1
|
Zuo L, Liu Q, Zhang K, Zhao L, Lin S, Dai Y, Sun Y, Li Y, Zhang P, Shen H, He D, Ma S, Long X, Chen Y, Luo Y, Wong G. Self-amplifying mRNA vaccines protect elderly BALB/c mice against a lethal respiratory syncytial virus infection. Mol Ther 2024:S1525-0016(24)00812-8. [PMID: 39673128 DOI: 10.1016/j.ymthe.2024.12.013] [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: 05/03/2024] [Revised: 11/05/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024] Open
Abstract
Respiratory syncytial virus (RSV) represents a significant threat, being a primary cause of critical lower respiratory tract infections and fatalities among infants and the elderly worldwide, and poses a challenge to global public health. This urgent public health challenge necessitates the swift development of safe and effective vaccines capable of eliciting robust immune responses at low doses. Addressing this need, our study investigated five self-amplifying mRNA (sa-mRNA) candidate vaccines that encode the various pre-fusion conformations of the RSV fusion protein. When administered via low-dose intramuscular injection to 8-month-old elderly mice, these vaccines triggered potent humoral reactions and T helper type 1-biased cellular immunity. A prime-boost strategy followed by challenge with a lethal, mouse-adapted RSV strain showed that three of these sa-mRNA candidates achieved greater than 80% survival rates. An immune correlates of protection analysis contrasting immunized survivors with non-survivors suggest that the titers of IgG and neutralizing antibody are associated with vaccine-mediated protection from RSV infection. Our results highlight the usefulness of sa-mRNA vaccines to play a crucial role in forging an effective defense against RSV, addressing a critical need in protecting vulnerable populations against this virus.
Collapse
Affiliation(s)
- Lulu Zuo
- Viral Hemorrhagic Fevers Research Unit, Institut Pasteur of Shanghai (now Shanghai Institute of Immunity and Infection), Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 201203, China
| | - Qixing Liu
- Viral Hemorrhagic Fevers Research Unit, Institut Pasteur of Shanghai (now Shanghai Institute of Immunity and Infection), Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; Department of Nucleic Acid Research, Hongene Biotech, Shanghai 201203, China
| | - Ke Zhang
- Viral Hemorrhagic Fevers Research Unit, Institut Pasteur of Shanghai (now Shanghai Institute of Immunity and Infection), Chinese Academy of Sciences, Shanghai 201203, China; Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control/Institute of Virology, School of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
| | - Lu Zhao
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 201203, China
| | - Siyu Lin
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control/Institute of Virology, School of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
| | - You Dai
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control/Institute of Virology, School of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
| | - Yun Sun
- College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Yingwen Li
- Viral Hemorrhagic Fevers Research Unit, Institut Pasteur of Shanghai (now Shanghai Institute of Immunity and Infection), Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pingping Zhang
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control/Institute of Virology, School of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
| | - Huyan Shen
- Guizhou Key Laboratory of Microbio and Infectious Disease Prevention & Control/Institute of Virology, School of Basic Medicine, Guizhou Medical University, Guiyang 550025, China
| | - Dongmei He
- Institute of Pathogenic Microorganisms, Guangdong Provincial Center for Disease Control and Prevention, Guangzhou 511430, China; Guangdong Provincial Key Laboratory of Pathogen Detection for Emerging Infectious Disease Response, Guangzhou 511430, China
| | - Shuang Ma
- Department of Clinical Laboratory, Huadu Maternal and Child Health Care Hospital, Guangzhou 511430, China
| | - Xianhua Long
- Guangzhou DAAN Clinical Laboratory Center, Guangzhou 510665, China
| | - Yanhua Chen
- Viral Hemorrhagic Fevers Research Unit, Institut Pasteur of Shanghai (now Shanghai Institute of Immunity and Infection), Chinese Academy of Sciences, Shanghai 201203, China
| | - Yusi Luo
- Department of Emergency ICU, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China.
| | - Gary Wong
- Viral Hemorrhagic Fevers Research Unit, Institut Pasteur of Shanghai (now Shanghai Institute of Immunity and Infection), Chinese Academy of Sciences, Shanghai 201203, China; Virology Laboratory, Institut Pasteur du Laos, Vientiane 01030, Laos.
