75007 Uppsala, Sweden; (S.H.); (K.N.); (J.F.V.)
| |
Collapse
|
31
|
McFarland EJ, Karron RA, Muresan P, Cunningham CK, Libous J, Perlowski C, Thumar B, Gnanashanmugam D, Moye J, Schappell E, Barr E, Rexroad V, Fearn L, Spector SA, Aziz M, Cielo M, Beneri C, Wiznia A, Luongo C, Collins P, Buchholz UJ. Live Respiratory Syncytial Virus Attenuated by M2-2 Deletion and Stabilized Temperature Sensitivity Mutation 1030s Is a Promising Vaccine Candidate in Children. J Infect Dis 2020; 221:534-543. [PMID: 31758177 PMCID: PMC6996856 DOI: 10.1093/infdis/jiz603] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 11/13/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The safety and immunogenicity of live respiratory syncytial virus (RSV) candidate vaccine, LID/ΔM2-2/1030s, with deletion of RSV ribonucleic acid synthesis regulatory protein M2-2 and genetically stabilized temperature-sensitivity mutation 1030s in the RSV polymerase protein was evaluated in RSV-seronegative children. METHODS Respiratory syncytial virus-seronegative children ages 6-24 months received 1 intranasal dose of 105 plaque-forming units (PFU) of LID/ΔM2-2/1030s (n = 21) or placebo (n = 11). The RSV serum antibodies, vaccine shedding, and reactogenicity were assessed. During the following RSV season, medically attended acute respiratory illness (MAARI) and pre- and postsurveillance serum antibody titers were monitored. RESULTS Eighty-five percent of vaccinees shed LID/ΔM2-2/1030s vaccine (median peak nasal wash titers: 3.1 log10 PFU/mL by immunoplaque assay; 5.1 log10 copies/mL by reverse-transcription quantitative polymerase chain reaction) and had ≥4-fold rise in serum-neutralizing antibodies. Respiratory symptoms and fever were common (60% vaccinees and 27% placebo recipients). One vaccinee had grade 2 wheezing with rhinovirus but without concurrent LID/ΔM2-2/1030s shedding. Five of 19 vaccinees had ≥4-fold increases in antibody titers postsurveillance without RSV-MAARI, indicating anamnestic responses without significant illness after infection with community-acquired RSV. CONCLUSIONS LID/ΔM2-2/1030s had excellent infectivity without evidence of genetic instability, induced durable immunity, and primed for anamnestic antibody responses, making it an attractive candidate for further evaluation.
Collapse
Affiliation(s)
- Elizabeth J McFarland
- Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Ruth A Karron
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Petronella Muresan
- Center for Biostatistics in AIDS Research, Harvard T. H. Chan School of Public Health/Frontier Science, Boston, Massachusetts, USA
| | - Coleen K Cunningham
- Department of Pediatrics, Duke University Medical Center, Durham, North Carolina, USA
| | | | | | - Bhagvanji Thumar
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Devasena Gnanashanmugam
- Maternal, Adolescent and Pediatric Research Branch, Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jack Moye
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Elizabeth Schappell
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Emily Barr
- Department of Pediatrics, University of Colorado Anschutz Medical Campus and Children’s Hospital Colorado, Aurora, Colorado, USA
| | - Vivian Rexroad
- Investigational Drug Service Pharmacy, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Laura Fearn
- Department of Pediatrics, Northwestern University Medical School and Ann & Robert H. Lurie Children’s Hospital of Chicago, Chicago, Illinois, USA
| | - Stephen A Spector
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
- Rady Children’s Hospital, San Diego, California, USA
| | - Mariam Aziz
- Section of Infectious Disease, Rush University Medical Center, Chicago, Illinois, USA
| | - Mikhaela Cielo
- Division of Infectious Diseases, Maternal Child and Adolescent Center, University of Southern California Keck School of Medicine, Los Angeles, California, USA
| | - Christy Beneri
- Department of Pediatrics, SUNY Stony Brook, Stony Brook, New York, USA
| | - Andrew Wiznia
- Department of Pediatrics, Albert Einstein College of Medicine and Jacobi Medical Center, Bronx, New York, USA
| | - Cindy Luongo
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Peter Collins
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ursula J Buchholz
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
32
|
Browne SK, Beeler JA, Roberts JN. Summary of the Vaccines and Related Biological Products Advisory Committee meeting held to consider evaluation of vaccine candidates for the prevention of respiratory syncytial virus disease in RSV-naïve infants. Vaccine 2020; 38:101-106. [DOI: 10.1016/j.vaccine.2019.10.048] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/07/2019] [Accepted: 10/17/2019] [Indexed: 10/25/2022]
|
33
|
Kwon YM, Lee Y, Kim KH, Jung YJ, Li Z, Jeeva S, Lee S, Moore ML, Kang SM. Antigenicity and immunogenicity of unique prefusion-mimic F proteins presented on enveloped virus-like particles. Vaccine 2019; 37:6656-6664. [PMID: 31542260 DOI: 10.1016/j.vaccine.2019.09.041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 09/07/2019] [Accepted: 09/10/2019] [Indexed: 11/27/2022]
Abstract
Pre-fusion stabilizing mutations (DS-Cav1) in soluble fusion (F) proteins of human respiratory syncytial virus (RSV) were previously reported. Here we investigated the antigenic and immunogenic properties of pre-fusion like RSV F proteins on enveloped virus-like particles (VLP). Additional mutations were introduced to DS-Cav1 (F-dcmTM VLP); fusion peptide deletion and cleavage mutation site 1 (F1d-dcmTM VLP) or both sites (F12d-dcmTM VLP). F1d-dcmTM VLP and F12d-dcmTM VLP displayed higher reactivity against pre-fusion specific site Ø and antigenic site I and II specific monoclonal antibodies, compared to F-dcmTM VLP with DS-Cav1 only. Mice immunized with F1d-dcmTM VLP and F12d-dcmTM VLP induced higher levels of DS-Cav1 pre-fusion specific IgG antibodies, RSV neutralizing activity titers, and effective lung viral clearance after challenge. These results suggest that cleavage site mutations and fusion peptide deletion in addition to DS-Cav1 mutations have contributed to structural stabilization of pre-fusion like F conformation on enveloped VLP, capable of inducing high levels of pre-fusion F specific and RSV neutralizing antibodies.
