1
|
Cnossen VM, van Leeuwen RP, Mazur NI, Vernhes C, ten Voorde W, Burggraaf J, de Visser SJ, Roestenberg M, Kamerling IMC. From setbacks to success: lessons from the journey of RSV vaccine development. Ther Adv Vaccines Immunother 2024; 12:25151355241308305. [PMID: 39711948 PMCID: PMC11660060 DOI: 10.1177/25151355241308305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 12/03/2024] [Indexed: 12/24/2024] Open
Abstract
Respiratory syncytial virus (RSV) causes high worldwide infant mortality, as well as a high disease burden in the elderly. Efforts in vaccine development over the past 60 years have recently delivered three approved vaccines and two monoclonal antibodies (mAbs). Looking back at the eventful history of RSV vaccine development, several factors can be identified that have hampered the developmental pathway, including the occurrence of enhanced RSV disease (ERD) in the first vaccine attempt and the difficulty in characterizing and stabilizing the pre-fusion F protein as a vaccine target. Moreover, the need for large trials to test vaccine efficacy, usually done late in development, and the lack of a correlate of protection (CoP) result in significant uncertainties in RSV vaccine development. The use of controlled human infection models (CHIMs) may provide a solution for some of these problems: through swift, cost-efficient and closely monitored assessment of vaccine safety and efficacy in early clinical phases, vaccines can either 'fail fast' or show results supporting further investments. Moreover, CHIMs facilitate the assessment of disease and could assist in the identification of a CoP supporting late-stage development. Although some factors may affect translatability to real-world vaccine efficacy, CHIMs can support the clinical development pathway in various ways. We advocate for, and demonstrate, a conceptual and rational design of RSV vaccine development. Assessing protective efficacy early on would result in the most cost-efficient pathway and identification of target populations should be done as early as possible. For RSV, elderly individuals and people in low- and middle-income countries are high-impact populations for RSV prevention. While RSV immunization is now available in certain regions, global access is not accomplished yet, and worldwide prevention does not seem within reach. Quick and cost-effective assessments of candidates currently in the pipeline could contribute to future successes in the battle against RSV.
Collapse
Affiliation(s)
- Victor M. Cnossen
- Centre for Human Drug Research, Zernikedreef 8, 2333 CL Leiden, The Netherlands
| | | | | | - Charlotte Vernhes
- Vaccines Europe, European Federation of Pharmaceutical Industries and Associations, Brussels, Belgium
| | | | | | - Saco J. de Visser
- Centre for Future Affordable & Sustainable Therapy Development (FAST), The Hague, The Netherlands
| | - Meta Roestenberg
- Centre for Human Drug Research, Leiden, The Netherlands
- Leiden University Centre for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, The Netherlands
| | - Ingrid M. C. Kamerling
- Centre for Human Drug Research, Leiden, The Netherlands
- Leiden University Centre for Infectious Diseases (LU-CID), Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
2
|
Vijayan A, Vogels R, Groppo R, Jin Y, Khan S, Van Kampen M, Jorritsma S, Boedhoe S, Baert M, van Diepen H, Kuipers H, Serroyen J, Del Valle JR, Broman A, Nguyen L, Ray S, Jarai B, Arora J, Lifton M, Mildenberg B, Morton G, Santra S, Grossman TR, Schuitemaker H, Custers J, Zahn R. A self-amplifying RNA RSV prefusion-F vaccine elicits potent immunity in pre-exposed and naïve non-human primates. Nat Commun 2024; 15:9884. [PMID: 39543172 PMCID: PMC11564874 DOI: 10.1038/s41467-024-54289-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024] Open
Abstract
Newly approved subunit and mRNA vaccines for respiratory syncytial virus (RSV) demonstrate effectiveness in preventing severe disease, with protection exceeding 80% primarily through the generation of antibodies. An alternative vaccine platform called self-amplifying RNA (saRNA) holds promise in eliciting humoral and cellular immune responses. We evaluate the immunogenicity of a lipid nanoparticle (LNP)-formulated saRNA vaccine called SMARRT.RSV.preF, encoding a stabilized form of the RSV fusion protein, in female mice and in non-human primates (NHPs) that are either RSV-naïve or previously infected. Intramuscular vaccination with SMARRT.RSV.preF vaccine induces RSV neutralizing antibodies and cellular responses in naïve mice and NHPs. Importantly, a single dose of the vaccine in RSV pre-exposed NHPs elicits a dose-dependent anamnestic humoral immune response comparable to a subunit RSV preF vaccine. Notably, SMARRT.RSV.preF immunization significantly increases polyfunctional RSV.F specific memory CD4+ and CD8+ T-cells compared to RSV.preF protein vaccine. Twenty-four hours post immunization with SMARRT.RSV.preF, there is a dose-dependent increase in the systemic levels of inflammatory and chemotactic cytokines associated with the type I interferon response in NHPs, which is not observed with the protein vaccine. We identify a cluster of analytes including IL-15, TNFα, CCL4, and CXCL10, whose levels are significantly correlated with each other after SMARRT.RSV.preF immunization. These findings suggest saRNA vaccines have the potential to be developed as a prophylactic RSV vaccine based on innate, cellular, and humoral immune profiles they elicit.