| |
Collapse
|
2
|
Matsuyama-Ito R, Hogiri T, Kishida H, Takedomi K, Okada O, Nishizawa A, Higashi-Nakatani S, Omasa T. Generation of novel respiratory syncytial virus vaccine candidate antigens that can induce high levels of prefusion-specific antibodies. J Biosci Bioeng 2024; 138:127-136. [PMID: 38851988 DOI: 10.1016/j.jbiosc.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 04/27/2024] [Accepted: 05/19/2024] [Indexed: 06/10/2024]
Abstract
Respiratory syncytial virus (RSV) infection is an acute respiratory infection caused by RSV. It occurs worldwide, and for over 50 years, several attempts have been made to research and develop vaccines to prevent RSV infection; effective preventive vaccines are eagerly awaited. The RSV fusion (F) protein, which has gained attention as a vaccine antigen, causes a dynamic structural change from the preF to postF state. Therefore, the structural changes in proteins must be regulated to produce a vaccine antigen that can efficiently induce antibodies with high virus-neutralizing activity. We successfully discovered several mutations that stabilized the antigen site Ø in the preF state, trimerized it, and improved the level of protein expression through observation and computational analysis of the RSV-F protein structure and amino acid mutation analysis of RSV strains. The four RSV-F protein mutants that resulted from the combination of these effective mutations stably conserved a wide range of preF- and trimeric preF-specific epitopes with high virus-neutralizing activity. Absorption assay using human serum revealed that mutants constructed bound to antibodies with virus-neutralizing activity that were induced by natural RSV infection, whereas they hardly bound to anti-postF antibodies without virus-neutralizing activity. Furthermore, mouse immunization demonstrated that our constructed mutants induced a high percentage of antibodies that bind to the preF-specific antigen site. These characteristics suggest that the mutants constructed can be superior vaccine antigens from the viewpoint of RSV infection prevention effect and safety.
Collapse
Affiliation(s)
- Rima Matsuyama-Ito
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Modality Laboratories, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 2-26-1 Muraoka-Higashi, Fujisawa, Kanagawa 251-8555, Japan.
| | - Tomoharu Hogiri
- Modality Laboratories, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Hiroyuki Kishida
- Discovery Technology Laboratories, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Kei Takedomi
- Discovery Technology Laboratories, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Okimasa Okada
- Discovery Technology Laboratories, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Akitoshi Nishizawa
- Modality Laboratories, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 2-26-1 Muraoka-Higashi, Fujisawa, Kanagawa 251-8555, Japan
| | - Sakiko Higashi-Nakatani
- Modality Laboratories, Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000 Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa 227-0033, Japan
| | - Takeshi Omasa
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
3
|
Ma J, Chen L, Tang S, Shi Y. Efficacy and safety of respiratory syncytial virus vaccination during pregnancy to prevent lower respiratory tract illness in newborns and infants: a systematic review and meta-analysis of randomized controlled trials. Front Pediatr 2024; 11:1260740. [PMID: 38357264 PMCID: PMC10864603 DOI: 10.3389/fped.2023.1260740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/28/2023] [Indexed: 02/16/2024] Open
Abstract
To evaluate the effectiveness and safety of respiratory syncytial virus (RSV) vaccination during pregnancy in preventing lower respiratory tract infection (LRTI) in infants and neonates, we conducted a systematic search of randomized controlled trials (RCTs) in five databases (PubMed, Embase and Cochrane Library, Web of Science, Cochrane Center Register of Controlled trial) until 1 May 2023. We performed a meta-analysis of the eligible trials using RevMan5.4.1 software. Our analysis included six articles and five RCTs. The meta-analysis revealed significant differences in the incidences of LRTI [risk ratio (RR): 0.64; 95% confidence interval (CI): 0.43, 0.96; p = 0.03)] and severe LRTI (RR: 0.37; 95% CI: 0.18, 0.79; p = 0.01) between the vaccine group and the placebo group for newborns and infants. These differences were observed at 90, 120, and 150 days after birth (p = 0.003, p = 0.05, p = 0.02, p = 0.03, p = 0.009, p = 0.05). At 180 days after birth, there was a significant difference observed in the incidence of LRTI between the two groups (RR: 0.43; 95% CI: 0.21, 0.90; p = 0.02). The safety results showed a significant difference in the incidence of common adverse events between the two groups (RR: 1.08; 95% CI: 1.04, 1.12; p < 0.0001). However, there was no significant difference observed in the incidence of serious adverse events (RR: 1.05; 95% CI: 0.97, 1.15; p = 0.23), common and serious adverse events (RR: 1.02; 95% CI: 0.96, 1.10; p = 0.23), or common and serious adverse events among pregnant women and newborns and infants (RR: 0.98; 95% CI: 0.93, 1.04; p = 0.52). In conclusion, maternal RSV vaccination is an effective and safe immunization strategy for preventing LRTI in postpartum infants, with greater efficacy observed within the first 150 days after birth.