Collapse
Affiliation(s)
- Young-Man Kwon
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Ki Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Yu Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Zhuo Li
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Subbiah Jeeva
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Sujin Lee
- Department of Pediatrics, Division of Infectious Disease, Emory University School of Medicine, Atlanta, GA, USA; Children's Healthcare of Atlanta, Atlanta, GA, USA
| | | | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
34
|
Atherton LJ, Jorquera PA, Bakre AA, Tripp RA. Determining Immune and miRNA Biomarkers Related to Respiratory Syncytial Virus (RSV) Vaccine Types. Front Immunol 2019; 10:2323. [PMID: 31649663 PMCID: PMC6794384 DOI: 10.3389/fimmu.2019.02323] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 09/13/2019] [Indexed: 12/19/2022] Open
Abstract
Respiratory Syncytial Virus (RSV) causes serious respiratory tract illness and substantial morbidity and some mortality in populations at the extremes of age, i.e., infants, young children, and the elderly. To date, RSV vaccine development has been unsuccessful, a feature linked to the lack of biomarkers available to assess the safety and efficacy of RSV vaccine candidates. We examined microRNAs (miR) as potential biomarkers for different types of RSV vaccine candidates. In this study, mice were vaccinated with a live attenuated RSV candidate that lacks the small hydrophobic (SH) and attachment (G) proteins (CP52), an RSV G protein microparticle (GA2-MP) vaccine, a formalin-inactivated RSV (FI-RSV) vaccine or were mock-treated. Several immunological endpoints and miR expression profiles were determined in mouse serum and bronchoalveolar lavage (BAL) following vaccine priming, boost, and RSV challenge. We identified miRs that were linked with immunological parameters of disease and protection. We show that miRs are potential biomarkers providing valuable insights for vaccine development.
Collapse
Affiliation(s)
- Lydia J Atherton
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Patricia A Jorquera
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Abhijeet A Bakre
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| | - Ralph A Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, United States
| |
Collapse
|
35
|
Beugeling M, De Zee J, Woerdenbag HJ, Frijlink HW, Wilschut JC, Hinrichs WLJ. Respiratory syncytial virus subunit vaccines based on the viral envelope glycoproteins intended for pregnant women and the elderly. Expert Rev Vaccines 2019; 18:935-950. [PMID: 31446807 DOI: 10.1080/14760584.2019.1657013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Introduction: Respiratory syncytial virus (RSV) causes high morbidity and mortality rates among infants, young children, and the elderly worldwide. Unfortunately, a safe and effective vaccine is still unavailable. In 1966, a formalin-inactivated RSV vaccine failed and resulted in the death of two young children. This failure shifted research toward the development of subunit-based vaccines for pregnant women (to passively vaccinate infants) and the elderly. Among these subunit-based vaccines, the viral envelope glycoproteins show great potential as antigens. Areas covered: In this review, progress in the development of safe and effective subunit RSV vaccines based on the viral envelope glycoproteins and intended for pregnant women and the elderly, are reviewed and discussed. Studies published in the period 2012-2018 were included. Expert opinion: Researchers are close to bringing safe and effective subunit-based RSV vaccines to the market using the viral envelope glycoproteins as antigens. However, it remains a major challenge to elicit protective immunity, with a formulation that has sufficient (storage) stability. These issues may be overcome by using the RSV fusion protein in its pre-fusion conformation, and by formulating this protein as a dry powder. It may further be convenient to administer this powder via the pulmonary route.