Collapse
Affiliation(s)
- Aneesh Vijayan
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands.
- Artemis Bioservices, Delft, The Netherlands.
| | - Ronald Vogels
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
| | - Rachel Groppo
- Johnson & Johnson Innovative Medicine, La Jolla, USA
| | - Yi Jin
- Johnson & Johnson Innovative Medicine, La Jolla, USA
| | - Selina Khan
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
- Oncode Accelerator Foundation, Utrecht, The Netherlands
| | | | - Sytze Jorritsma
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
| | - Satish Boedhoe
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
| | - Miranda Baert
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
- LUCID research centre, Leiden Medical University, Leiden, The Netherlands
| | | | - Harmjan Kuipers
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
| | - Jan Serroyen
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
| | | | - Ann Broman
- Johnson & Johnson Innovative Medicine, La Jolla, USA
| | - Lannie Nguyen
- Johnson & Johnson Innovative Medicine, La Jolla, USA
| | - Sayoni Ray
- Johnson & Johnson Innovative Medicine, La Jolla, USA
| | - Bader Jarai
- Johnson & Johnson Innovative Medicine, La Jolla, USA
| | - Jayant Arora
- Johnson & Johnson Innovative Medicine, La Jolla, USA
| | - Michelle Lifton
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School., Boston, USA
| | - Benjamin Mildenberg
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School., Boston, USA
| | - Georgeanna Morton
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School., Boston, USA
| | - Sampa Santra
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School., Boston, USA
| | | | | | - Jerome Custers
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands
| | - Roland Zahn
- Janssen Vaccines and Prevention B.V., Leiden, The Netherlands.
| |
Collapse
|
3
|
Coindy EL, Efstathiou C, Talwar S, Moureau A, Vernhes C, Openshaw PJM, Thwaites RS. Antibody-mediated protection against respiratory syncytial virus in children. Eur Respir Rev 2024; 33:240106. [PMID: 39384305 PMCID: PMC11462297 DOI: 10.1183/16000617.0106-2024] [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: 05/17/2024] [Accepted: 07/31/2024] [Indexed: 10/11/2024] Open
Abstract
Respiratory syncytial virus (RSV) is a major global pathogen, causing lower respiratory tract disease in at-risk populations including young children. Antibodies form a crucial layer of protection from RSV disease, particularly in immunologically naïve infants. Such antibodies are derived from the mother via transplacental transfer and breast milk, but may be particularly low in high-risk infants such as those born preterm. Maternally derived antibodies can now be supplemented by the administration of anti-RSV monoclonal antibodies, while a rising wave of maternal and paediatric vaccine strategies are approaching. The implementation of these prophylactics may profoundly decrease the healthcare burden of RSV. In this article, we review the role of antibody-mediated immunity in protecting children from RSV. We focus on maternally derived antibodies as the main source of protection against RSV and study factors that influence the scale of this transfer. The role of passive and active prophylactic approaches in protecting infants against RSV are discussed and knowledge gaps in our understanding of antibody-mediated protection against RSV are identified.