Collapse
Affiliation(s)
- Juan Ma
- Department of Neonatology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
- Department of Neonatology, SongShan General Hospital, Chongqing, China
| | - Long Chen
- Department of Neonatology, Women and Children’s Hospital of Chongqing Medical University (Chongqing Health Center for Women and Children), Chongqing, China
| | - ShiFang Tang
- Department of Neonatology, SongShan General Hospital, Chongqing, China
| | - Yuan Shi
- Department of Neonatology, Children’s Hospital of Chongqing Medical University, Chongqing, China
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, China
- Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| |
Collapse
|
4
|
Sacconnay L, De Smedt J, Rocha-Perugini V, Ong E, Mascolo R, Atas A, Vanden Abeele C, de Heusch M, De Schrevel N, David MP, Bouzya B, Stobbelaar K, Vanloubbeeck Y, Delputte PL, Mallett CP, Dezutter N, Warter L. The RSVPreF3-AS01 vaccine elicits broad neutralization of contemporary and antigenically distant respiratory syncytial virus strains. Sci Transl Med 2023; 15:eadg6050. [PMID: 37611082 DOI: 10.1126/scitranslmed.adg6050] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 07/25/2023] [Indexed: 08/25/2023]
Abstract
The RSVPreF3-AS01 vaccine, containing the respiratory syncytial virus (RSV) prefusion F protein and the AS01 adjuvant, was previously shown to boost neutralization responses against historical RSV strains and to be efficacious in preventing RSV-associated lower respiratory tract diseases in older adults. Although RSV F is highly conserved, variation does exist between strains. Here, we characterized variations in the major viral antigenic sites among contemporary RSV sequences when compared with RSVPreF3 and showed that, in older adults, RSVPreF3-AS01 broadly boosts neutralization responses against currently dominant and antigenically distant RSV strains. RSV-neutralizing responses are thought to play a central role in preventing RSV infection. Therefore, the breadth of RSVPreF3-AS01-elicited neutralization responses may contribute to vaccine efficacy against contemporary RSV strains and those that may emerge in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Kim Stobbelaar
- Department of Biomedical Sciences and Infla-Med Centre of Excellence, University of Antwerp, 2610 Wilrijk, Belgium
| | | | - Peter L Delputte
- Department of Biomedical Sciences and Infla-Med Centre of Excellence, University of Antwerp, 2610 Wilrijk, Belgium
| | | | | | | |
Collapse
|
5
|
Miller RJ, Mousa JJ. Structural basis for respiratory syncytial virus and human metapneumovirus neutralization. Curr Opin Virol 2023; 61:101337. [PMID: 37544710 PMCID: PMC10421620 DOI: 10.1016/j.coviro.2023.101337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 05/23/2023] [Accepted: 05/27/2023] [Indexed: 08/08/2023]
Abstract
Respiratory syncytial virus (RSV) and human metapneumovirus (hMPV) continue to be a global burden to infants, the elderly, and immunocompromised individuals. In the past ten years, there has been substantial progress in the development of new vaccine candidates and therapies against these viruses. These advancements were guided by the structural elucidation of the major surface glycoproteins for these viruses, the fusion (F) protein and attachment (G) protein. The identification of immunodominant epitopes on the RSV F and hMPV F proteins has expanded current knowledge on antibody-mediated immune responses, which has led to new approaches for vaccine and therapeutic development through the stabilization of pre-fusion constructs of the F protein and pre-fusion-specific monoclonal antibodies with high potency and efficacy. In this review, we describe structural characteristics of known antigenic sites on the RSV and hMPV proteins, their influence on the immune response, and current progress in vaccine and therapeutic development.