Collapse
Affiliation(s)
- Max Beugeling
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen , The Netherlands
| | - Jildou De Zee
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen , The Netherlands
| | - Herman J Woerdenbag
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen , The Netherlands
| | - Henderik W Frijlink
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen , The Netherlands
| | - Jan C Wilschut
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen , Groningen , The Netherlands
| | - Wouter L J Hinrichs
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen , Groningen , The Netherlands
| |
Collapse
|
36
|
Schneider-Ohrum K, Snell Bennett A, Rajani GM, Hostetler L, Maynard SK, Lazzaro M, Cheng LI, O'Day T, Cayatte C. CD4 + T Cells Drive Lung Disease Enhancement Induced by Immunization with Suboptimal Doses of Respiratory Syncytial Virus Fusion Protein in the Mouse Model. J Virol 2019; 93:e00695-19. [PMID: 31092578 PMCID: PMC6639276 DOI: 10.1128/jvi.00695-19] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 05/08/2019] [Indexed: 12/23/2022] Open
Abstract
Respiratory syncytial virus (RSV) infection of seronegative children previously immunized with formalin-inactivated (FI) RSV has been associated with serious enhanced respiratory disease (ERD). The phenomenon was reproduced in the cotton rat and the mouse, and both preclinical models have been routinely used to evaluate the safety of new RSV vaccine candidates. More recently, we demonstrated that immunizations with suboptimal doses of the RSV fusion (F) antigen, in its post- or prefusion conformation, and in the presence of a Th1-biasing adjuvant, unexpectedly led to ERD in the cotton rat model. To assess if those observations are specific to the cotton rat and to elucidate the mechanism by which vaccination with low antigen doses can drive ERD post-RSV challenge, we evaluated RSV post-F antigen dose de-escalation in BALB/c mice in the presence of a Th1-biasing adjuvant. While decreasing antigen doses, we observed an increase in lung inflammation associated with an upregulation of proinflammatory cytokines. The amplitude of the lung histopathology was comparable to that of FI-RSV-induced ERD, confirming the observations made in the cotton rat. Importantly, depletion of CD4+ T cells prior to viral challenge completely abrogated ERD, preventing proinflammatory cytokine upregulation and the infiltration of T cells, neutrophils, eosinophils, and macrophages into the lung. Overall, low-antigen-dose-induced ERD resembles FI-RSV-induced ERD, except that the former appears in the absence of detectable levels of viral replication and in the context of a Th1-biased immune response. Taken together, our observations reinforce the recent concept that vaccines developed for RSV-naïve individuals should be systematically tested under suboptimal dosing conditions.IMPORTANCE RSV poses a significant health care burden and is the leading cause of serious lower-respiratory-tract infections in young children. A formalin-inactivated RSV vaccine developed in the 1960s not only showed a complete lack of efficacy against RSV infection but also induced severe lung disease enhancement in vaccinated children. Since then, establishing safety in preclinical models has been one of the major challenges to RSV vaccine development. We recently observed in the cotton rat model that suboptimal immunizations with RSV fusion protein could induce lung disease enhancement. In the present study, we extended suboptimal dosing evaluation to the mouse model. We confirmed the induction of lung disease enhancement by vaccinations with low antigen doses and dissected the associated immune mechanisms. Our results stress the need to evaluate suboptimal dosing for any new RSV vaccine candidate developed for seronegative infants.
Collapse
Affiliation(s)
| | - Angie Snell Bennett
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | | | - Leigh Hostetler
- Laboratory Animal Resources, MedImmune, Gaithersburg, Maryland, USA
| | - Sean K Maynard
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | - Michelle Lazzaro
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| | - Lily I Cheng
- Pathology Department, MedImmune, Gaithersburg, Maryland, USA
| | - Terrence O'Day
- Statistical Sciences, MedImmune, Gaithersburg, Maryland, USA
| | - Corinne Cayatte
- Department of Infectious Disease/Vaccines, MedImmune, Gaithersburg, Maryland, USA
| |
Collapse
|
37
|
The Optimal Concentration of Formaldehyde is Key to Stabilizing the Pre-Fusion Conformation of Respiratory Syncytial Virus Fusion Protein. Viruses 2019; 11:v11070628. [PMID: 31288455 PMCID: PMC6669674 DOI: 10.3390/v11070628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 07/01/2019] [Accepted: 07/06/2019] [Indexed: 12/12/2022] Open
Abstract
Background: To date, there is no licensed vaccine available to prevent respiratory syncytial virus (RSV) infection. The valuable pre-fusion conformation of the fusion protein (pre-F) is prone to lose high neutralizing antigenic sites. The goals of this study were to stabilize pre-F protein by fixatives and try to find the possibility of developing an inactivated RSV vaccine. Methods: The screen of the optimal fixative condition was performed with flow cytometry. BALB/c mice were immunized intramuscularly with different immunogens. The serum neutralizing antibody titers of immunized mice were determined by neutralization assay. The protection and safety of these immunogens were assessed. Results: Fixation in an optimal concentration of formaldehyde (0.0244%–0.0977%) or paraformaldehyde (0.0625%–1%) was able to stabilize pre-F. Additionally, BALB/c mice inoculated with optimally stabilized pre-F protein (opti-fixed) induced a higher anti-RSV neutralization (9.7 log2, mean value of dilution rate) than those inoculated with unstable (unfixed, 8.91 log2, p < 0.01) or excessively fixed (exce-fixed, 7.28 log2, p < 0.01) pre-F protein. Furthermore, the opti-fixed immunogen did not induce enhanced RSV disease. Conclusions: Only the proper concentration of fixatives could stabilize pre-F and the optimal formaldehyde condition provides a potential reference for development of an inactivated RSV vaccine.
Collapse
|
38
|
Boukhvalova MS, Yim KC, Blanco J. Cotton rat model for testing vaccines and antivirals against respiratory syncytial virus. Antivir Chem Chemother 2019; 26:2040206618770518. [PMID: 29768937 PMCID: PMC5987903 DOI: 10.1177/2040206618770518] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Respiratory syncytial virus is the leading cause of pneumonia and bronchiolitis in infants and is a serious health risk for elderly and immunocompromised individuals. No vaccine has yet been approved to prevent respiratory syncytial virus infection and the only available treatment is immunoprophylaxis of severe respiratory syncytial virus disease in high-risk infants with Palivizumab (Synagis®). The development of respiratory syncytial virus vaccine has been hampered by the phenomenon of enhanced respiratory syncytial virus disease observed during trials of a formalin-inactivated respiratory syncytial virus in 1960s. A search for effective respiratory syncytial virus therapeutics has been complicated by the fact that some of the most advanced respiratory syncytial virus antivirals, while highly effective in a prophylactic setting, had not demonstrated clinical efficacy when given after infection. A number of respiratory syncytial virus vaccines and antivirals are currently under development, including several vaccines proposed for maternal immunization. The cotton rat Sigmodon hispidus is an animal model of respiratory syncytial virus infection with demonstrated translational value. Special cohort scenarios, such as infection under conditions of immunosuppression and maternal immunization have been modeled in the cotton rat and are summarized here. In this review, we focus on the recent use of the cotton rat model for testing respiratory syncytial virus vaccine and therapeutic candidates in preclinical setting, including the use of special cohort models. An overview of published studies spanning the period of the last three years is provided. The emphasis, where possible, is made on candidates in the latest stages of preclinical development or currently in clinical trials.