Collapse
Affiliation(s)
- Emma L Coindy
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Shubha Talwar
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | | | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
4
|
Langley JM, Nolan TM, Rämet M, Richmond PC, Rosário Filho N, Haazen W, van den Berg SPH, Williams K, Bastian AR, Omoruyi E, Williams Durkin J, Salisch N, Van Geet G, van Duijnhoven W, Heijnen E, Callendret B. A Phase 1/2a Study Evaluating Safety and Immunogenicity of Ad26.RSV.preF in RSV-seronegative Toddlers Aged 12-24 Months. Open Forum Infect Dis 2024; 11:ofae453. [PMID: 39220658 PMCID: PMC11365064 DOI: 10.1093/ofid/ofae453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Background Respiratory syncytial virus (RSV) causes serious illness in children. The Ad26.RSV.preF vaccine candidate was immunogenic with acceptable safety in a phase 1/2a study of RSV-seropositive children. Here, we assessed its safety and immunogenicity in RSV-seronegative children. Methods In this randomized, observer-blinded, placebo-controlled, phase 1/2a study (NCT03606512; https://www.clinicaltrials.gov/ct2/show/NCT03606512), RSV-seronegative toddlers aged 12-24 months received Ad26.RSV.preF (2.5 × 1010 viral particles) or placebo on days 1, 29, and 57 (a meningococcal vaccine [Nimenrix] could substitute for day 57 placebo). Primary endpoints were solicited local and systemic adverse events (AEs; 7 days after each vaccination), unsolicited AEs (28 days postvaccination), and serious AEs (first vaccination until study end). Participants were monitored for RSV-respiratory tract infection to assess infection rates and for severe RSV-lower respiratory tract infection as an indication of enhanced disease. RSV-A2 neutralizing, RSV (A and B) preF binding, and RSV postF immunoglobulin G-binding antibodies were evaluated on days 1 (predose), 8, and 85, and after RSV season 1. Results Thirty-eight participants were enrolled and vaccinated (Ad26.RSV.preF, n = 20; placebo, placebo/Nimenrix, n = 18). Solicited AEs were more common following Ad26.RSV.preF than placebo; most were mild/moderate. No vaccine-related serious AEs were reported. Five of 19 participants receiving Ad26.RSV.preF and 2/18 receiving placebo or placebo/Nimenrix had confirmed RSV-respiratory tract infection or RSV-associated otitis media; none were considered severe. At the final season 1 study visit, most Ad26.RSV.preF recipients had ≥2-fold increases from baseline in RSV-A2 neutralizing, RSV A and B preF binding, and RSV postF antibodies. Conclusions Ad26.RSV.preF was well tolerated and immunogenic in RSV-seronegative toddlers.
Collapse
Affiliation(s)
- Joanne M Langley
- Canadian Center for Vaccinology, Dalhousie University, IWK and Nova Scotia Health, Halifax, Nova Scotia, Canada
| | - Terry M Nolan
- Department of Infectious Diseases, Peter Doherty Institute for Infection and Immunity at The University of Melbourne, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Mika Rämet
- FVR – Finnish Vaccine Research Ltd., and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Peter C Richmond
- Wesfarmers Centre of Vaccines and Infectious Diseases, Telethon Kids Institute, University of Western Australia, Perth, Western Australia, Australia
- Discipline of Paediatrics, School of Medicine, University of Western Australia, Perth, Western Australia, Australia
| | - Nelson Rosário Filho
- Division of Allergy and Immunology, Complexo Hospital de Clínicas da Universidade Federal do Paraná, Curitiba, Brazil
| | - Wouter Haazen
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | | | | | | | | | | | - Nadine Salisch
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | | | | | - Esther Heijnen
- Janssen Vaccines & Prevention B.V., Leiden, The Netherlands
| | | |
Collapse
|
5
|
Ma J, Gong T, Luo T, Li S, Zhong L, Zhao X, Mei C, Bu H, Jia Z, Kuang X, Wang X, Fu Z, Tian D. Exacerbated lung inflammation in offspring with high maternal antibody levels following secondary RSV exposure. Front Immunol 2024; 15:1377374. [PMID: 38745662 PMCID: PMC11091276 DOI: 10.3389/fimmu.2024.1377374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024] Open
Abstract
Respiratory syncytial virus (RSV) is the primary cause of bronchiolitis-related hospitalizations among children under 5 years of age, with reinfection being common throughout life. Maternal vaccination has emerged as a promising strategy, delivering elevated antibody levels to newborns for immediate protection. However, limited research has explored the protective efficacy of maternal antibodies (matAbs) against secondary RSV infections in offspring. To address this gap, we employed a mouse model of maternal RSV vaccination and secondary infection of offspring to evaluate lung pathology following RSV reinfection in mice with varying levels of maternal antibody (matAb). Additionally, we aimed to investigate the potential causes of exacerbated lung inflammation in offspring with high matAb levels following secondary RSV exposure. Our findings revealed that offspring with elevated levels of maternal pre-F antibody demonstrated effective protection against lung pathology following the initial RSV infection. However, this protection was compromised upon reinfection, manifesting as heightened weight loss, exacerbated lung pathology, increased expression of RSV-A N genes, eosinophilia, enhanced IL-5, IL-13, MUC5AC, and eosinophils Major Basic Protein (MBP) production in lung tissue compared to offspring lacking matAbs. Importantly, these unexpected outcomes were not attributed to antibody-dependent enhancement (ADE) resulting from declining matAb levels over time. Notably, our findings showed a decline in secretory IgA (sIgA), mucosal IgA, and mucosal IgG levels in offspring with high matAb levels post-primary RSV challenge. We propose that this decline may be a critical factor contributing to the ineffective protection observed during secondary RSV exposure. Overall, these findings offer valuable insights into maternal vaccination against RSV, contributing to a comprehensive understanding and mitigation of potential risks associated with maternal RSV vaccination.