Collapse
Affiliation(s)
- Rose J Miller
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| | - Jarrod J Mousa
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA; Department of Biochemistry and Molecular Biology, Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA.
| |
Collapse
|
6
|
Ma L, Brecher M, Soufal A, Gaiotto T, Tian S, Chandramouli S, Dewar V, Ferrant L, Zhang M, Zhou X, Roy V. Structural interrogation of a trimeric prefusion RSV fusion protein vaccine candidate by a camelid nanobody. Vaccine 2023; 41:3308-3316. [PMID: 37085457 DOI: 10.1016/j.vaccine.2023.04.021] [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: 06/17/2022] [Revised: 03/31/2023] [Accepted: 04/05/2023] [Indexed: 04/23/2023]
Abstract
In the past decade, camelid nanobodies have been developed for multiple applications, including immuno-imaging, cancer immunotherapy, and antiviral therapeutics. Despite the prevalence of these approaches, nanobodies have rarely been used to assess the potency of vaccine antigen candidates, which are primarily based on mAb binding approaches. In this work, we demonstrate that a nanobody-based ELISA method is suitable for characterization of a leading respiratory syncytial virus (RSV) vaccine candidate, RSVPreF3. This nanobody, F-VHH-L66, compares similarly with AM14, an antibody well-known to be specific for the prefusion form of the RSV surface fusion glycoprotein, RSV F. ELISA assays based on F-VHH-L66 were specific for the trimeric, prefusion form of RSV F, the antigen conformation that best generates neutralizing antibodies. Additionally, the F-VHH-L66-based ELISA proved accurate, linear, and stability-indicating. Statistical analysis of 65 independent F-VHH-L66-based ELISA experiments indicated assay performance similar to that of ELISA assays based on AM14. Moreover, the binding kinetics of F-VHH-L66 to RSVPreF3 are comparable to those of AM14 when measured by surface plasmon resonance (SPR). Finally, F-VHH-L66 neutralized RSV(A) with similar efficacy as AM14; this bioactivity data further supports its use as an alternative to AM14 for pre-fusion specific structural characterization of RSVPreF3.
Collapse
Affiliation(s)
- Li Ma
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States
| | - Matthew Brecher
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States.
| | - Allison Soufal
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States
| | | | - Sai Tian
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States
| | - Sumana Chandramouli
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States
| | | | | | - Meng Zhang
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States
| | - Xianzhi Zhou
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States
| | - Varnika Roy
- GSK, Rockville Center for Vaccines Research, Rockville, MD 20850, United States.
| |
Collapse
|
7
|
Che Y, Gribenko AV, Song X, Handke LD, Efferen KS, Tompkins K, Kodali S, Nunez L, Prasad AK, Phelan LM, Ammirati M, Yu X, Lees JA, Chen W, Martinez L, Roopchand V, Han S, Qiu X, DeVincenzo JP, Jansen KU, Dormitzer PR, Swanson KA. Rational Design of a Highly Immunogenic Prefusion-Stabilized F Glycoprotein Antigen for a Respiratory Syncytial Virus Vaccine. Sci Transl Med 2023; 15:eade6422. [PMID: 37023209 DOI: 10.1126/scitranslmed.ade6422] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Abstract
Respiratory syncytial virus (RSV) is the leading, global cause of serious respiratory disease in infants and is an important cause of respiratory illness in older adults. No RSV vaccine is currently available. The RSV fusion (F) glycoprotein is a key antigen for vaccine development, and its prefusion conformation is the target of the most potent neutralizing antibodies. Here, we describe a computational and experimental strategy for designing immunogens that enhance the conformational stability and immunogenicity of RSV prefusion F. We obtained an optimized vaccine antigen after screening nearly 400 engineered F constructs. Through in vitro and in vivo characterization studies, we identified F constructs that are more stable in the prefusion conformation and elicit ~10-fold higher serum neutralizing titers in cotton rats than DS-Cav1. The stabilizing mutations of the lead construct (847) were introduced onto F glycoprotein backbones of strains representing the dominant circulating genotypes of the two major RSV subgroups, A and B. Immunization of cotton rats with a bivalent vaccine formulation of these antigens conferred complete protection against RSV challenge, with no evidence of disease enhancement. The resulting bivalent RSV prefusion F investigational vaccine has recently been shown to be efficacious against RSV disease in two pivotal phase 3 efficacy trials, one for passive protection of infants by immunization of pregnant women and the second for active protection of older adults by direct immunization.