Collapse
Affiliation(s)
| | - K C Yim
- Sigmovir Biosystems, Inc., Rockville, MD, USA
| | - Jcg Blanco
- Sigmovir Biosystems, Inc., Rockville, MD, USA
| |
Collapse
|
39
|
Lee Y, Ko EJ, Kim KH, Lee YT, Hwang HS, Kwon YM, Graham BS, Kang SM. A unique combination adjuvant modulates immune responses preventing vaccine-enhanced pulmonary histopathology after a single dose vaccination with fusion protein and challenge with respiratory syncytial virus. Virology 2019; 534:1-13. [PMID: 31163351 DOI: 10.1016/j.virol.2019.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 10/26/2022]
Abstract
Alum adjuvanted formalin-inactivated respiratory syncytial virus (RSV) vaccination resulted in enhanced respiratory disease in young children upon natural infection. Here, we investigated the adjuvant effects of monophosphoryl lipid A (MPL) and oligodeoxynucleotide CpG (CpG) on vaccine-enhanced respiratory disease after fusion (F) protein prime vaccination and RSV challenge in infant and adult mouse models. Combination CpG + MPL adjuvant in RSV F protein single dose priming of infant and adult age mice was found to promote the induction of IgG2a isotype antibodies and neutralizing activity, and lung viral clearance after challenge. CpG + MPL adjuvanted F protein (Fp) priming of infant and adult age mice was effective in avoiding lung histopathology, in reducing interleukin-4+ CD4 T cells and cellular infiltration of monocytes and neutrophils after RSV challenge. This study suggests that combination CpG and MPL adjuvant in RSV subunit vaccination might contribute to priming protective immune responses and preventing inflammatory RSV disease after infection.
Collapse
Affiliation(s)
- Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Hye Suk Hwang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA; Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, South Korea
| | - Young-Man Kwon
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sang Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
40
|
Lee Y, Ko EJ, Kim KH, Lee YT, Hwang HS, Jung YJ, Jeeva S, Kwon YM, Seong BL, Kang SM. The efficacy of inactivated split respiratory syncytial virus as a vaccine candidate and the effects of novel combination adjuvants. Antiviral Res 2019; 168:100-108. [PMID: 31150678 DOI: 10.1016/j.antiviral.2019.05.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/18/2019] [Accepted: 05/27/2019] [Indexed: 12/24/2022]
Abstract
Clinical trials with alum-adjuvanted formalin-inactivated human respiratory syncytial virus (FI-RSV) vaccine failed in children due to vaccine-enhanced disease upon RSV infection. In this study, we found that inactivated, detergent-split RSV vaccine (Split) displayed higher reactivity against neutralizing antibodies in vitro and less histopathology in primed adult mice after challenge, compared to FI-RSV. The immunogenicity and efficacy of FI-RSV and Split RSV vaccine were further determined in 2 weeks old mice after a single dose in the absence or presence of monophosphoryl lipid A (MPL) + CpG combination adjuvant. Split RSV with MPL + CpG adjuvant was effective in increasing T helper type 1 (Th1) immune responses and IgG2a isotype antibodies, neutralizing activity, and lung viral clearance as well as modulating immune responses to prevent pulmonary histopathology after RSV vaccination and challenge. This study demonstrates the efficacy of Split RSV as an effective vaccine candidate.
Collapse
Affiliation(s)
- Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Eun-Ju Ko
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA; Vaccine Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Hye Suk Hwang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA; Department of Microbiology, Chonnam National University, Hwasun-gun, Jeonnam, Republic of Korea
| | - Yu-Jin Jung
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Subbiah Jeeva
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Young-Man Kwon
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Baik Lin Seong
- Institute of Life Science and Biotechnology, Yonsei University, Seodaemun-Gu, Seoul, Republic of Korea
| | - Sang Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA.
| |
Collapse
|
41
|
Kwon YM, Hwang HS, Lee YT, Kim KH, Lee Y, Kim MC, Lee YN, Quan FS, Moore ML, Kang SM. Respiratory Syncytial Virus Fusion Protein-encoding DNA Vaccine Is Less Effective in Conferring Protection against Inflammatory Disease than a Virus-like Particle Platform. Immune Netw 2019; 19:e18. [PMID: 31281715 PMCID: PMC6597443 DOI: 10.4110/in.2019.19.e18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/17/2022] Open
Abstract
Formalin-inactivated respiratory syncytial virus (RSV) vaccination causes vaccine-enhanced disease (VED) after RSV infection. It is considered that vaccine platforms enabling endogenous synthesis of RSV immunogens would induce favorable immune responses than non-replicating subunit vaccines in avoiding VED. Here, we investigated the immunogenicity, protection, and disease in mice after vaccination with RSV fusion protein (F) encoding plasmid DNA (F-DNA) or virus-like particles presenting RSV F (F-VLP). F-DNA vaccination induced CD8 T cells and RSV neutralizing Abs, whereas F-VLP elicited higher levels of IgG2a isotype and neutralizing Abs, and germinal center B cells, contributing to protection by controlling lung viral loads after RSV challenge. However, mice that were immunized with F-DNA displayed weight loss and pulmonary histopathology, and induced F specific CD8 T cell responses and recruitment of monocytes and plasmacytoid dendritic cells into the lungs. These innate immune parameters, RSV disease, and pulmonary histopathology were lower in mice that were immunized with F-VLP after challenge. This study provides important insight into developing effective and safe RSV vaccines.