Collapse
Affiliation(s)
- Jinhua Ma
- Department of Respiratory Medicine Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Ting Gong
- Department of Respiratory Medicine Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Tingting Luo
- Department of Respiratory Medicine Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Shuanglian Li
- Department of Respiratory Medicine Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Li Zhong
- Department of Respiratory Medicine Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Xin Zhao
- Department of Respiratory Medicine Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Chenghao Mei
- Department of Respiratory Medicine Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Huaqin Bu
- Department of Respiratory Medicine Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Zhenxing Jia
- Department of mAbs Discovery, Zhuhai Trinomab Pharmaceutical Co., Ltd, Zhuhai, China
| | - Xiaohu Kuang
- Department of mAbs Discovery, Zhuhai Trinomab Pharmaceutical Co., Ltd, Zhuhai, China
| | - Xiaoli Wang
- Department of mAbs Discovery, Zhuhai Trinomab Pharmaceutical Co., Ltd, Zhuhai, China
| | - Zhou Fu
- Department of Respiratory Medicine Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
| | - Daiyin Tian
- Department of Respiratory Medicine Children’s Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China
- Department of Respiratory Medicine, Yibin Hospital Affiliated to Children’s Hospital of Chongqing Medical University, Yibin, China
| |
Collapse
|
6
|
Honda-Okubo Y, Sakala IG, André G, Tarbet EB, Hurst BL, Petrovsky N. An Advax-CpG55.2 adjuvanted recombinant hemagglutinin vaccine provides immunity against H7N9 influenza in adult and neonatal mice. Vaccine 2023; 41:5592-5602. [PMID: 37532610 DOI: 10.1016/j.vaccine.2023.07.061] [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: 05/30/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/04/2023]
Abstract
There is a major unmet need for strategies to improve the immunogenicity and effectiveness of pandemic influenza vaccines, particularly in poor responder populations such as neonates. Recombinant protein approaches to pandemic influenza offer advantages over more traditional inactivated virus approaches, as they are free of problems such as egg adaptation or need for high level biosecurity containment for manufacture. However, a weakness of recombinant proteins is their low immunogenicity. We asked whether the use of an inulin polysaccharide adjuvant (Advax) alone or combined with a TLR9 agonist (CpG55.2) would enhance the immunogenicity and protection of a recombinant hemagglutinin vaccine against H7N9 influenza (rH7HA), including in neonatal mice. Advax adjuvant induced predominantly IgG1 responses against H7HA, whereas Advax-CpG55.2 adjuvant also induced IgG2a, IgG2b and IgG3 responses, consistent with the TLR9 agonist component inducing a Th1 bias. Advax-CpG55.2 adjuvanted rH7HA induced high serum neutralizing antibody titers in adult mice. In newborns it similarly overcame immune hypo-responsiveness and enhanced serum anti-rH7HA IgG levels in 7-day-old BALB/C and C57BL/6 mice. Immunized adult mice were protected against a lethal H7N9 virus challenge. When formulated with Advax-CpG55.2 adjuvant, greater protection was seen with rH7HA than with inactivated H7 whole virus antigen. Advax-CpG55.2 adjuvanted rH7HA represents a promising influenza vaccine platform for further development.