Collapse
Affiliation(s)
- Ye Che
- Discovery Sciences, Pfizer Inc; Groton, CT 06340, USA
| | - Alexey V Gribenko
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| | - Xi Song
- Discovery Sciences, Pfizer Inc; Groton, CT 06340, USA
| | - Luke D Handke
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| | - Kari S Efferen
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| | - Kristin Tompkins
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| | - Srinivas Kodali
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| | - Lorna Nunez
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| | - A Krishna Prasad
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| | - Lynn M Phelan
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| | - Mark Ammirati
- Discovery Sciences, Pfizer Inc; Groton, CT 06340, USA
| | - Xiaodi Yu
- Discovery Sciences, Pfizer Inc; Groton, CT 06340, USA
| | - Joshua A Lees
- Discovery Sciences, Pfizer Inc; Groton, CT 06340, USA
| | - Wei Chen
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| | - Lyndsey Martinez
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| | - Vidia Roopchand
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| | - Seungil Han
- Discovery Sciences, Pfizer Inc; Groton, CT 06340, USA
| | - Xiayang Qiu
- Discovery Sciences, Pfizer Inc; Groton, CT 06340, USA
| | - John P DeVincenzo
- Children's Foundation Research Institute at Le Bonheur Children's Hospital; Memphis, TN 38103, USA
| | - Kathrin U Jansen
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| | - Philip R Dormitzer
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| | - Kena A Swanson
- Vaccine Research and Development, Pfizer Inc; Pearl River, NY 10965, USA
| |
Collapse
|
8
|
Hägglund S, Näslund K, Svensson A, Lefverman C, Enül H, Pascal L, Siltenius J, Holzhauer M, Delabouglise A, Österberg J, Alvåsen K, Olsson U, Eléouët JF, Riffault S, Taylor G, Rodriguez MJ, Garcia Duran M, Valarcher JF. Longitudinal study of the immune response and memory following natural bovine respiratory syncytial virus infections in cattle of different age. PLoS One 2022; 17:e0274332. [PMID: 36112582 PMCID: PMC9481050 DOI: 10.1371/journal.pone.0274332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/25/2022] [Indexed: 11/24/2022] Open
Abstract
Human and bovine respiratory syncytial virus (HRSV and BRSV) are closely genetically related and cause respiratory disease in their respective host. Whereas HRSV vaccines are still under development, a multitude of BRSV vaccines are used to reduce clinical signs. To enable the design of vaccination protocols to entirely stop virus circulation, we aimed to investigate the duration, character and efficacy of the immune responses induced by natural infections. The systemic humoral immunity was monitored every two months during two years in 33 dairy cattle in different age cohorts following a natural BRSV outbreak, and again in selected individuals before and after a second outbreak, four years later. Local humoral and systemic cellular responses were also monitored, although less extensively. Based on clinical observations and economic losses linked to decreased milk production, the outbreaks were classified as moderate. Following the first outbreak, most but not all animals developed neutralising antibody responses, BRSV-specific IgG1, IgG2 and HRSV F- and HRSV N-reactive responses that lasted at least two years, and in some cases at least four years. In contrast, no systemic T cell responses were detected and only weak IgA responses were detected in some animals. Seronegative sentinels remained negative, inferring that no new infections occurred between the outbreaks. During the second outbreak, reinfections with clinical signs and virus shedding occurred, but the signs were milder, and the virus shedding was significantly lower than in naïve animals. Whereas the primary infection induced similar antibody titres against the prefusion and the post fusion form of the BRSV F protein, memory responses were significantly stronger against prefusion F. In conclusion, even if natural infections induce a long-lasting immunity, it would probably be necessary to boost memory responses between outbreaks, to stop the circulation of the virus and limit the potential role of previously infected adult cattle in the chain of BRSV transmission.
Collapse
Affiliation(s)
- Sara Hägglund
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
- * E-mail:
| | - Katarina Näslund
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Anna Svensson
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Cecilia Lefverman
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Hakan Enül
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Leonore Pascal
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Jari Siltenius
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Menno Holzhauer
- Ruminant Health Department Royal GD Animal Health, Deventer, The Netherlands
| | - Alexis Delabouglise
- CIRAD, UMR ASTRE, F-34398 Montpellier, France and UMR ASTRE, Univ Montpellier, CIRAD, INRAE, Montpellier, France
| | - Julia Österberg
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Karin Alvåsen
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Ulf Olsson
- Department of Energy and Technology, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | | | - Sabine Riffault
- Université Paris-Saclay, UVSQ, INRAE, VIM, Jouy-en-Josas, France
| | - Geraldine Taylor
- The Pirbright Institute, Ash Road, Pirbright, Woking, Surrey, United Kingdom
| | | | | | - Jean François Valarcher
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| |
Collapse
|