Collapse
Affiliation(s)
- Young-Man Kwon
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Hye Suk Hwang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.,Department of Microbiology, Chonnam National University Medical School, Hwasun 58128, Korea
| | - Young-Tae Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.,Green Cross Cell Corp., Yongin 16924, Korea
| | - Ki-Hye Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Youri Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Min-Chul Kim
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.,Komipharm Co., Ltd., Siheung 15094, Korea
| | - Yu-Na Lee
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.,Animal and Plant Quarantine Agency, Gimcheon 39660, Korea
| | - Fu-Shi Quan
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.,Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul 02447, Korea
| | | | - Sang-Moo Kang
- Center for Inflammation, Immunity & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| |
Collapse
|
42
|
Russell MS, Creskey M, Muralidharan A, Li C, Gao J, Chen W, Larocque L, Lavoie JR, Farnsworth A, Rosu-Myles M, Hashem AM, Yauk CL, Cao J, Van Domselaar G, Cyr T, Li X. Unveiling Integrated Functional Pathways Leading to Enhanced Respiratory Disease Associated With Inactivated Respiratory Syncytial Viral Vaccine. Front Immunol 2019; 10:597. [PMID: 30984178 PMCID: PMC6449435 DOI: 10.3389/fimmu.2019.00597] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 03/06/2019] [Indexed: 02/01/2023] Open
Abstract
Respiratory syncytial virus (RSV) infection is a severe threat to young children and the elderly. Despite decades of research, no vaccine has been approved. Notably, instead of affording protection, a formalin-inactivated RSV vaccine induced severe respiratory disease including deaths in vaccinated children in a 1960s clinical trial; however, recent studies indicate that other forms of experimental vaccines can also induce pulmonary pathology in pre-clinical studies. These findings suggest that multiple factors/pathways could be involved in the development of enhanced respiratory diseases. Clearly, a better understanding of the mechanisms underlying such adverse reactions is critically important for the development of safe and efficacious vaccines against RSV infection, given the exponential growth of RSV vaccine clinical trials in recent years. By employing an integrated systems biology approach in a pre-clinical cotton rat model, we unraveled a complex network of pulmonary canonical pathways leading to disease development in vaccinated animals upon subsequent RSV infections. Cytokines including IL-1, IL-6 GRO/IL-8, and IL-17 in conjunction with mobilized pulmonary inflammatory cells could play important roles in disease development, which involved a wide range of host responses including exacerbated pulmonary inflammation, oxidative stress, hyperreactivity, and homeostatic imbalance between coagulation and fibrinolysis. Moreover, the observed elevated levels of MyD88 implicate the involvement of this critical signal transduction module as the central node of the inflammatory pathways leading to exacerbated pulmonary pathology. Finally, the immunopathological consequences of inactivated vaccine immunization and subsequent RSV exposure were further substantiated by histological analyses of these key proteins along with inflammatory cytokines, while hypercoagulation was supported by increased pulmonary fibrinogen/fibrin accompanied by reduced levels of plasma D-dimers. Enhanced respiratory disease associated with inactivated RSV vaccine involves a complex network of host responses, resulting in significant pulmonary lesions and clinical manifestations such as tachypnea and airway obstruction. The mechanistic insight into the convergence of different signal pathways and identification of biomarkers could help facilitate the development of safe and effective RSV vaccine and formulation of new targeted interventions.