Collapse
Affiliation(s)
- Yoshikazu Honda-Okubo
- Vaxine Pty Ltd, Bedford Park, Adelaide, SA 5042, Australia; Flinders University, Bedford Park, Adelaide, SA 5042, Australia
| | - Isaac G Sakala
- Vaxine Pty Ltd, Bedford Park, Adelaide, SA 5042, Australia; Flinders University, Bedford Park, Adelaide, SA 5042, Australia
| | | | - E Bart Tarbet
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, 5600 Old Main Hill, Utah State University, Logan, UT 84322, USA
| | - Brett L Hurst
- Institute for Antiviral Research, Department of Animal, Dairy, and Veterinary Sciences, 5600 Old Main Hill, Utah State University, Logan, UT 84322, USA
| | | |
Collapse
|
7
|
Kherfan T, Sallam M. Prospective Attitudes towards Respiratory Syncytial Virus (RSV) Vaccination: Validation of a Survey Instrument among Young Females in Jordan Pending Vaccine Authorization. Vaccines (Basel) 2023; 11:1386. [PMID: 37631954 PMCID: PMC10459998 DOI: 10.3390/vaccines11081386] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/11/2023] [Accepted: 08/18/2023] [Indexed: 08/28/2023] Open
Abstract
In May 2023, the U.S. FDA advisors endorsed Pfizer's pregnancy-administered vaccine (branded ABRYSVO) to protect infants from respiratory syncytial virus (RSV) infection. Vaccination can reduce the burden of RSV-related respiratory disease, with previous studies showing its substantial medical and financial burden in Jordan. However, pregnant women may exhibit hesitancy to get vaccinated due to concerns about potential risks to themselves or their fetuses. This study aimed to assess the acceptance of the RSV vaccine among young females and identify the determinants influencing their decision using a newly constructed instrument. A survey instrument was developed and validated, comprising 26 items to measure RSV vaccine acceptance. A cross-sectional study design was employed, with data collection from a sample of females aged 18 to 45 residing in Jordan during 5-6 July 2023, using a convenient approach via an online distributed questionnaire. The final study sample comprised 315 respondents, with 67.6% who have heard of RSV before the study. If the vaccine was safe, effective, and provided freely, 70.2% showed willingness to get the RSV vaccine, 15.2% resisted, and 14.6% were hesitant. Principal component analysis identified six internally consistent sub-scales with the following suggested themes: Advice, Burden, Conspiracy, Dangers, Efficiency, and Fear, comprising 21 items collectively as assigned as the "ABCDEF" scale for RSV vaccine acceptance. RSV vaccine acceptance in this study was associated with the advice and fear constructs. The validated survey instrument successfully captured important determinants of RSV vaccine acceptance among young females. RSV vaccine promotion efforts should focus on the following: enhancing vaccine education, improving trust in healthcare institutions and providers, reducing burdens through resolving cost issues and focusing on the role of social support, addressing safety concerns, and tailoring communication strategies to effectively promote the benefits of the vaccine. These insights can inform public health policies and interventions aiming to promote RSV vaccination and mitigate the burden of RSV-related diseases among infants. Follow-up studies are recommended with pregnant women as the target group to assess their attitude towards RSV vaccination and to confirm the validity of the conceived ABCDEF survey instrument.
Collapse
Affiliation(s)
- Tleen Kherfan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Malik Sallam
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman 11942, Jordan
- Department of Translational Medicine, Faculty of Medicine, Lund University, 22184 Malmö, Sweden
| |
Collapse
|
8
|
Leroux-Roels I, Bruhwyler J, Stergiou L, Sumeray M, Joye J, Maes C, Lambert PH, Leroux-Roels G. Double-Blind, Placebo-Controlled, Dose-Escalating Study Evaluating the Safety and Immunogenicity of an Epitope-Specific Chemically Defined Nanoparticle RSV Vaccine. Vaccines (Basel) 2023; 11:vaccines11020367. [PMID: 36851245 PMCID: PMC9967611 DOI: 10.3390/vaccines11020367] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND V-306 is a virus-like particle-based vaccine candidate displaying respiratory syncytial virus (RSV) F site II protein mimetics (FsIIm) as an antigenic epitope. METHODS This was a randomized, placebo-controlled, double-blind, dose-escalating, first-in-human study, conducted in 60 women aged 18-45 years. Twenty subjects per cohort (15 vaccine and five placebo) received two V-306 intramuscular administrations on Days 0 and 56 at 15 µg, 50 µg, or 150 µg. Safety and immunogenicity were assessed after each vaccination and for 1 year in total. RESULTS V-306 was safe and well tolerated at all dose levels, with no increase in reactogenicity and unsolicited adverse events between the first and second administrations. At 50 µg and 150 µg, V-306 induced an increase in FsIIm-specific immunoglobulin G (IgG) titers, which lasted at least 4 months. This did not translate into an increase in RSV-neutralizing antibody titers, which were already high at baseline. No increase in the anti-F protein-specific IgG titers was observed, which were also high in most subjects at baseline due to past natural infections. CONCLUSIONS V-306 was safe and well-tolerated. Future modifications of the vaccine and assay conditions will likely improve the results of vaccination.