Collapse
Affiliation(s)
- Marsha S Russell
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Marybeth Creskey
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Abenaya Muralidharan
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Changgui Li
- National Institutes for Food and Drug Control, WHO Collaborating Center for Standardization and Evaluation of Biologicals, Beijing, China
| | - Jun Gao
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Wangxue Chen
- Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Louise Larocque
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Jessie R Lavoie
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Aaron Farnsworth
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Michael Rosu-Myles
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Anwar M Hashem
- Immunotherapy Unit, Department of Medical Microbiology and Parasitology, Faculty of Medicine and Vaccines, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Carole L Yauk
- Mechanistic Studies Division, Environmental and Radiation Health Sciences Directorate, Healthy Environments and Consumer Safety Branch (HECSB), Health Canada, Ottawa, ON, Canada
| | - Jingxin Cao
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Gary Van Domselaar
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Terry Cyr
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Xuguang Li
- Centre for Biologics Evaluation, Biologics and Genetic Therapies Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada.,Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
43
|
van Erp EA, Luytjes W, Ferwerda G, van Kasteren PB. Fc-Mediated Antibody Effector Functions During Respiratory Syncytial Virus Infection and Disease. Front Immunol 2019; 10:548. [PMID: 30967872 PMCID: PMC6438959 DOI: 10.3389/fimmu.2019.00548] [Citation(s) in RCA: 184] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 02/28/2019] [Indexed: 12/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract infections and hospitalization in infants under 1 year of age and there is currently no market-approved vaccine available. For protection against infection, young children mainly depend on their innate immune system and maternal antibodies. Traditionally, antibody-mediated protection against viral infections is thought to be mediated by direct binding of antibodies to viral particles, resulting in virus neutralization. However, in the case of RSV, virus neutralization titers do not provide an adequate correlate of protection. The current lack of understanding of the mechanisms by which antibodies can protect against RSV infection and disease or, alternatively, contribute to disease severity, hampers the design of safe and effective vaccines against this virus. Importantly, neutralization is only one of many mechanisms by which antibodies can interfere with viral infection. Antibodies consist of two structural regions: a variable fragment (Fab) that mediates antigen binding and a constant fragment (Fc) that mediates downstream effector functions via its interaction with Fc-receptors on (innate) immune cells or with C1q, the recognition molecule of the complement system. The interaction with Fc-receptors can lead to killing of virus-infected cells through a variety of immune effector mechanisms, including antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP). Antibody-mediated complement activation may lead to complement-dependent cytotoxicity (CDC). In addition, both Fc-receptor interactions and complement activation can exert a broad range of immunomodulatory functions. Recent studies have emphasized the importance of Fc-mediated antibody effector functions in both protection and pathogenesis for various infectious agents. In this review article, we aim to provide a comprehensive overview of the current knowledge on Fc-mediated antibody effector functions in the context of RSV infection, discuss their potential role in establishing the balance between protection and pathogenesis, and point out important gaps in our understanding of these processes. Furthermore, we elaborate on the regulation of these effector functions on both the cellular and humoral side. Finally, we discuss the implications of Fc-mediated antibody effector functions for the rational design of safe and effective vaccines and monoclonal antibody therapies against RSV.
Collapse
Affiliation(s)
- Elisabeth A. van Erp
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Radboud Center for Infectious Diseases, Nijmegen, Netherlands
| | - Willem Luytjes
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Gerben Ferwerda
- Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Nijmegen, Netherlands
- Radboud Center for Infectious Diseases, Nijmegen, Netherlands
| | - Puck B. van Kasteren
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
44
|
Smatti MK, Al Thani AA, Yassine HM. Viral-Induced Enhanced Disease Illness. Front Microbiol 2018; 9:2991. [PMID: 30568643 PMCID: PMC6290032 DOI: 10.3389/fmicb.2018.02991] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 11/19/2018] [Indexed: 12/24/2022] Open
Abstract
Understanding immune responses to viral infections is crucial to progress in the quest for effective infection prevention and control. The host immunity involves various mechanisms to combat viral infections. Under certain circumstances, a viral infection or vaccination may result in a subverted immune system, which may lead to an exacerbated illness. Clinical evidence of enhanced illness by preexisting antibodies from vaccination, infection or maternal passive immunity is available for several viruses and is presumptively proposed for other viruses. Multiple mechanisms have been proposed to explain this phenomenon. It has been confirmed that certain infection- and/or vaccine-induced immunity could exacerbate viral infectivity in Fc receptor- or complement bearing cells- mediated mechanisms. Considering that antibody dependent enhancement (ADE) is a major obstacle in vaccine development, there are continues efforts to understand the underlying mechanisms through identification of the epitopes and antibodies responsible for disease enhancement or protection. This review discusses the recent findings on virally induced ADE, and highlights the potential mechanisms leading to this condition.
Collapse
Affiliation(s)
- Maria K Smatti
- Biomedical Research Center, Qatar University, Doha, Qatar
| | | | - Hadi M Yassine
- Biomedical Research Center, Qatar University, Doha, Qatar
| |
Collapse
|
45
|
Lee Y, Lee YT, Ko EJ, Kim KH, Hwang HS, Park S, Kwon YM, Kang SM. Soluble F proteins exacerbate pulmonary histopathology after vaccination upon respiratory syncytial virus challenge but not when presented on virus-like particles. Hum Vaccin Immunother 2018; 13:2594-2605. [PMID: 28854003 DOI: 10.1080/21645515.2017.1362514] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Respiratory syncytial virus (RSV) fusion (F) protein is suggested to be a protective vaccine target although its efficacy and safety concerns remain not well understood. We investigated immunogenicity, efficacy, and safety of F proteins in a soluble form or on virus-like particle (F-VLP). F VLP preferentially elicited IgG2a antibody and T helper type 1 (Th1) immune responses whereas F protein induced IgG1 isotype and Th2 responses. Despite lung viral clearance after prime or prime-boost and then RSV challenge, F protein immune mice displayed weight loss and lung histopathology and high mucus production and eosinophils. In contrast, prime or prime-boost vaccination of F VLP induced effective protection, prevented infiltration of eosinophils and vaccine- enhanced disease after challenge. This study provides insight into developing an effective and safe RSV vaccine candidate.