Collapse
Affiliation(s)
- Isabel Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University Hospital, Corneel Heymanslaan 10, B-9000 Ghent, Belgium
| | - Jacques Bruhwyler
- Expert Clinical Services Organization (ECSOR) sa/nv, Rue de la Station 78, B-1630 Linkebeek, Belgium
| | - Lilli Stergiou
- Virometix AG, Wagistrasse 14, 8952 Schlieren, Switzerland
- Correspondence: ; Tel.: +41-4343-38660
| | - Mark Sumeray
- Virometix AG, Wagistrasse 14, 8952 Schlieren, Switzerland
| | - Jasper Joye
- Center for Vaccinology (CEVAC), Ghent University Hospital, Corneel Heymanslaan 10, B-9000 Ghent, Belgium
| | - Cathy Maes
- Center for Vaccinology (CEVAC), Ghent University Hospital, Corneel Heymanslaan 10, B-9000 Ghent, Belgium
| | - Paul-Henri Lambert
- Department of Paediatrics, Gynecology and Obstetrics, University of Geneva, Rue du Général Dufour 24, 1211 Geneva, Switzerland
| | - Geert Leroux-Roels
- Center for Vaccinology (CEVAC), Ghent University Hospital, Corneel Heymanslaan 10, B-9000 Ghent, Belgium
| |
Collapse
|
9
|
Abstract
Nirsevimab (Beyfortus®), a long-acting intramuscular recombinant neutralising human IgG1ĸ monoclonal antibody to the prefusion conformation of the respiratory syncytial virus (RSV) F protein that has been modified with a triple amino acid substitution in the Fc region to extend the serum half-life, is being jointly developed by AstraZeneca and Sanofi for the prevention of RSV disease. The extended serum half-life allows administration of nirsevimab as a single dose to cover the RSV season. Nirsevimab was approved in the EU on 3 November 2022 and in the UK on 7 November 2022 for the prevention of RSV lower respiratory tract disease in neonates and infants during their first RSV season. This article summarizes the milestones in the development of nirsevimab leading to this first approval for the prevention of RSV disease in all infants.
Collapse
Affiliation(s)
- Susan J Keam
- Springer Nature, Mairangi Bay, Private Bag 65901, Auckland, 0754, New Zealand.
| |
Collapse
|
10
|
Sevilla JP. Immunization, not vaccination: monoclonal antibodies for infant RSV prevention and the US vaccines for children program. J Med Econ 2023; 26:991-997. [PMID: 37498791 DOI: 10.1080/13696998.2023.2242169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 07/29/2023]
Abstract
In the US, RSV imposes significant burdens on infants, households, and the health system. Yet the only licensed immunization is accessible to only certain risk groups comprising 2% of the infant population, leaving the remaining 98% unprotected. An effective immunization for all infants is a significant public health priority. One possible solution is the FDA-approved monoclonal antibody nirsevimab, which recent evidence suggests is safe and effective in preventing RSV in all infants, and which is currently being considered for inclusion in the pediatric immunization schedule and the federal Vaccines for Children (VFC) program. But the question arises whether passive immunization products like nirsevimab ought to be eligible for the VFC, which nominally and traditionally centers on vaccines providing active immunity. Addressing this is urgent because VFC inclusion will be decided on imminently. I argue there are strong policy grounds, i.e., reasons grounded in the ultimate health system goals of maximizing population health or social welfare subject to resource constraints, not to exclude passive immunization from VFC eligibility. Active and passive immunizations both provide adaptive immunity and can therefore produce qualitatively similar effects on risks of infection, disease, and transmission; on disease severity and duration; and on health, welfare, and health resource use. The distinction between active and passive immunization does not intrinsically matter since what matters for the attainment of health system goals is the extent of immunity conferred, not whether immunity is active or passive. Nor can passivity be considered a useful proxy for conferring a lesser extent of immunity, since no such proxy is needed (existing valuation methods can cope with variations in product attributes), and it is a poor proxy (passive immunizations can be better for individuals with impaired immune systems and can have comparable effectiveness durations and economic value as vaccines).
Collapse
|
11
|
Characterisation of RSV Fusion Proteins from South African Patients with RSV Disease, 2019 to 2020. Viruses 2022; 14:v14112321. [PMID: 36366419 PMCID: PMC9698603 DOI: 10.3390/v14112321] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 11/07/2022] Open
Abstract
Respiratory syncytial virus (RSV) is classified into RSV-A and RSV-B, which are further classified into genotypes based on variability in the G gene. The fusion (F) protein is highly conserved; however, variability within antigenic sites has been reported. This study aimed to characterise F proteins from RSV strains detected in South Africa from 2019 to 2020. Patients of all ages, from whom respiratory samples were submitted to the National Health Laboratory Service at Charlotte Maxeke Johannesburg Academic Hospital, South Africa during 2019 to 2020, were included. Complete RSV F genes were amplified for next-generation sequencing. MEGA X software was used for phylogenetic analysis. The overall prevalence of RSV was 5.8% (101/1734). Among 101 RSV positive samples only 69.3% (70/101) were available for characterization of the RSV F protein gene. Among cases included for F gene characterisation, viral co-infections were observed in 50% (35/70) and 25.7% (18/70) were admitted to intensive care units (ICU). About 74.2% (23/31) of F gene sequences cluster with other African NA1/ON1 genotypes. At antigenic site I, the V384I mutation was replaced by V384T in South African strains. The S275F mutation was seen in a single South African strain. The N120 N-linked glycosylation site was present in 25.8% (8/31) of RSV-A F proteins described in this study. For the first time, we detected the rare S275F mutation that is associated with palivizumab resistance.