Collapse
Affiliation(s)
- Youri Lee
- a Center for Inflammation, Immunity & Infection , Institute for Biomedical Sciences, Georgia State University , Atlanta , GA , USA.,b Department of Biology Institute for Biomedical Sciences , Georgia State University , Atlanta , GA , USA
| | - Young-Tae Lee
- a Center for Inflammation, Immunity & Infection , Institute for Biomedical Sciences, Georgia State University , Atlanta , GA , USA
| | - Eun-Ju Ko
- a Center for Inflammation, Immunity & Infection , Institute for Biomedical Sciences, Georgia State University , Atlanta , GA , USA
| | - Ki-Hye Kim
- a Center for Inflammation, Immunity & Infection , Institute for Biomedical Sciences, Georgia State University , Atlanta , GA , USA
| | - Hye Suk Hwang
- a Center for Inflammation, Immunity & Infection , Institute for Biomedical Sciences, Georgia State University , Atlanta , GA , USA
| | - Soojin Park
- a Center for Inflammation, Immunity & Infection , Institute for Biomedical Sciences, Georgia State University , Atlanta , GA , USA
| | - Young-Man Kwon
- a Center for Inflammation, Immunity & Infection , Institute for Biomedical Sciences, Georgia State University , Atlanta , GA , USA
| | - Sang Moo Kang
- a Center for Inflammation, Immunity & Infection , Institute for Biomedical Sciences, Georgia State University , Atlanta , GA , USA.,b Department of Biology Institute for Biomedical Sciences , Georgia State University , Atlanta , GA , USA
| |
Collapse
|
46
|
Blanco JCG, Boukhvalova MS, Morrison TG, Vogel SN. A multifaceted approach to RSV vaccination. Hum Vaccin Immunother 2018; 14:1734-1745. [PMID: 29771625 PMCID: PMC6067850 DOI: 10.1080/21645515.2018.1472183] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/12/2018] [Accepted: 04/29/2018] [Indexed: 12/15/2022] Open
Abstract
Respiratory Syncytial Virus (RSV) is the leading cause of pneumonia and bronchiolitis in infants, resulting in significant morbidity and mortality worldwide. In addition, RSV infections occur throughout different ages, thus, maintaining the virus in circulation, and increasing health risk to more susceptible populations such as infants, the elderly, and the immunocompromised. To date, there is no vaccine approved to prevent RSV infection or minimize symptoms of infection. Current clinical trials for vaccines against RSV are being carried out in four very different populations. There are vaccines that target two different pediatric populations, infants 2 to 6 month of age and seropositive children over 6 months of age, as well as women (non-pregnant or pregnant in their third trimester). There are vaccines that target adult and elderly populations. In this review, we will present and discuss RSV vaccine candidates currently in clinical trials. We will describe the preclinical studies instrumental for their advancement, with the goal of introducing new preclinical models that may more accurately predict the outcome of clinical vaccine studies.
Collapse
|
47
|
Boukhvalova MS, Mbaye A, Kovtun S, Yim KC, Konstantinova T, Getachew T, Khurana S, Falsey AR, Blanco JCG. Improving ability of RSV microneutralization assay to detect G-specific and cross-reactive neutralizing antibodies through immortalized cell line selection. Vaccine 2018; 36:4657-4662. [PMID: 29960801 DOI: 10.1016/j.vaccine.2018.06.045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/08/2018] [Accepted: 06/16/2018] [Indexed: 11/29/2022]
Abstract
Respiratory syncytial virus (RSV) is a significant cause of bronchiolitis and pneumonia. Protection against RSV is associated with neutralizing antibodies against the fusion (F) and attachment (G) glycoproteins. Several RSV vaccine candidates are in development, but their immunogenicity is hard to compare due to the little-understood differences between multiple RSV neutralizing antibody assays used. Existing assays utilize primarily Vero or HEp-2 cells, but their ability to detect G-neutralizing antibodies or antibodies against specific RSV strains is unclear. In this work, we developed an RSV microneutralization assay (MNA) using unmodified RSV and immortalized cell line derived from human airway epithelial cells (A549). Performance of A549-, HEp-2- and Vero-based MNA was compared under the same assay conditions (fixed amount of virus and cells) with regards to detection of neutralizing antibodies against RSV A or B viruses, G-reactive neutralizing antibodies, and effect of complement. Our results indicate that A549 cells yield the highest MNA titers, particularly in the RSV A/A2 MNA, are least susceptible to complement-enhancing effect of neutralizing titer readout and are superior to Vero or HEp-2 MNA at recognizing G-reactive neutralizing antibodies when no complement is used. Vero cells, however, can be more consistent at recognizing neutralizing antibodies against multiple RSV strains. The choice of substrate cells thus affects the outcome of MNA, as some immortalized cells better support detection of broader range of neutralizing antibodies, while others facilitate detection of G-targeting neutralizing antibodies, a long-thought prerogative of primary airway epithelial cells.
Collapse
Affiliation(s)
- M S Boukhvalova
- Sigmovir Biosystems, Inc, 9610 Medical Center Drive, Suite 100, Rockville, MD 20850, USA.