Collapse
|
12
|
Bont L, Weil Olivier C, Herting E, Esposito S, Navarro Alonso JA, Lega F, Mader S, Morioka I, Shen K, Syrogiannopoulos GA, Faust SN, Bozzola E. The assessment of future RSV immunizations: How to protect all infants? Front Pediatr 2022; 10:981741. [PMID: 36016878 PMCID: PMC9396232 DOI: 10.3389/fped.2022.981741] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Affiliation(s)
- Louis Bont
- Department of Paediatrics, Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, Netherlands
| | | | - Egbert Herting
- Department of Pediatrics, University of Lübeck, Lübeck, Germany
| | - Susanna Esposito
- Department of Medicine and Surgery, Pediatric Clinic, Pietro Barilla Children's Hospital, University of Parma, Parma, Italy
| | | | - Federico Lega
- Department of Biomedical Science, Research Center in Health Administration, University of Milan, Milan, Italy
| | - Silke Mader
- European Foundation for the Care of Newborn Infants (EFCNI), Munich, Germany
| | - Ichiro Morioka
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Kunling Shen
- Department of Respiratory Medicine, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | | | - Saul N. Faust
- Faculty of Medicine, Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
- National Institute for Health Research (NIHR) Southampton Clinical Research Facility, NIHR Southampton Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
| | - Elena Bozzola
- Pediatric and Infectious Diseases Unit, Bambino Gesù Children Hospital, Rome, Italy
| |
Collapse
|
13
|
Esposito S, Abu Raya B, Baraldi E, Flanagan K, Martinon Torres F, Tsolia M, Zielen S. RSV Prevention in All Infants: Which Is the Most Preferable Strategy? Front Immunol 2022; 13:880368. [PMID: 35572550 PMCID: PMC9096079 DOI: 10.3389/fimmu.2022.880368] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/28/2022] [Indexed: 11/30/2022] Open
Abstract
Respiratory syncytial virus (RSV) causes a spectrum of respiratory illnesses in infants and young children that may lead to hospitalizations and a substantial number of outpatient visits, which result in a huge economic and healthcare burden. Most hospitalizations happen in otherwise healthy infants, highlighting the need to protect all infants against RSV. Moreover, there is evidence on the association between early-life RSV respiratory illness and recurrent wheezing/asthma-like symptoms As such, RSV is considered a global health priority. However, despite this, the only prevention strategy currently available is palivizumab, a monoclonal antibody (mAb) indicated in a subset of preterm infants or those with comorbidities, hence leaving the majority of the infant population unprotected against this virus. Therefore, development of prevention strategies against RSV for all infants entering their first RSV season constitutes a large unmet medical need. The aim of this review is to explore different immunization approaches to protect all infants against RSV. Prevention strategies include maternal immunization, immunization of infants with vaccines, immunization of infants with licensed mAbs (palivizumab), and immunization of infants with long-acting mAbs (e.g., nirsevimab, MK-1654). Of these, palivizumab use is restricted to a small population of infants and does not offer a solution for all-infant protection, whereas vaccine development in infants has encountered various challenges, including the immaturity of the infant immune system, highlighting that future pediatric vaccines will most likely be used in older infants (>6 months of age) and children. Consequently, maternal immunization and immunization of infants with long-acting mAbs represent the two feasible strategies for protection of all infants against RSV. Here, we present considerations regarding these two strategies covering key areas which include mechanism of action, "consistency" of protection, RSV variability, duration of protection, flexibility and optimal timing of immunization, benefit for the mother, programmatic implementation, and acceptance of each strategy by key stakeholders. We conclude that, based on current data, immunization of infants with long-acting mAbs might represent the most effective approach for protecting all infants entering their first RSV season.