| | - A Mbaye
- Sigmovir Biosystems, Inc, 9610 Medical Center Drive, Suite 100, Rockville, MD 20850, USA
| | - S Kovtun
- Sigmovir Biosystems, Inc, 9610 Medical Center Drive, Suite 100, Rockville, MD 20850, USA
| | - K C Yim
- Sigmovir Biosystems, Inc, 9610 Medical Center Drive, Suite 100, Rockville, MD 20850, USA
| | - T Konstantinova
- Sigmovir Biosystems, Inc, 9610 Medical Center Drive, Suite 100, Rockville, MD 20850, USA
| | - T Getachew
- Sigmovir Biosystems, Inc, 9610 Medical Center Drive, Suite 100, Rockville, MD 20850, USA
| | - S Khurana
- Division of Viral Products, Center for Biologics Evaluation and Research (CBER), Food and Drug Administration (FDA), 10903 New Hampshire Avenue, Silver Spring, MD 20993, USA
| | - A R Falsey
- University of Rochester Medical Center, Rochester General Hospital, 1425 Portland Avenue, Infectious Diseases Unit, Rochester, NY 14621, USA
| | - J C G Blanco
- Sigmovir Biosystems, Inc, 9610 Medical Center Drive, Suite 100, Rockville, MD 20850, USA
| |
Collapse
|
48
|
Enhancing the Thermostability and Immunogenicity of a Respiratory Syncytial Virus (RSV) Live-Attenuated Vaccine by Incorporating Unique RSV Line19F Protein Residues. J Virol 2018; 92:JVI.01568-17. [PMID: 29263264 DOI: 10.1128/jvi.01568-17] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/12/2017] [Indexed: 12/20/2022] Open
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in infants, and an effective vaccine is not yet available. We previously generated an RSV live-attenuated vaccine (LAV) candidate, DB1, which was attenuated by a low-fusion subgroup B F protein (BAF) and codon-deoptimized nonstructural protein genes. DB1 was immunogenic and protective in cotton rats but lacked thermostability and stability of the prefusion conformation of F compared to strains with the line19F gene. We hypothesized that substitution of unique residues from the thermostable A2-line19F strain could thermostabilize DB1 and boost its immunogenicity. We therefore substituted 4 unique line19F residues into the BAF protein of DB1 by site-directed mutagenesis and rescued the recombinant virus, DB1-QUAD. Compared to DB1, DB1-QUAD had improved thermostability at 4°C and higher levels of prefusion F as measured by enzyme-linked immunosorbent assays (ELISAs). DB1-QUAD was attenuated in normal human bronchial epithelial cells, in BALB/c mice, and in cotton rats but grew to wild-type titers in Vero cells. In mice, DB1-QUAD was highly immunogenic and generated significantly higher neutralizing antibody titers to a panel of RSV A and B strains than did DB1. DB1-QUAD was also efficacious against wild-type RSV challenge in mice and cotton rats. Thus, substitution of unique line19F residues into RSV LAV DB1 enhanced vaccine thermostability, incorporation of prefusion F, and immunogenicity and generated a promising vaccine candidate that merits further investigation.IMPORTANCE We boosted the thermostability and immunogenicity of an RSV live-attenuated vaccine candidate by substituting 4 unique residues from the RSV line19F protein into the F protein of the heterologous vaccine strain DB1. The resultant vaccine candidate, DB1-QUAD, was thermostable, attenuated in vivo, highly immunogenic, and protective against RSV challenge in mice and cotton rats.
Collapse
|
49
|
Kulkarni PS, Hurwitz JL, Simões EAF, Piedra PA. Establishing Correlates of Protection for Vaccine Development: Considerations for the Respiratory Syncytial Virus Vaccine Field. Viral Immunol 2018; 31:195-203. [PMID: 29336703 DOI: 10.1089/vim.2017.0147] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Correlates of protection (CoPs) can play a significant role in vaccine development by assisting the selection of vaccine candidates for clinical trials, supporting clinical trial design and implementation, and simplifying tests of vaccine modifications. Because of this important role in vaccine development, it is essential that CoPs be defined by well-designed immunogenicity and efficacy studies, with attention paid to benefits and limitations. The respiratory syncytial virus (RSV) field is unique in that a great deal of information about the humoral response is available from basic research and clinical studies. Polyclonal and monoclonal antibodies have been used routinely in the clinic to protect vulnerable infants from infection, providing a wealth of information about correlations between neutralizing antibodies and disease prevention. Considerations for the establishment of future CoPs to support RSV vaccine development in different populations are therefore discussed.
Collapse
Affiliation(s)
| | - Julia L Hurwitz
- 2 Department of Infectious Diseases, St. Jude Children's Research Hospital , Memphis, Tennessee.,3 Department of Microbiology, Immunology, and Biochemistry, The University of Tennessee Health Science Center , Memphis, Tennessee
| | - Eric A F Simões
- 4 Department of Pediatrics, University of Colorado School of Medicine , Aurora, Colorado.,5 Department of Epidemiology, Colorado School of Public Health , Section of Infectious Diseases, Children's Hospital Colorado, Aurora, Colorado
| | - Pedro A Piedra
- 6 Department of Molecular Virology and Microbiology, Baylor College of Medicine , Houston, Texas
| |
Collapse
|
50
|
Clark CM, Guerrero-Plata A. Respiratory Syncytial Virus Vaccine Approaches: a Current Overview. CURRENT CLINICAL MICROBIOLOGY REPORTS 2017; 4:202-207. [PMID: 30009126 PMCID: PMC6040676 DOI: 10.1007/s40588-017-0074-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
PURPOSE OF REVIEW Respiratory syncytial virus (RSV) is a global human pathogen responsible for lower respiratory tract infections (LRTI). While RSV infection is innocuous in healthy adults, it is the leading cause of infant hospitalization for respiratory tract infection. Nearly everyone shows evidence of an RSV infection by the age of 3. However, there is still not a vaccine commercially available. This review will provide an update on the clinical and preclinical vaccine studies and different approaches to prevent RSV infection. RECENT FINDINGS Novel vaccine approaches that induce protection against RSV without enhancement of respiratory tract disease. SUMMARY Recent technological approaches have led to generation of different strategies to prevent RSV infection, including live attenuated, chimeric, and subunit vaccines, virus-like particles, and nanoparticles. These vaccine approaches represent promising candidates towards an efficient RSV vaccine that effectively protects infants, children, and adults.
Collapse
Affiliation(s)
- Carolyn M Clark
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Antonieta Guerrero-Plata
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
- Center for Experimental Infectious Disease Research, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|