Collapse
Affiliation(s)
- Susanna Esposito
- Pediatric Clinic, Pietro Barilla Children’s Hospital, University of Parma, Parma, Italy
| | - Bahaa Abu Raya
- Department of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Eugenio Baraldi
- Neonatal Intensive Care Unit, Department of Woman’s and Child’s Health, Padova University Hospital, Padova, Italy
| | - Katie Flanagan
- School of Medicine, Faculty of Health Sciences, University of Tasmania, Launceston, TAS, Australia
- School of Health and Biomedical Science, RMIT University, Melbourne, VIC, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
- Tasmanian Vaccine Trial Centre, Clifford Craig Foundation, Launceston General Hospital, Launceston, TAS, Australia
| | - Federico Martinon Torres
- Genetics, Vaccines, Infections and Pediatrics Research group (GENVIP), Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Maria Tsolia
- Second Department of Pediatrics, National and Kapodistrian University of Athens, “A&P Kyriakou” Children’s Hospital, Athens, Greece
| | - Stefan Zielen
- Department for Children and Adolescents, Division of Allergology, Pulmonology and Cystic Fibrosis, Goethe-University Hospital, Frankfurt am Main, Germany
| |
Collapse
|
14
|
Russell MS, Thulasi Raman SN, Gravel C, Zhang W, Pfeifle A, Chen W, Van Domselaar G, Safronetz D, Johnston M, Sauve S, Wang L, Rosu-Myles M, Cao J, Li X. Single Immunization of a Vaccine Vectored by a Novel Recombinant Vaccinia Virus Affords Effective Protection Against Respiratory Syncytial Virus Infection in Cotton Rats. Front Immunol 2021; 12:747866. [PMID: 34603336 PMCID: PMC8484905 DOI: 10.3389/fimmu.2021.747866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/27/2021] [Indexed: 11/13/2022] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of respiratory infections worldwide and disease management measures are hampered by the lack of a safe and effective vaccine against the infection. We constructed a novel recombinant RSV vaccine candidate based on a deletion mutant vaccinia virus platform, in that the host range genes E3L and K3L were deleted (designated as VACVΔE3LΔK3L) and a poxvirus K3L ortholog gene was used as a marker for the rapid and efficient selection of recombinant viruses. The safety of the modified vaccinia virus was investigated by intranasal administration of BALB/c mice with the modified vaccinia vector using a dose known to be lethal in the wild-type Western Reserve. Only a minor loss of body weight by less than 5% and mild pulmonary inflammation were observed, both of which were transient in nature following nasal administration of the high-dose modified vaccinia virus. In addition, the viruses were cleared from the lung in 2 days with no viral invasions of the brain and other vital organs. These results suggest that the virulence of the virus has been essentially abolished. We then investigated the efficiency of the vector for the delivery of vaccines against RSV through comparison with another RSV vaccine delivered by the widely used Modified Vaccinia virus Ankara (MVA) backbone. In the cotton rats, we found a single intramuscular administration of VACVΔE3LΔK3L-vectored vaccine elicited immune responses and protection at a level comparable to the MVA-vectored vaccine against RSV infection. The distinct features of this novel VACV vector, such as an E3L deletion for attenuation and a K3L ortholog for positive selection and high efficiency for vaccine delivery, could provide unique advantages to the application of VACV as a platform for vaccine development.
Collapse
Affiliation(s)
- Marsha S Russell
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Sathya N Thulasi Raman
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Caroline Gravel
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Wanyue Zhang
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs 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
| | - Annabelle Pfeifle
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs 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
| | - Wangxue Chen
- National Research Council of Canada, Human Health Therapeutics, Ottawa, ON, Canada
| | - Gary Van Domselaar
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - David Safronetz
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Michael Johnston
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada.,Department of Chemistry, Carleton University, Ottawa, ON, Canada
| | - Simon Sauve
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs Directorate, Health Products and Food Branch (HPFB), Health Canada and WHO Collaborating Center for Standardization and Evaluation of Biologicals, Ottawa, ON, Canada
| | - Lisheng Wang
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael Rosu-Myles
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs 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
| | - Jingxin Cao
- National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, MB, Canada
| | - Xuguang Li
- Centre for Biologics Evaluation, Biologic and Radiopharmaceutical Drugs 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
|
15
|
Tripp RA, Stambas J. Intervention Strategies for Seasonal and Emerging Respiratory Viruses with Drugs and Vaccines Targeting Viral Surface Glycoproteins. Viruses 2021; 13:v13040625. [PMID: 33917411 PMCID: PMC8067509 DOI: 10.3390/v13040625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/31/2021] [Accepted: 04/03/2021] [Indexed: 12/12/2022] Open
Abstract
Vaccines and therapeutics targeting viral surface glycoproteins are a major component of disease prevention for respiratory viral diseases. Over the years, vaccines have proven to be the most successful intervention for preventing disease. Technological advances in vaccine platforms that focus on viral surface glycoproteins have provided solutions for current and emerging pathogens like SARS-CoV-2, and our understanding of the structural basis for antibody neutralization is guiding the selection of other vaccine targets for respiratory viruses like RSV. This review discusses the role of viral surface glycoproteins in disease intervention approaches.
Collapse
Affiliation(s)
- Ralph A. Tripp
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30605, USA
- Correspondence:
| | - John Stambas
- School of Medicine, Geelong Waurn Ponds, Deakin University, Melbourne, VIC 3125, Australia;
| |
Collapse
|